Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 353
Filtrar
1.
J Virol ; 97(4): e0007323, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37022165

RESUMO

Stress-mediated activation of the glucocorticoid receptor (GR) and specific stress-induced transcription factors stimulate herpes simplex virus 1 (HSV-1) productive infection, explant-induced reactivation, and immediate early (IE) promoters that drive expression of infected cell protein 0 (ICP0), ICP4, and ICP27. Several published studies concluded the virion tegument protein VP16, ICP0, and/or ICP4 drives early steps of reactivation from latency. Notably, VP16 protein expression was induced in trigeminal ganglionic neurons of Swiss Webster or C57BL/6J mice during early stages of stress-induced reactivation. If VP16 mediates reactivation, we hypothesized stress-induced cellular transcription factors would stimulate its expression. To address this hypothesis, we tested whether stress-induced transcription factors transactivate a VP16 cis-regulatory module (CRM) located upstream of the VP16 TATA box (-249 to -30). Initial studies revealed the VP16 CRM cis-activated a minimal promoter more efficiently in mouse neuroblastoma cells (Neuro-2A) than mouse fibroblasts (NIH-3T3). GR and Slug, a stress-induced transcription factor that binds enhancer boxes (E-boxes), were the only stress-induced transcription factors examined that transactivated the VP16 CRM construct. GR- and Slug-mediated transactivation was reduced to basal levels when the E-box, two 1/2 GR response elements (GREs), or NF-κB binding site was mutated. Previous studies revealed GR and Slug cooperatively transactivated the ICP4 CRM, but not ICP0 or ICP27. Silencing of Slug expression in Neuro-2A cells significantly reduced viral replication, indicating Slug-mediated transactivation of ICP4 and VP16 CRM activity correlates with enhanced viral replication and reactivation from latency. IMPORTANCE Herpes simplex virus 1 (HSV-1) establishes lifelong latency in several types of neurons. Periodically cellular stressors trigger reactivation from latency. Viral regulatory proteins are not abundantly expressed during latency, indicating cellular transcription factors mediate early stages of reactivation. Notably, the glucocorticoid receptor (GR) and certain stress-induced transcription factors transactivate cis-regulatory modules (CRMs) essential for expression of infected cell protein 0 (ICP0) and ICP4, key viral transcriptional regulatory proteins linked to triggering reactivation from latency. Virion protein 16 (VP16) specifically transactivates IE promoter and was also reported to mediate early stages of reactivation from latency. GR and Slug, a stress-induced enhancer box (E-box) binding protein, transactivate a minimal promoter downstream of VP16 CRM, and these transcription factors occupy VP16 CRM sequences in transfected cells. Notably, Slug stimulates viral replication in mouse neuroblastoma cells suggesting Slug, by virtue of transactivating VP16 and ICP4 CRM sequences, can trigger reactivation in certain neurons.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples , Herpesvirus Humano 1 , Regiões Promotoras Genéticas , Replicação Viral , Animais , Camundongos , Regulação Viral da Expressão Gênica , Infecções por Herpesviridae/virologia , Herpesvirus Humano 1/fisiologia , Camundongos Endogâmicos C57BL , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Replicação Viral/genética , Feminino , Proteína Vmw65 do Vírus do Herpes Simples/genética , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Células NIH 3T3 , Latência Viral/genética , Mutação , RNA Interferente Pequeno/metabolismo
2.
Rev Med Virol ; 32(6): e2394, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36069169

RESUMO

Herpes simplex virus type 1 (HSV-1) is an important human pathogen with neurotropism. Following lytic infection in mucosal or skin epithelium, life-long latency is established mainly in sensory neurons, which can periodically reactivate by stress, leading to recurrent disease and virus transmission. During the virus's productive infection, the tegument protein VP16, a component of HSV-1 virion, is physically associated with two cellular factors, host cell factor-1 (HCF-1), and POU domain protein Oct-1, to construct the VP16-induced complex, which is essential to stimulate immediate early (IE)-gene transcription as well as initiate the lytic programme. Apart from HCF-1 and Oct-1, VP16 also associates with a series of other host factors, making a VP16-induced regulatory switch to either activate or inactivate virus gene transcription. In addition, VP16 has effects on distinct signalling pathways via binding to various host molecules that are essentially related to innate immune responses, RNA polymerases, molecular chaperones, and virus infection-induced host shutoff. VP16 also functionally compensates for given host factors, such as PPAR-γ and ß-catenin. In this review, we provide an overview of the updated insights on the interplay between VP16 and the host factors that coordinate virus infection.


Assuntos
Herpesvirus Humano 1 , Fatores de Transcrição , Humanos , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Herpesvirus Humano 1/metabolismo , Proteína Vmw65 do Vírus do Herpes Simples/química , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Fator C1 de Célula Hospedeira , Etoposídeo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo
3.
Autophagy ; 18(8): 1801-1821, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-34822318

RESUMO

Alphaherpesvirus infection results in severe health consequences in a wide range of hosts. USPs are the largest subfamily of deubiquitinating enzymes that play critical roles in immunity and other cellular functions. To investigate the role of USPs in alphaherpesvirus replication, we assessed 13 USP inhibitors for PRV replication. Our data showed that all the tested compounds inhibited PRV replication, with the USP14 inhibitor b-AP15 exhibiting the most dramatic effect. Ablation of USP14 also influenced PRV replication, whereas replenishment of USP14 in USP14 null cells restored viral replication. Although inhibition of USP14 induced the K63-linked ubiquitination of PRV VP16 protein, its degradation was not dependent on the proteasome. USP14 directly bound to ubiquitin chains on VP16 through its UBL domain during the early stage of viral infection. Moreover, USP14 inactivation stimulated EIF2AK3/PERK- and ERN1/IRE1-mediated signaling pathways, which were responsible for VP16 degradation through SQSTM1/p62-mediated selective macroautophagy/autophagy. Ectopic expression of non-ubiquitinated VP16 fully rescued PRV replication. Challenge of mice with b-AP15 activated ER stress and autophagy and inhibited PRV infection in vivo. Our results suggested that USP14 was a potential therapeutic target to treat alphaherpesvirus-induced infectious diseases.Abbreviations ATF4: activating transcription factor 4; ATF6: activating transcription factor 6; ATG5: autophagy related 5; ATG12: autophagy related 12; CCK-8: cell counting kit-8; Co-IP: co-immunoprecipitation; CRISPR: clustered regulatory interspaced short palindromic repeat; Cas9: CRISPR associated system 9; DDIT3/CHOP: DNA-damage inducible transcript 3; DNAJB9/ERdj4: DnaJ heat shock protein family (Hsp40) member B9; DUBs: deubiquitinases; EIF2A/eIF2α: eukaryotic translation initiation factor 2A; EIF2AK3/PERK: eukaryotic translation initiation factor 2 alpha kinase 3; EP0: ubiquitin E3 ligase ICP0; ER: endoplasmic reticulum; ERN1/IRE1: endoplasmic reticulum (ER) to nucleus signaling 1; FOXO1: forkhead box O1; FRET: Förster resonance energy transfer; HSPA5/BiP: heat shock protein 5; HSV: herpes simplex virus; IE180: transcriptional regulator ICP4; MAP1LC3/LC3: microtube-associated protein 1 light chain 3; MOI: multiplicity of infection; MTOR: mechanistic target of rapamycin kinase; PPP1R15A/GADD34: protein phosphatase 1, regulatory subunit 15A; PRV: pseudorabies virus; PRV gB: PRV glycoprotein B; PRV gE: PRV glycoprotein E; qRT-PCR: quantitative real-time polymerase chain reaction; sgRNA: single guide RNA; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; TCID50: tissue culture infective dose; UB: ubiquitin; UBA: ubiquitin-associated domain; UBL: ubiquitin-like domain; UL9: DNA replication origin-binding helicase; UPR: unfolded protein response; USPs: ubiquitin-specific proteases; VHS: virion host shutoff; VP16: viral protein 16; XBP1: X-box binding protein 1; XBP1s: small XBP1; XBP1(t): XBP1-total.


Assuntos
Alphaherpesvirinae , Autofagia , Estresse do Retículo Endoplasmático , Proteína Vmw65 do Vírus do Herpes Simples , Ubiquitina Tiolesterase , Alphaherpesvirinae/patogenicidade , Alphaherpesvirinae/fisiologia , Animais , Proliferação de Células , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Macroautofagia , Camundongos , Proteína Sequestossoma-1 , Ubiquitina Tiolesterase/metabolismo
4.
Nat Commun ; 12(1): 6160, 2021 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-34697317

RESUMO

Pparg, a nuclear receptor, is downregulated in basal subtype bladder cancers that tend to be muscle invasive and amplified in luminal subtype bladder cancers that tend to be non-muscle invasive. Bladder cancers derive from the urothelium, one of the most quiescent epithelia in the body, which is composed of basal, intermediate, and superficial cells. We find that expression of an activated form of Pparg (VP16;Pparg) in basal progenitors induces formation of superficial cells in situ, that exit the cell cycle, and do not form tumors. Expression in basal progenitors that have been activated by mild injury however, results in luminal tumor formation. We find that these tumors are immune deserted, which may be linked to down-regulation of Nf-kb, a Pparg target. Interestingly, some luminal tumors begin to shift to basal subtype tumors with time, down-regulating Pparg and other luminal markers. Our findings have important implications for treatment and diagnosis of bladder cancer.


Assuntos
PPAR gama/metabolismo , Transdução de Sinais , Neoplasias da Bexiga Urinária/imunologia , Neoplasias da Bexiga Urinária/patologia , Animais , Biomarcadores Tumorais/metabolismo , Carcinogênese , Carcinógenos/toxicidade , Diferenciação Celular , Proliferação de Células , Proteína Vmw65 do Vírus do Herpes Simples/genética , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , PPAR gama/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Neoplasias da Bexiga Urinária/induzido quimicamente , Neoplasias da Bexiga Urinária/metabolismo , Urotélio/efeitos dos fármacos , Urotélio/imunologia , Urotélio/patologia
5.
J Immunol Res ; 2020: 9630452, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32537474

RESUMO

Duck enteritis virus (DEV) can successfully evade the host innate immune responses and establish a lifelong latent infection in the infected host. However, the study about how DEV escapes host innate immunity is still deficient up to now. In this study, for the first time, we identified a viral protein VP16 by which DEV can obviously downregulate the production of IFN-ß in duck embryo fibroblast (DEF). Our results showed that ectopic expression of VP16 decreased duck IFN-ß (duIFN-ß) promoter activation and significantly inhibited the mRNA transcription of IFN-ß. Further study showed that VP16 can also obviously inhibit the mRNA transcription of interferon-stimulated genes (ISGs), such as myxovirus resistance protein (Mx) and interferon-induced oligoadenylate synthetase-like (OASL). Furthermore, we found that this anti-interferon activity of VP16 depended on its N-terminus (aa1-200). Coexpression analysis revealed that VP16 selectively blocked duIFN-ß promoter activity at the duIRF7 level rather than duIRF1. Based on the results of coimmunoprecipitation analysis (co-IP) and indirect immunofluorescence assay (IFA), VP16 was able to bind to duck IRF7 (duIRF7) directly, but did not interact with duck IRF1 (duIRF1) in vitro.


Assuntos
Patos/imunologia , Fibroblastos/fisiologia , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Infecções por Herpesviridae/imunologia , Herpesvirus Galináceo 1/fisiologia , Interferon beta/metabolismo , Mardivirus/fisiologia , 2',5'-Oligoadenilato Sintetase/genética , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Células Cultivadas , Patos/virologia , Fibroblastos/virologia , Regulação da Expressão Gênica , Infecções por Herpesviridae/virologia , Evasão da Resposta Imune , Imunidade Inata , Fator Regulador 1 de Interferon/genética , Fator Regulador 7 de Interferon/genética , Interferon beta/genética , Proteínas de Resistência a Myxovirus/genética , Regiões Promotoras Genéticas/genética
6.
Int J Biol Sci ; 16(9): 1586-1603, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32226304

RESUMO

Herpes simplex virus (HSV) type 1 (HSV-1) infection exhibited high heterogeneity at individual cells level, including the different gene expression patterns and varying amounts of progeny virus. However, the underlying mechanism of such variability remains obscure. The importance of host long noncoding RNAs (lncRNAs) in virus infection had been recognized, while the contribution of lncRNAs to the heterogeneous infection remains unknown. Herein, a prior single-cell RNA sequencing data using HSV-1 reporter strain expressing ICP4-YFP was re-analyzed to obtain the differentially expressed lncRNA between the successfully initiated viral gene expression (ICP4-YFP+) cells and the aborted infection cells (ICP4-YFP-). The ICP4-YFP+ population show a higher abundance of MAMDC2 antisense 1 (MAMDC2-AS1) lncRNA than ICP4-YFP- population. MAMDC2-AS1 silencing reduces the expression of HSV-1 immediate early (IE) genes and limit HSV-1 infection in human host cells. Consistently, ectopic expression of MAMDC2-AS1 enhances HSV-1 IE genes transcription and facilitates the formation of HSV-1-induced plaques. Mechanically, both RNA-pull down and RNA immunoprecipitation assays show that MAMDC2-AS1 interacts with the RNA binding protein heat shock protein 90α (Hsp90α), a molecular chaperone involving in the nuclear import of HSV-1. The MAMDC2-AS1-Hsp90α interaction facilitates the nuclear transport of viral tegument protein VP16, the core factor initiating the expression of HSV-1 IE genes. The transcription factor YY1 mediates the induction of MAMDC2-AS1 upon HSV-1 infection. Our study elucidates the contribution of lncRNA to HSV-1 infection susceptibility in human cells and the role of Hsp90α RNA binding activity in HSV-1 infection.


Assuntos
Núcleo Celular/virologia , Herpesvirus Humano 1/metabolismo , RNA Longo não Codificante/fisiologia , Transporte Ativo do Núcleo Celular , Linhagem Celular , Genes Precoces , Proteínas de Choque Térmico HSP90/metabolismo , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/genética , Humanos , RNA Longo não Codificante/biossíntese , RNA Longo não Codificante/metabolismo , Proteínas de Ligação a RNA/metabolismo , RNA-Seq , Análise de Célula Única , Fator de Transcrição YY1/fisiologia
7.
J Virol ; 93(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30971470

RESUMO

Herpes simplex virus 1 (HSV-1) establishes lifelong latent infections in neurons. Reactivation from latency can lead to serious recurrent disease, including stromal keratitis, corneal scarring, blindness, and encephalitis. Although numerous studies link stress to an increase in the incidence of reactivation from latency and recurrent disease, the mechanism of action is not well understood. We hypothesized that stress, via corticosteroid-mediated activation of the glucocorticoid receptor (GR), stimulates viral gene expression and productive infection during reactivation from latency. Consequently, we tested whether GR activation by the synthetic corticosteroid dexamethasone influenced virus shedding during reactivation from latency using trigeminal ganglion (TG) explants from Swiss Webster mice latently infected with HSV-1, strain McKrae. TG explants from the latently infected mice shed significantly higher levels of virus when treated with dexamethasone. Conversely, virus shedding from TG explants was significantly impaired when they were incubated with medium containing a GR-specific antagonist (CORT-108297) or stripped fetal bovine serum, which lacks nuclear hormones and other growth factors. TG explants from latently infected, but not uninfected, TG contained significantly more GR-positive neurons following explant when treated with dexamethasone. Strikingly, VP16 protein expression was detected in TG neurons at 8 hours after explant whereas infected-cell protein 0 (ICP0) and ICP4 protein expression was not readily detected until 16 hours after explant. Expression of all three viral regulatory proteins was stimulated by dexamethasone. These studies indicated corticosteroid-mediated GR activation increased the number of TG neurons expressing viral regulatory proteins, which enhanced virus shedding during explant-induced reactivation from latency.IMPORTANCE Herpes simplex virus 1 (HSV-1) establishes lifelong latent infections in neurons within trigeminal ganglia (TG); periodically, reactivation from latency occurs, leading to virus transmission and recurrent disease. Chronic or acute stress increases the frequency of reactivation from latency; how this occurs is not well understood. Here, we demonstrate that the synthetic corticosteroid dexamethasone stimulated explant-induced reactivation from latency. Conversely, a glucocorticoid receptor (GR) antagonist significantly impaired reactivation from latency, indicating that GR activation stimulated explant-induced reactivation. The viral regulatory protein VP16 was readily detected in TG neurons prior to infected-cell protein 0 (ICP0) and ICP4 during explant-induced reactivation. Dexamethasone induced expression of all three viral regulatory proteins following TG explant. Whereas the immunosuppressive properties of corticosteroids would facilitate viral spread during reactivation from latency, these studies indicate GR activation increases the number of TG neurons that express viral regulatory proteins during early stages of explant-induced reactivation.


Assuntos
Herpesvirus Humano 1/fisiologia , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/metabolismo , Latência Viral/efeitos dos fármacos , Latência Viral/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Regulação Viral da Expressão Gênica , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/genética , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Neurônios/virologia , Estresse Fisiológico , Gânglio Trigeminal/patologia , Gânglio Trigeminal/virologia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ativação Viral/genética , Ativação Viral/fisiologia , Latência Viral/genética , Eliminação de Partículas Virais
8.
Mol Med ; 24(1): 65, 2018 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-30577726

RESUMO

BACKGROUND: Numerous host cellular factors are exploited by viruses to facilitate infection. Our previous studies and those of others have shown heat-shock protein 90 (Hsp90), a cellular molecular chaperone, is involved in herpes simplex virus (HSV)-1 infection. However, the function of the dominant Hsp90 isoform and the relationship between Hsp90 and HSV-1 α genes remain unclear. METHODS AND RESULTS: Hsp90α knockdown or inhibition significantly inhibited the promoter activity of HSV-1 α genes and downregulated virion protein 16(VP16) expression from virus and plasmids. The Hsp90α knockdown-induced suppression of α genes promoter activity and downregulation of α genes was reversed by VP16 overexpression, indicating that Hsp90α is involved in VP16-mediated transcription of HSV-1 α genes. Co-immunoprecipitation experiments indicated that VP16 interacted with Hsp90α through the conserved core domain within VP16. Based on using autophagy inhibitors and the presence of Hsp90 inhibitors in ATG7-/- (autophagy-deficient) cells, Hsp90 inhibition-induced degradation of VP16 is dependent on macroautophagy-mediated degradation but not chaperone-mediated autophagy (CMA) pathway. In vivo studies demonstrated that treatment with gels containing Hsp90 inhibitor effectively reduced the level of VP16 and α genes, which may contribute to the amelioration of the skin lesions in an HSV-1 infection mediated zosteriform model. CONCLUSION: Our study provides new insights into the mechanisms by which Hsp90α facilitates the transactivation of HSV-1 α genes and viral infection, and highlights the importance of developing selective inhibitors targeting the interaction between Hsp90α and VP16 to reduce toxicity, a major challenge in the clinical use of Hsp90 inhibitors.


Assuntos
Proteínas de Choque Térmico HSP90/genética , Proteína Vmw65 do Vírus do Herpes Simples/genética , Herpesvirus Humano 1/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Feminino , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Herpes Simples/tratamento farmacológico , Herpes Simples/genética , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Ativação Transcricional
9.
Sci Rep ; 8(1): 15876, 2018 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-30367157

RESUMO

Analysis of a genome-scale RNA interference screen of host factors affecting herpes simplex virus type 1 (HSV-1) revealed that the mineralocorticoid receptor (MR) inhibits HSV-1 replication. As a ligand-activated transcription factor the MR regulates sodium transport and blood pressure in the kidney in response to aldosterone, but roles have recently been elucidated for the MR in other cellular processes. Here, we show that the MR and other members of the mineralocorticoid signalling pathway including HSP90 and FKBP4, possess anti-viral activity against HSV-1 independent of their effect on sodium transport, as shown by sodium channel inhibitors. Expression of the MR is upregulated upon infection in an interferon (IFN) and viral transcriptional activator VP16-dependent fashion. Furthermore, the MR and VP16, together with the cellular co-activator Oct-1, transactivate the hormone response element (HRE) present in the MR promoter and those of its transcriptional targets. As the MR induces IFN expression, our data suggests the MR is involved in a positive feedback loop that controls HSV-1 infection.


Assuntos
Antivirais/farmacologia , Herpesvirus Humano 1/fisiologia , Receptores de Mineralocorticoides/metabolismo , Replicação Viral/efeitos dos fármacos , Antivirais/uso terapêutico , Células HeLa , Herpes Simples/tratamento farmacológico , Herpes Simples/patologia , Proteína Vmw65 do Vírus do Herpes Simples/genética , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/isolamento & purificação , Humanos , Interferons/farmacologia , Interferons/uso terapêutico , Fator 1 de Transcrição de Octâmero/genética , Fator 1 de Transcrição de Octâmero/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Mineralocorticoides/química , Receptores de Mineralocorticoides/genética , Ativação Transcricional/efeitos dos fármacos
10.
Virus Res ; 256: 29-37, 2018 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-30077727

RESUMO

The ability of herpes simplex virus 1 (HSV-1) to replicate efficiently in differentiated cells is regulated by cellular factors that stimulate viral gene expression, cell survival, and viral morphogenesis. Activation of the canonical Wnt signaling pathway generally increases ß-catenin protein levels, cell survival, and growth in dividing cells suggesting this important signaling pathway regulates productive infection. In this study, we demonstrated that a ß-catenin specific small molecule inhibitor (iCRT14) reduced HSV-1 titers approximately 10-fold in primary human lung fibroblasts and Vero cells. Furthermore, ß-catenin dependent transcription was increased at late times after infection and as expected iCRT14 reduced ß-catenin dependent transcription. Although HSV-1 infection increased ß-catenin steady state protein levels approximately 4-fold in Vero cells, there was only a nominal increase in human lung fibroblasts. We hypothesized that VP16 regulates ß-catenin dependent transcription because VP16 is a viral regulatory protein expressed at late times after infection. In the absence of other viral proteins, VP16 increased ß-catenin dependent transcription and ß-catenin steady state protein levels. Collectively, these studies suggested the cellular transcription factor ß-catenin stimulates productive infection, in part because VP16 enhances ß-catenin dependent transcription.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Replicação Viral , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Células Cultivadas , Células Epiteliais/virologia , Fibroblastos/virologia , Humanos , Carga Viral
11.
J Virol ; 92(17)2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29925667

RESUMO

The herpes simplex virus 1 (HSV-1) virion host shutoff (vhs) protein is an endoribonuclease that binds to the cellular translation initiation machinery and degrades associated mRNAs, resulting in the shutoff of host protein synthesis. Hence, its unrestrained activity is considered lethal, and it has been proposed that vhs is regulated by two other virus proteins, VP22 and VP16. We have found that during infection, translation of vhs requires VP22 but not the VP22-VP16 complex. Moreover, in the absence of VP22, vhs is not overactive against cellular or viral transcripts. In transfected cells, vhs was also poorly translated, correlating with the aberrant localization of its mRNA. Counterintuitively, vhs mRNA was predominantly nuclear in cells where vhs protein was detected. Likewise, transcripts from cotransfected plasmids were also retained in the same nuclei where vhs mRNA was located, while poly(A) binding protein (PABP) was relocalized to the nucleus in a vhs-dependent manner, implying a general block to mRNA export. Coexpression of VP16 and VP22 rescued the cytoplasmic localization of vhs mRNA but failed to rescue vhs translation. We identified a 230-nucleotide sequence in the 5' region of vhs that blocked its translation and, when transferred to a heterologous green fluorescent protein transcript, reduced translation without altering mRNA levels or localization. We propose that expression of vhs is tightly regulated by a combination of inherent untranslatability and autoinduced nuclear retention of its mRNA that results in a negative feedback loop, with nuclear retention but not translation of vhs mRNA being the target of rescue by the vhs-VP16-VP22 complex.IMPORTANCE A myriad of gene expression strategies has been discovered through studies carried out on viruses. This report concerns the regulation of the HSV-1 vhs endoribonuclease, a virus factor that is important for counteracting host antiviral responses by degrading their mRNAs but that must be regulated during infection to ensure that it does not act against and inhibit the virus itself. We show that regulation of vhs involves multifaceted posttranscriptional cellular and viral processes, including aberrant mRNA localization and a novel, autoregulated negative feedback loop to target its own and coexpressed mRNAs for nuclear retention, an activity that is relieved by coexpression of two other virus proteins, VP22 and VP16. These studies reveal the interplay of strategies by which multiple virus-encoded factors coordinate gene expression at the time that they are needed. These findings are broadly relevant to both virus and cellular gene expression.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/enzimologia , Ribonucleases/genética , Proteínas Virais/genética , Proteínas Estruturais Virais/metabolismo , Regiões 5' não Traduzidas , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Viral da Expressão Gênica , Herpesvirus Humano 1/genética , Fases de Leitura Aberta , Processamento Pós-Transcricional do RNA , Ribonucleases/química , Ribonucleases/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo
12.
Cell Rep ; 20(7): 1585-1596, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28813671

RESUMO

SOX2 and OCT4, in conjunction with KLF4 and cMYC, are sufficient to reprogram human fibroblasts to induced pluripotent stem cells (iPSCs), but it is unclear if they function as transcriptional activators or as repressors. We now show that, like OCT4, SOX2 functions as a transcriptional activator. We substituted SOX2-VP16 (a strong activator) for wild-type (WT) SOX2, and we saw an increase in the efficiency and rate of reprogramming, whereas the SOX2-HP1 fusion (a strong repressor) eliminated reprogramming. We report that, at an early stage of reprogramming, virtually all DNA-bound OCT4, SOX2, and SOX2-VP16 were embedded in putative enhancers, about half of which were created de novo. Those associated with SOX2-VP16 were, on average, stronger than those bearing WT SOX2. Many newly created putative enhancers were transient, and many transcription factor locations on DNA changed as reprogramming progressed. These results are consistent with the idea that, during reprogramming, there is an intermediate state that is distinct from both parental cells and iPSCs.


Assuntos
Reprogramação Celular , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Proteínas Recombinantes de Fusão/genética , Fatores de Transcrição SOXB1/genética , Diferenciação Celular , Fibroblastos/citologia , Proteína Vmw65 do Vírus do Herpes Simples/genética , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Ativação Transcricional
13.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28275191

RESUMO

Several virulence genes have been identified thus far in the herpes simplex virus 1 genome. It is also generally accepted that protein heterogeneity among virions further impacts viral fitness. However, linking this variability directly with infectivity has been challenging at the individual viral particle level. To address this issue, we resorted to flow cytometry (flow virometry), a powerful approach we recently employed to analyze individual viral particles, to identify which tegument proteins vary and directly address if such variability is biologically relevant. We found that the stoichiometry of the UL37, ICP0, and VP11/12 tegument proteins in virions is more stable than the VP16 and VP22 tegument proteins, which varied significantly among viral particles. Most interestingly, viruses sorted for their high VP16 or VP22 content yielded modest but reproducible increases in infectivity compared to their corresponding counterparts containing low VP16 or VP22 content. These findings were corroborated for VP16 in short interfering RNA experiments but proved intriguingly more complex for VP22. An analysis by quantitative Western blotting revealed substantial alterations of virion composition upon manipulation of individual tegument proteins and suggests that VP22 protein levels acted indirectly on viral fitness. These findings reaffirm the interdependence of the virion components and corroborate that viral fitness is influenced not only by the genome of viruses but also by the stoichiometry of proteins within each virion.IMPORTANCE The ability of viruses to spread in animals has been mapped to several viral genes, but other factors are clearly involved, including virion heterogeneity. To directly probe whether the latter influences viral fitness, we analyzed the protein content of individual herpes simplex virus 1 particles using an innovative flow cytometry approach. The data confirm that some viral proteins are incorporated in more controlled amounts, while others vary substantially. Interestingly, this correlates with the VP16 trans-activating viral protein and indirectly with VP22, a second virion component whose modulation profoundly alters virion composition. This reaffirms that not only the presence but also the amount of specific tegument proteins is an important determinant of viral fitness.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Proteínas Estruturais Virais/metabolismo , Animais , Western Blotting , Chlorocebus aethiops , Citometria de Fluxo , Genes Virais , Proteína Vmw65 do Vírus do Herpes Simples/análise , Proteína Vmw65 do Vírus do Herpes Simples/química , Herpesvirus Humano 1/patogenicidade , RNA Interferente Pequeno , Células Vero , Proteínas Estruturais Virais/análise , Proteínas Estruturais Virais/química , Vírion/genética , Vírion/fisiologia , Montagem de Vírus
14.
Virol J ; 14(1): 35, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28222744

RESUMO

BACKGROUND: Herpes simplex virus 1 (HSV-1) is an archetypal member of the alphaherpesvirus subfamily with a large genome encoding over 80 proteins, many of which play a critical role in virus-host interactions and immune modulation. Upon viral infections, the host cells activate innate immune responses to restrict their replications. Peroxisomes, which have long been defined to regulate metabolic activities, are reported to be important signaling platforms for antiviral innate immunity. It has been verified that signaling from peroxisomal MAVS (MAVS-Pex) triggers a rapid interferon (IFN) independent IFN-stimulated genes (ISGs) production against invading pathogens. However, little is known about the interaction between DNA viruses such as HSV-1 and the MAVS-Pex mediated signaling. RESULTS: HSV-1 could activate the MAVS-Pex signaling pathway at a low multiplicity of infection (MOI), while infection at a high MOI dampens MAVS-Pex induced immediately early ISGs production. A high-throughput screen assay reveals that HSV-1 tegument protein VP16 inhibits the immediate early ISGs expression downstream of MAVS-Pex signaling. Moreover, the expression of ISGs was recovered when VP16 was knockdown with its specific short hairpin RNA. CONCLUSION: HSV-1 blocks MAVS-Pex mediated early ISGs production through VP16 to dampen the immediate early antiviral innate immunity signaling from peroxisomes.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/patogenicidade , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Imunidade Inata , Peroxissomos/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular , Humanos , Endopeptidase Neutra Reguladora de Fosfato PHEX/metabolismo
15.
Protein Sci ; 25(8): 1371-7, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27213278

RESUMO

Transcriptional activators coordinate the dynamic assembly of multiprotein coactivator complexes required for gene expression to occur. Here we combine the power of in vivo covalent chemical capture with p-benzoyl-L-phenylalanine (Bpa), a genetically incorporated photo-crosslinking amino acid, and chromatin immunoprecipitation (ChIP) to capture the direct protein interactions of the transcriptional activator VP16 with the general transcription factor TBP at the GAL1 promoter in live yeast.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples/genética , Saccharomyces cerevisiae/genética , Proteína de Ligação a TATA-Box/genética , Transativadores/genética , Ativação Transcricional , Benzofenonas/química , Benzofenonas/metabolismo , Imunoprecipitação da Cromatina , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/metabolismo , Galactoquinase/genética , Galactoquinase/metabolismo , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Fenilalanina/análogos & derivados , Fenilalanina/química , Fenilalanina/metabolismo , Processos Fotoquímicos , Regiões Promotoras Genéticas , Ligação Proteica , Multimerização Proteica , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Proteína de Ligação a TATA-Box/metabolismo , Transativadores/metabolismo
16.
Exp Neurol ; 280: 115-20, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27060489

RESUMO

Axonal regeneration after spinal cord injury (SCI) is intrinsically and extrinsically inhibited by multiple factors. One major factor contributing to intrinsic regeneration failure is the inability of mature neurons in the central nervous system (CNS) to activate regeneration-associated transcription factors (TFs) post-injury. A prior study identified TFs overexpressed in neurons of the peripheral nervous system (PNS) compared to the CNS; some of these could be involved in the ability of PNS neurons to regenerate. Of these, signal transducer and activator of transcription 3 (STAT3), as well its downstream regeneration-associated targets, showed a significant upregulation in PNS neurons relative to CNS neurons, and a constitutively active variant of Stat3 (Stat3CA) promoted neurite growth when expressed in cerebellar neurons (Lerch et al., 2012; Smith et al., 2011). To further enhance STAT3's neurite outgrowth enhancing activity, Stat3CA was fused with a viral activation domain (VP16). VP16 hyperactivates TFs by recruiting transcriptional co-factors to the DNA binding domain (Hirai et al., 2010). Overexpression of this VP16-Stat3CA chimera in primary cortical neurons led to a significant increase of neurite outgrowth as well as Stat3 transcriptional activity in vitro. Furthermore, in vivo transduction of retinal ganglion cells (RGCs) with AAV constructs expressing VP16-Stat3CA resulted in regeneration of optic nerve axons after injury, to a greater degree than for those expressing Stat3CA alone. These findings confirm and extend the concept that overexpression of hyperactivated transcription factors identified as functioning in PNS regeneration can promote axon regeneration in the CNS.


Assuntos
Sistema Nervoso Central/patologia , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Regeneração Nervosa/fisiologia , Traumatismos do Nervo Óptico/patologia , Fator de Transcrição STAT3/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Axônios/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Toxina da Cólera/toxicidade , Feminino , Proteína Vmw65 do Vírus do Herpes Simples/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Neuritos , Ratos , Fator de Transcrição STAT3/genética , Transdução Genética , Regulação para Cima/genética
17.
J Virol ; 90(11): 5368-5383, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27009950

RESUMO

UNLABELLED: Herpes simplex virus (HSV) replicates in the skin and mucous membranes, and initiates lytic or latent infections in sensory neurons. Assembly of progeny virions depends on the essential large tegument protein pUL36 of 3,164 amino acid residues that links the capsids to the tegument proteins pUL37 and VP16. Of the 32 tryptophans of HSV-1-pUL36, the tryptophan-acidic motifs (1766)WD(1767) and (1862)WE(1863) are conserved in all HSV-1 and HSV-2 isolates. Here, we characterized the role of these motifs in the HSV life cycle since the rare tryptophans often have unique roles in protein function due to their large hydrophobic surface. The infectivity of the mutants HSV-1(17(+))Lox-pUL36-WD/AA-WE/AA and HSV-1(17(+))Lox-CheVP26-pUL36-WD/AA-WE/AA, in which the capsid has been tagged with the fluorescent protein Cherry, was significantly reduced. Quantitative electron microscopy shows that there were a larger number of cytosolic capsids and fewer enveloped virions compared to their respective parental strains, indicating a severe impairment in secondary capsid envelopment. The capsids of the mutant viruses accumulated in the perinuclear region around the microtubule-organizing center and were not dispersed to the cell periphery but still acquired the inner tegument proteins pUL36 and pUL37. Furthermore, cytoplasmic capsids colocalized with tegument protein VP16 and, to some extent, with tegument protein VP22 but not with the envelope glycoprotein gD. These results indicate that the unique conserved tryptophan-acidic motifs in the central region of pUL36 are required for efficient targeting of progeny capsids to the membranes of secondary capsid envelopment and for efficient virion assembly. IMPORTANCE: Herpesvirus infections give rise to severe animal and human diseases, especially in young, immunocompromised, and elderly individuals. The structural hallmark of herpesvirus virions is the tegument, which contains evolutionarily conserved proteins that are essential for several stages of the herpesvirus life cycle. Here we characterized two conserved tryptophan-acidic motifs in the central region of the large tegument protein pUL36 of herpes simplex virus. When we mutated these motifs, secondary envelopment of cytosolic capsids and the production of infectious particles were severely impaired. Our data suggest that pUL36 and its homologs in other herpesviruses, and in particular such tryptophan-acidic motifs, could provide attractive targets for the development of novel drugs to prevent herpesvirus assembly and spread.


Assuntos
Capsídeo/metabolismo , Herpesvirus Humano 1/fisiologia , Triptofano/química , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/metabolismo , Montagem de Vírus , Motivos de Aminoácidos , Capsídeo/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Citoplasma/virologia , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Humanos , Estágios do Ciclo de Vida , Microscopia Eletrônica , Mutação , Ligação Proteica , Domínios Proteicos , Triptofano/metabolismo , Proteínas Estruturais Virais/genética
18.
J Neuroimmunol ; 291: 110-4, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26857504

RESUMO

Environmental factors are implicated in the development of Parkinson's disease (PD). We have investigated on the role of molecular mimicry between HSV1 and α-synuclein that could foster the progression of PD. The antibody response against homologous peptides in PD patients and healthy controls was evaluated, showing that these antibodies are highly prevalent among PD patients to healthy controls. The competitive assay demonstrated cross-reactivity between HSV1 and human α-synuclein peptides. The results may suggest the hypothesis of the involvement of HSV1 in stimulating the immune cells against the neurons of the substantia nigra as a consequence of the cross reactivity.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Doença de Parkinson/complicações , alfa-Sinucleína/metabolismo , Idoso , Estudos de Casos e Controles , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mimetismo Molecular , Peptídeos/metabolismo , Curva ROC , Substância Negra/metabolismo , Substância Negra/patologia
19.
Bing Du Xue Bao ; 32(6): 817-24, 2016 11.
Artigo em Chinês | MEDLINE | ID: mdl-30004657

RESUMO

Viral protein(VP)16is an important tegument protein of the herpes virus. It is involved in early transcription activation of viral genes as well as virion assembly and release in host cells.VP16 of some herpes viruses have deubiquitinating protease activity and can help the virus counteract the host immune response. In this review, we explain the function and complex interactions between VP16 and other proteins based of the structural characteristics of VP16.This summary provides a reference for further research of maturation of the herpes virus as well as interactions between VP16 and other proteins.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/metabolismo , Animais , Proteína Vmw65 do Vírus do Herpes Simples/genética , Herpesvirus Humano 1/genética , Humanos , Ligação Proteica
20.
Intervirology ; 59(4): 187-196, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28178699

RESUMO

OBJECT: Duck enteritis virus (DEV) is a member of the Alphaherpesvirinae viruses. VP16 and pUL14 are both predicted to be tegument proteins of DEV. METHODS: An indirect immunofluorescence assay (IFA) was performed for preliminary analysis of the colocalization of pUL14 and VP16, which detected their subcellular localization in duck embryo fibroblasts (DEFs) during virus replication. The coexpression of pUL14 and VP16 was detected in transfected DEFs. A bimolecular fluorescence complementation (BiFC) assay was used to confirm a direct interaction between pUL14 and VP16. To better characterize the nuclear localization domain of pUL14, we designed a series of deletion mutants and transfected them with VP16. RESULTS: Our IFA findings indicated that pUL14 binds to VP16 in the cytoplasm and that pUL14 leads VP16 import into the nucleus during DEV replication. The BiFC assay revealed the presence of pUL14 and VP16 complexes. Furthermore, 1-98 amino acid (aa) at the N-terminus of pUL14 played a role in the nuclear localization signal (NLS) region and promoted translocation of VP16 into the nucleus to complete the virus life cycle. CONCLUSIONS: Our findings indicated that pUL14 could transport VP16 into the nucleus and that the N-terminal 1-98 aa may contain the NLS domain of pUL14.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples/genética , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Mardivirus/genética , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Proteínas Virais/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Núcleo Celular/genética , Patos/virologia , Fibroblastos/ultraestrutura , Fibroblastos/virologia , Microscopia de Fluorescência , Mutação , Transfecção , Proteínas Virais/metabolismo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...