Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
J Virol ; 94(13)2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32295924

RESUMO

hnRNPA2B1, an abundant cellular protein, has been reported to recruit RNAs bearing a specific sequence (EXO motif) into exosomes. We characterized an exosome population averaging 100 ± 50 nm in diameter and containing a defined set of constitutive exosome markers. This population packages microRNAs (miRNAs) and can be directed to block targeted gene expression in a dose-dependent fashion. The objective of this study was to characterize the role of hnRNPA2B1 in the recruitment of miRNA. We report the following four key findings. (i) hnRNPA2B1 is not a component of exosomes produced in HEp-2 or HEK293T cells. Hence, hnRNPA2B1 carries its cargo, at most, to the site of exosome assembly, but it is not itself incorporated into exosomes. (ii) The accumulation of exosomes produced by cells in which the gene encoding hnRNPA2B1 has been knocked out (ΔhnRNPA2B1 cells) was reduced 3-fold. (iii) In uninfected HEp-2 cells, hnRNPA2B1 is localized in the nucleus. In cells infected with herpes simplex virus 1 (HSV-1), hnRNPA2B1 was quantitatively exported to the cytoplasm and at least a fraction of hnRNPA2B1 colocalized with a Golgi marker. (iv) Lastly, in ΔhnRNPA2B1 cells, there was a 2- to 3-fold reduction in virus yield but a significant (>10-fold) reduction in HSV-1 released through the apical surface into the extracellular environment. The absence of hnRNPA2B1 had no significant impact on the basolateral export of HSV-1 from infected to uninfected cells by direct cell-to-cell contact. The results suggest that hnRNPA2B1 plays a key role in the transport of enveloped virus from its site of assembly to the extracellular environment.IMPORTANCE In this report, we show that hnRNPA2B1 is not a component of exosomes produced in HEp-2 or HEK293T cells. In herpes simplex virus 1 (HSV-1)-infected cells, hnRNPA2B1 was quantitatively translocated from the nucleus into the cytoplasm. In infected ΔhnRNPA2B1 cells, Golgi-dependent transport of virus from the apical surface to the extracellular medium was significantly reduced. In essence, this report supports the hypothesis that hnRNPA2B1 plays a key role in the egress of exosomes and HSV-1 from infected cells.


Assuntos
Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Herpesvirus Humano 1/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Animais , Chlorocebus aethiops , Exossomos/metabolismo , Células HEK293 , Herpes Simples/virologia , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/patogenicidade , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Humanos , MicroRNAs/genética , RNA Mensageiro/genética , Células Vero , Proteínas Virais/metabolismo , Replicação Viral
2.
Proc Natl Acad Sci U S A ; 115(14): E3230-E3237, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29559532

RESUMO

Analyses of the levels of mRNAs encoding IFIT1, IFI16, RIG-1, MDA5, CXCL10, LGP2, PUM1, LSD1, STING, and IFNß in cell lines from which the gene encoding LGP2, LSD1, PML, HDAC4, IFI16, PUM1, STING, MDA5, IRF3, or HDAC 1 had been knocked out, as well as the ability of these cell lines to support the replication of HSV-1, revealed the following: (i) Cell lines lacking the gene encoding LGP2, PML, or HDAC4 (cluster 1) exhibited increased levels of expression of partially overlapping gene networks. Concurrently, these cell lines produced from 5 fold to 12 fold lower yields of HSV-1 than the parental cells. (ii) Cell lines lacking the genes encoding STING, LSD1, MDA5, IRF3, or HDAC 1 (cluster 2) exhibited decreased levels of mRNAs of partially overlapping gene networks. Concurrently, these cell lines produced virus yields that did not differ from those produced by the parental cell line. The genes up-regulated in cell lines forming cluster 1, overlapped in part with genes down-regulated in cluster 2. The key conclusions are that gene knockouts and subsequent selection for growth causes changes in expression of multiple genes, and hence the phenotype of the cell lines cannot be ascribed to a single gene; the patterns of gene expression may be shared by multiple knockouts; and the enhanced immunity to viral replication by cluster 1 knockout cell lines but not by cluster 2 cell lines suggests that in parental cells, the expression of innate resistance to infection is specifically repressed.


Assuntos
Biomarcadores/análise , Redes Reguladoras de Genes , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Imunidade Inata/genética , Neoplasias Laríngeas/imunologia , Replicação Viral/genética , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Herpes Simples/genética , Herpes Simples/virologia , Humanos , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/virologia , Células Tumorais Cultivadas
3.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31413129

RESUMO

An earlier report showed that herpes simplex virus 1 (HSV-1) expresses two microRNAs (miRNAs), miR-H28 and miR-H29, late in the infectious cycle. The miRNAs are packed in exosomes and, in recipient cells, restrict the transmission of virus from infected cells to uninfected cells. We now report that (i) miR-H28 induced the synthesis of gamma interferon (IFN-γ) in both infected cells and cells transfected with miR-H28, (ii) IFN-γ accumulated concurrently with viral proteins in infected cells, (iii) IFN-γ was produced in HEp-2 cells derived from cancer tissue and in HEK293T cells derived from normal tissue, and (iv) HSV-1 replication was affected by exposure to IFN-γ before infection but not during or after infection. The results presented in this report support the growing body of evidence indicating that HSV-1 encodes functions designed to reduce the spread of infection from infected cells to uninfected cells, possibly in order to maximize the transmission of virus from infected individuals to uninfected individuals.IMPORTANCE In this report, we show that IFN-γ is produced by HSV-1 viral miR-H28 and viral replication is blocked in cells exposed to IFN-γ before infection but not during or after infection. The inevitable conclusion is that HSV-1 induces IFN-γ to curtail its spread from infected cells to uninfected cells. In essence, this report supports the hypothesis that HSV-1 encodes functions that restrict the transmission of virus from cell to cell.


Assuntos
Antivirais/metabolismo , Herpes Simples/transmissão , Herpesvirus Humano 1/fisiologia , Interferon gama/metabolismo , MicroRNAs/genética , RNA Mensageiro/metabolismo , Replicação Viral/efeitos dos fármacos , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/virologia , Comunicação Celular , Células HEK293 , Herpes Simples/metabolismo , Herpes Simples/virologia , Humanos , Interferon gama/genética , Neoplasias Laríngeas/imunologia , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , Neoplasias Laríngeas/virologia , RNA Mensageiro/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
4.
J Virol ; 93(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30700604

RESUMO

The stress response genes encoding GADD45γ, and to a lesser extent GADD45ß, are activated early in infection with herpes simplex virus 1 (HSV-1). Cells that had been depleted of GADD45γ by transfection of short hairpin RNA (shRNA) or in which the gene had been knocked out (ΔGADD45γ) yielded significantly less virus than untreated infected cells. Consistent with lower virus yields, the ΔGADD45γ cells (either uninfected or infected with HSV-1) exhibited significantly higher levels of transcripts of a cluster of innate immunity genes, including those encoding IFI16, IFIT1, MDA5, and RIG-I. Members of this cluster of genes were reported by this laboratory to be activated concurrently with significantly reduced virus yields in cells depleted of LGP2 or HDAC4. We conclude that innate immunity to HSV-1 is normally repressed in unstressed cells and repression appears to be determined by two mechanisms. The first, illustrated here, is through activation by HSV-1 infection of the gene encoding GADD45γ. The second mechanism requires constitutively active expression of LGP2 and HDAC4.IMPORTANCE Previous studies from our laboratory reported that knockout of some innate immunity genes was associated with increases in the expression of overlapping networks of genes and significant loss of the ability to support the replication of HSV-1; knockout of other genes was associated with decreases in the expression of overlapping networks of genes and had no effect on virus replication. In this report, we document that depletion of GADD45γ reduced virus yields concurrently with significant upregulation of the expression of a cluster of innate immunity genes comprising IFI16, IFIT1, MDA5, and RIG-I. This report differs from the preceding study in an important respect; i.e., the preceding study found no evidence to support the hypothesis that HSV-1 maintained adequate levels of LGP2 or HDAC4 to block upregulation of the cluster of innate immunity genes. We show that HSV-1 causes upregulation of the GADD45γ gene to prevent the upregulation of innate immunity genes.


Assuntos
Proteínas de Ciclo Celular/imunologia , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Imunidade Inata/imunologia , Proteínas Nucleares/imunologia , Animais , Chlorocebus aethiops , Histona Desacetilases/imunologia , Proteínas de Membrana/imunologia , RNA Helicases/imunologia , RNA Interferente Pequeno/imunologia , Células Vero , Replicação Viral/imunologia
5.
Proc Natl Acad Sci U S A ; 114(19): E3823-E3829, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28439026

RESUMO

Nuclear domain 10 (ND10) bodies are small (0.1-1 µM) nuclear structures containing both constant [e.g., promyelocytic leukemia protein (PML), SP100, death domain-associated protein (Daxx)] and variable proteins, depending on the function of the cells or the stress to which they are exposed. In herpes simplex virus (HSV)-infected cells, ND10 bodies assemble at the sites of DNA entering the nucleus after infection. In sequence, the ND10 bodies become viral replication compartments, and ICP0, a viral E3 ligase, degrades both PML and SP100. The amounts of PML and SP100 and the number of ND10 structures increase in cells exposed to IFN-ß. Earlier studies have shown that PML has three key functions. Thus, (i) the interaction of PML with viral components facilitates the initiation of replication compartments, (ii) viral replication is significantly less affected by IFN-ß in PML-/- cells than in parental PML+/+ cells, and (iii) viral yields are significantly lower in PML-/- cells exposed to low ratios of virus per cell compared with parental PML+/+ cells. This report focuses on the function of SP100. In contrast to PML-/- cells, SP100-/- cells retain the sensitivity of parental SP100+/+ cells to IFN-ß and support replication of the ΔICP0 virus. At low multiplicities of infection, wild-type virus yields are higher in SP100-/- cells than in parental HEp-2 cells. In addition, the number of viral replication compartments is significantly higher in SP100-/- cells than in parental SP100+/+ cells or in PML-/- cells.


Assuntos
Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Estruturas do Núcleo Celular/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Simplexvirus/fisiologia , Montagem de Vírus/fisiologia , Animais , Antígenos Nucleares/genética , Autoantígenos/genética , Estruturas do Núcleo Celular/genética , Estruturas do Núcleo Celular/virologia , Chlorocebus aethiops , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína da Leucemia Promielocítica/genética , Células Vero
6.
Proc Natl Acad Sci U S A ; 114(33): E6902-E6911, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28760986

RESUMO

PUM1 is an RNA binding protein shown to regulate the stability and function of mRNAs bearing a specific sequence. We report the following: (i) A key function of PUM1 is that of a repressor of key innate immunity genes by repressing the expression of LGP2. Thus, between 12 and 48 hours after transfection of human cells with siPUM1 RNA there was an initial (phase 1) upsurge of transcripts encoding LGP2, CXCL10, IL6, and PKR. This was followed 24 hours later (phase 2) by a significant accumulation of mRNAs encoding RIG-I, SP100, MDA5, IFIT1, PML, STING, and IFNß. The genes that were not activated encoded HDAC4 and NF-κB1. (ii) Simultaneous depletion of PUM1 and LGP2, CXCL10, or IL6 revealed that up-regulation of phase 1 and phase 2 genes was the consequence of up-regulation of LGP2. (iii) IFNß produced 48-72 hours after transfection of siPUM1 was effective in up-regulating LGP2 and phase 2 genes and reducing the replication of HSV-1 in untreated cells. (iv) Because only half of genes up-regulated in phase 1 and 2 encode mRNAs containing PUM1 binding sites, the upsurge in gene expression could not be attributed solely to stabilization of mRNAs in the absence of PUM1. (v) Lastly, depletion of PUM2 does not result in up-regulation of phase 1 or phase 2 genes. The results of the studies presented here indicate that PUM1 is a negative regulator of LGP2, a master regulator of innate immunity genes expressed in a cascade fashion.


Assuntos
Regulação da Expressão Gênica , Imunidade Inata/genética , RNA Helicases/genética , Proteínas de Ligação a RNA/genética , Linhagem Celular Tumoral , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Humanos , Interferon beta/genética , Interferon beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , RNA Helicases/metabolismo , Interferência de RNA , Proteínas de Ligação a RNA/metabolismo
7.
Mol Ther ; 26(4): 1032-1039, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29526650

RESUMO

miRNAs are potent tools that in principle can be used to control the replication of infectious agents. The objectives of the studies reported here were to design miRNAs that can block the replication of herpes simplex virus 1 and which could be delivered to infected cells via exosomes. We report the following: (1) We designed three miRNAs targeting the mRNA encoding ICP4, an essential viral regulatory protein. Of the three miRNAs, one miRNA401 effectively blocked ICP4 accumulation and viral replication on transfection into susceptible cells. (2) To facilitate packaging of the miRNA into exosomes, we incorporated into the sequence of miRNA401 an exosome-packaging motif. miRNA401 was shown to be packaged into exosomes and successfully delivered by exosomes to susceptible cells, where it remained stable for at least 72 hr. Finally, the results show that miRNA401 delivered to cells via exosomes effectively reduced virus yields in a miRNA401 dose-dependent fashion. The protocol described in this report can be applied to study viral gene functions without actually deleting or mutagenizing the gene.


Assuntos
Regulação Viral da Expressão Gênica , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Proteínas Imediatamente Precoces/genética , MicroRNAs/genética , Interferência de RNA , Replicação Viral , Células Cultivadas , Exossomos/metabolismo , Vetores Genéticos/genética , Herpes Simples/metabolismo , Humanos , MicroRNAs/metabolismo , Transporte de RNA
8.
Proc Natl Acad Sci U S A ; 113(21): E3022-8, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27162364

RESUMO

After entry into the nucleus, herpes simplex virus (HSV) DNA is coated with repressive proteins and becomes the site of assembly of nuclear domain 10 (ND10) bodies. These small (0.1-1 µM) nuclear structures contain both constant [e.g., promyelocytic leukemia protein (PML), Sp100, death-domain associated protein (Daxx), and so forth] and variable proteins, depending on the function of the cells or the stress to which they are exposed. The amounts of PML and the number of ND10 structures increase in cells exposed to IFN-ß. On initiation of HSV-1 gene expression, ICP0, a viral E3 ligase, degrades both PML and Sp100. The earlier report that IFN-ß is significantly more effective in blocking viral replication in murine PML(+/+) cells than in sibling PML(-/-) cells, reproduced here with human cells, suggests that PML acts as an effector of antiviral effects of IFN-ß. To define more precisely the function of PML in HSV-1 replication, we constructed a PML(-/-) human cell line. We report that in PML(-/-) cells, Sp100 degradation is delayed, possibly because colocalization and merger of ICP0 with nuclear bodies containing Sp100 and Daxx is ineffective, and that HSV-1 replicates equally well in parental HEp-2 and PML(-/-) cells infected at 5 pfu wild-type virus per cell, but poorly in PML(-/-) cells exposed to 0.1 pfu per cell. Finally, ICP0 accumulation is reduced in PML(-/-) infected at low, but not high, multiplicities of infection. In essence, the very mechanism that serves to degrade an antiviral IFN-ß effector is exploited by HSV-1 to establish an efficient replication domain in the nucleus.


Assuntos
Herpesvirus Humano 1/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Replicação Viral/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Chlorocebus aethiops , Proteínas Correpressoras , Humanos , Proteínas Imediatamente Precoces/genética , Interferon beta/genética , Interferon beta/metabolismo , Corpos de Inclusão Intranuclear/genética , Corpos de Inclusão Intranuclear/virologia , Camundongos , Chaperonas Moleculares , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína da Leucemia Promielocítica/genética , Ubiquitina-Proteína Ligases/genética , Células Vero
9.
Proc Natl Acad Sci U S A ; 113(7): E894-901, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26831114

RESUMO

We report on the properties and function of two herpes simplex virus-1 (HSV-1) microRNAs (miRNAs) designated "miR-H28" and "miR-H29." Both miRNAs accumulate late in productive infection at a time when, for the most part, viral DNA and proteins have been made. Ectopic expression of miRNA mimics in human cells before infection reduced the accumulation of viral mRNAs and proteins, reduced plaque sizes, and at vey low multiplicities of infection reduced viral yields. The specificity of the miRNA mimics was tested in two ways. First, ectopic expression of mimics carrying mutations in the seed sequence was ineffective. Second, in similar tests two viral miRNAs made early in productive infection also had no effect. Both miR-H28 and miR-H29 are exported from infected cells in exosomes. A noteworthy finding is that both miR-H28 and miR-H29 were absent from murine ganglia harboring latent virus but accumulated in ganglia in which the virus was induced to reactivate. The significance of these findings rests on the principle that the transmission of HSV from person to person is by physical contact between the infected tissues of the donor and those of uninfected recipient. Diminished size of primary or recurrent lesions could be predicted to enhance person-to-person transmission. Reduction in the amount of reactivating latent virus would reduce the risk of retrograde transport to the CNS but would not interfere with anterograde transport to a site at or near the site of initial infection.


Assuntos
Herpesvirus Humano 1/fisiologia , MicroRNAs/genética , Replicação Viral/genética , Linhagem Celular , Humanos
10.
Annu Rev Microbiol ; 67: 355-74, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24024635

RESUMO

Herpes simplex virus (HSV) evolved an elegant strategy that enables the virus to impact a large fraction of the human population. The virus replicates at the portal of entry (mouth, genitals) and concurrently it is transported retrograde to sensory neurons. In sensory neurons it establishes a silent (latent) infection. A variety of stimuli can reactivate the virus. The reactivated virus is transmitted anterograde to a site at the portal of entry for transmission by physical contact between infected and uninfected tissues to other individuals. The central issue is how a virus that vigorously replicates and successfully blocks the innate immune defenses of the host at the portal of entry into the body remains silent in sensory neurons. The presentation focuses on three key issues: (a) current assessment of the impact of HSV on human health, (b) the mechanisms by which the virus overcomes a key host defense mechanism at the portal of entry into the body and yet is silenced in latently infected neurons, and (c) the mechanisms by which the virus reactivates.


Assuntos
Herpes Simples/virologia , Simplexvirus/fisiologia , Ativação Viral , Latência Viral , Animais , Herpes Simples/imunologia , Humanos , Simplexvirus/genética , Replicação Viral
11.
Proc Natl Acad Sci U S A ; 112(39): E5420-6, 2015 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-26305977

RESUMO

A key property of herpes simplex viruses (HSVs) is their ability to establish latent infection in sensory or autonomic ganglia and to reactivate on physical, hormonal, or emotional stress. In latently infected ganglia, HSVs express a long noncoding RNA, a latency-associated transcript (LAT), which plays a key role in maintaining latently infected neurons, but not viral proteins. To investigate the events leading to reactivation, we examined the use of ganglionic organ cultures that enable rapid reactivation in medium containing antibody to nerve growth factor (NGF) or delayed reactivation in medium containing NGF and epidermal growth factor (EGF). Here we report the discovery that activating transcription factor 3 (ATF3), a stress response protein, profoundly affects the interaction of HSV with its host. Specifically, (i) ATF3 is induced by stress, such as inhibition of protein synthesis or infection; (ii) in infected cells, ATF3 enhances the accumulation of LAT by acting on the response elements in the promoter of the LAT precursor RNA; (iii) ATF3 is induced nearly 100-fold in ganglionic organ cultures; and (iv) ATF3 plays a key role in the maintenance of the latent state, inasmuch as expression of ATF3 bereft of the C-terminal activation domain acts as a dominant negative factor, inducing HSV gene expression in ganglionic organ cultures harboring latent virus and incubated in medium containing NGF and EGF. Thus, ATF3 is a component of a cluster of cellular proteins that together with LAT maintain the integrity of the neurons harboring latent virus.


Assuntos
Fator 3 Ativador da Transcrição/metabolismo , Gânglios/virologia , Herpesvirus Humano 1/fisiologia , Latência Viral/fisiologia , Animais , Anticorpos Monoclonais , Primers do DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Células HEK293 , Humanos , Immunoblotting , Camundongos , Plasmídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Fisiológico/fisiologia , Estresse Psicológico/virologia
12.
Proc Natl Acad Sci U S A ; 112(1): E49-55, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25535379

RESUMO

The key events in herpes simplex virus (HSV) infections are (i) replication at a portal of entry into the body modeled by infection of cultured cells; (ii) establishment of a latent state characterized by a sole latency-associated transcript and microRNAs (miRNAs) modeled in murine peripheral ganglia 30 d after inoculation; and (iii) reactivation from the latent state modeled by excision and incubation of ganglia in medium containing anti-NGF antibody for a timespan of a single viral replicative cycle. In this report, we examine the pattern of synthesis and accumulation of 18 HSV-1 miRNAs in the three models. We report the following: (i) H2-3P, H3-3P, H4-3P, H5-3P, H6-3P, and H7-5P accumulated in ganglia harboring latent virus. All but H4-3P were readily detected in productively infected cells, and most likely they originate from three transcriptional units. (ii) H8-5P, H15, H17, H18, H26, and H27 accumulated during reactivation. Of this group, only H26 and H27 could be detected in productively infected cells. (iii) Of the 18 we have examined, only 10 miRNAs were found to accumulate above background levels in productively infected cells. The disparity in the accumulation of miRNAs in cell culture and during reactivation may reflect differences in the patterns of regulation of viral gene expression during productive infection and during reactivation from the latent state.


Assuntos
Herpes Simples/genética , Herpes Simples/virologia , MicroRNAs/metabolismo , Simplexvirus/fisiologia , Ativação Viral/genética , Latência Viral/genética , Animais , Feminino , Camundongos , MicroRNAs/genética , Modelos Biológicos , Gânglio Trigeminal/metabolismo , Gânglio Trigeminal/patologia , Gânglio Trigeminal/virologia
13.
Proc Natl Acad Sci U S A ; 111(5): E611-7, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24449861

RESUMO

STING (stimulator of IFN genes) activates the IFN pathway in response to cytosolic DNA. Knockout of STING in mice was reported to exacerbate the pathogenicity of herpes simplex virus 1 (HSV-1). Here we report the following: (i) STING is stable in cancer-derived HEp-2 or HeLa cells infected with wild-type HSV-1 but is degraded in cells infected with mutants lacking the genes encoding functional infected cell protein 0 (ICP0), ICP4, or the US3 protein kinase (US3-PK). In HEp-2 cells, depletion of STING by shRNA results in a decrease in the yields of wild-type or ΔICP0 viruses. (ii) STING is stable throughout infection with either wild-type or ICP0 mutant viruses in human embryonic lung cells (HEL) or HEK293T cells derived from normal tissues. In these cells, depletion of STING results in higher yields of both wild-type and ΔICP0 viruses. (iii) The US3-PK is also required for stabilization of IFI16, a nuclear DNA sensor. However, the stability of IFI16 does not correlate positively or negatively with that of STING. IFI16 is stable in STING-depleted HEL cells infected with wild-type virus. In contrast to HEL cells, IFI16 was undetectable in STING-depleted HEp-2 cells, and hence the role of HSV-1 in maintaining IFI16 could not be ascertained. The results indicate that in HSV-1-infected cells the stability of IFI16 and the function and stability of STING are dependent on cell derivation, the functional integrity of ICP0, and US3-PK, an indication that in wild-type virus-infected cells both proteins are actively stabilized. In HEp-2 cells, the stability of IFI16 requires STING.


Assuntos
Herpesvirus Humano 1/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais/metabolismo , Animais , Células HEK293 , Herpesvirus Humano 1/fisiologia , Humanos , Camundongos , Mutação/genética , Proteínas Quinases/metabolismo , Estabilidade Proteica , Replicação Viral
14.
Proc Natl Acad Sci U S A ; 111(46): E4991-6, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25368198

RESUMO

STING (stimulator of IFN genes) activates the IFN-dependent innate immune response to infection on sensing the presence of DNA in cytosol. The quantity of STING accumulating in cultured cells varies; it is relatively high in some cell lines [e.g., HEp-2, human embryonic lung fibroblasts (HEL), and HeLa] and low in others (e.g., Vero cells). In a preceding publication we reported that STING was stable in four cell lines infected with herpes simplex virus 1 and that it was actively stabilized in at least two cell lines derived from human cancers. In this report we show that STING is exported from HEp-2 cells to Vero cells along with virions, viral mRNAs, microRNAs, and the exosome marker protein CD9. The virions and exosomes copurified. The quantity of STING and CD9 exported from one cell line to another was inoculum-size-dependent and reflected the levels of STING and CD9 accumulating in the cells in which the virus inoculum was made. The export of STING, an innate immune sensor, and of viral mRNAs whose major role may be in silencing viral genes in latently infected neurons, suggests that the virus has evolved mechanisms that curtail rather than foster the spread of infection under certain conditions.


Assuntos
Exossomos/metabolismo , Fibroblastos/metabolismo , Fibroblastos/virologia , Herpesvirus Humano 1/patogenicidade , Interações Hospedeiro-Patógeno/fisiologia , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Animais , Transporte Biológico , Comunicação Celular/fisiologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Microambiente Celular , Chlorocebus aethiops , Exossomos/química , Herpes Simples/transmissão , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Humanos , Imunoprecipitação , Proteínas de Membrana/deficiência , Tetraspanina 29/análise , Células Vero , Proteínas Virais/genética , Proteínas Virais/fisiologia , Vírion/isolamento & purificação , Virulência , Ativação Viral , Replicação Viral
15.
Proc Natl Acad Sci U S A ; 111(4): E484-91, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24434553

RESUMO

An siRNA screen targeting 89 IFN stimulated genes in 14 different cancer cell lines pointed to the RIG-I (retinoic acid inducible gene I)-like receptor Laboratory of Genetics and Physiology 2 (LGP2) as playing a key role in conferring tumor cell survival following cytotoxic stress induced by ionizing radiation (IR). Studies on the role of LGP2 revealed the following: (i) Depletion of LGP2 in three cancer cell lines resulted in a significant increase in cell death following IR, (ii) ectopic expression of LGP2 in cells increased resistance to IR, and (iii) IR enhanced LGP2 expression in three cell lines tested. Studies designed to define the mechanism by which LGP2 acts point to its role in regulation of IFNß. Specifically (i) suppression of LGP2 leads to enhanced IFNß, (ii) cytotoxic effects following IR correlated with expression of IFNß inasmuch as inhibition of IFNß by neutralizing antibody conferred resistance to cell death, and (iii) mouse embryonic fibroblasts from IFN receptor 1 knockout mice are radioresistant compared with wild-type mouse embryonic fibroblasts. The role of LGP2 in cancer may be inferred from cumulative data showing elevated levels of LGP2 in cancer cells are associated with more adverse clinical outcomes. Our results indicate that cytotoxic stress exemplified by IR induces IFNß and enhances the expression of LGP2. Enhanced expression of LGP2 suppresses the IFN stimulated genes associated with cytotoxic stress by turning off the expression of IFNß.


Assuntos
Sobrevivência Celular/fisiologia , RNA Helicases DEAD-box/fisiologia , Neoplasias Experimentais/patologia , RNA Helicases/fisiologia , Radiação Ionizante , Animais , Apoptose , Neoplasias Encefálicas/patologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Glioblastoma/patologia , Humanos , Interferon Tipo I/biossíntese , Camundongos , Camundongos Knockout , Neoplasias Experimentais/metabolismo , RNA Helicases/metabolismo , Células Tumorais Cultivadas
17.
J Virol ; 89(10): 5643-50, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25762736

RESUMO

UNLABELLED: Herpes simplex viruses (HSV) package and bring into cells an RNase designated virion host shutoff (VHS) RNase. In infected cells, the VHS RNase targets primarily stress response mRNAs characterized by the presence of AU-rich elements in their 3' untranslated regions (UTRs). In uninfected cells, these RNAs are sequestered in exosomes or P bodies by host proteins that bind to the AU-rich elements. In infected cells, the AU-rich RNAs are deadenylated and cleaved close to the AU-rich elements, leading to long-term persistence of nontranslatable RNAs consisting of the 5' portions of the cleavage products. The host proteins that bind to the AU-rich elements are either resident in cells (e.g., TIA-1) or induced (e.g., tristetraprolin). Earlier, this laboratory reported that tristetraprolin binds VHS RNase. To test the hypothesis that tristetraprolin directs VHS RNase to the AU-rich elements, we mapped the domains of VHS and tristetraprolin required for their interactions. We report that VHS binds to the domain of tristetraprolin that enables its interaction with RNA. A single amino acid substitution in that domain abolished the interaction with RNA but did not block the binding to VHS RNase. In transfected cells, the mutant but not the wild-type tristetraprolin precluded the degradation of the AU-rich RNAs by VHS RNase. We conclude that TTP mediates the cleavage of the 3' UTRs of stress response mRNAs by recruiting the VHS RNase to the AU-rich elements. IMPORTANCE: The primary host response to HSV infection is the synthesis of stress response mRNAs characterized by the presence of AU-rich elements in their 3' UTRs. These mRNAs are the targets of the virion host shutoff (VHS) RNase. The VHS RNase binds both to mRNA cap structure and to tristetraprolin, an inducible host protein that sequesters AU-rich mRNAs in exosomes or P bodies. Here we show that tristetraprolin recruits VHS RNase to the AU-rich elements and enables the degradation of the stress response mRNAs.


Assuntos
Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonucleases/metabolismo , Tristetraprolina/metabolismo , Proteínas Virais/metabolismo , Regiões 3' não Traduzidas , Elementos Ricos em Adenilato e Uridilato , Sequência de Aminoácidos , Substituição de Aminoácidos , Proteínas Reguladoras de Apoptose/genética , Sequência de Bases , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Proteínas de Membrana/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ribonucleases/química , Ribonucleases/genética , Tristetraprolina/química , Tristetraprolina/genética , Proteínas Virais/química , Proteínas Virais/genética , Vírion/genética , Vírion/metabolismo
18.
Proc Natl Acad Sci U S A ; 110(6): E498-506, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23341636

RESUMO

HSVs transit from vigorous replication at the portal of entry into the body to a latent state in sensory neurons in which only noncoding (e.g., latency-associated transcript) and micro-RNAs are expressed. In productive infection, viral genes must be sequentially derepressed at two checkpoints. A leading role in the repression of viral genes is carried out by histone deacetylase (HDAC)/corepressor element-1 silencing transcription factor (CoREST)/lysinespecific demethylase1(LSD1)/RE1-silencing transcription factor (REST) repressor complex (HCLR). Previously, we reported that to define the role of the components of the HCLR complex in the establishment of latency, we constructed recombinant virus (R112) carrying a dominant-negative REST that bound response elements in DNA but could not recruit repressive proteins. This recombinant virus was unable to establish latency. In the current studies, we constructed a virus (R111) carrying WT REST with a WT genome. We report the following findings: (a) R111 readily established latent infection in trigeminal ganglia; however, although the amounts of viral DNAs in latently infected neurons were similar to those of WT virus, the levels of latency-associated transcript and micro-RNAs were 50- to 100-fold lower; (b) R111 did not spontaneously reactivate in ganglionic organ cultures; however, viral genes were expressed if the synthesis of REST was blocked by cycloheximide; and (c) histone deacetylase inhibitors reactivated the WT parent but not the R111 recombinant virus. The results suggest that REST plays a transient role in the establishment of latency but not in reactivation and suggest the existence of at least two phases at both establishment and reactivation.


Assuntos
Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidade , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Animais , Cicloeximida/farmacologia , Dexametasona/farmacologia , Feminino , Genes Virais , Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Camundongos , Camundongos Endogâmicos CBA , MicroRNAs/genética , MicroRNAs/metabolismo , Mutação , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Gânglio Trigeminal/virologia , Ativação Viral/genética , Ativação Viral/fisiologia , Latência Viral/genética , Latência Viral/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia
19.
Proc Natl Acad Sci U S A ; 110(28): E2621-8, 2013 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-23788661

RESUMO

A key property of herpes simplex viruses (HSVs) is their ability to establish latent infection in sensory or autonomic ganglia and to reactivate on physical, hormonal, or emotional stress. In latently infected ganglia, HSV expresses a long noncoding RNA and a set of microRNAs, but viral proteins are not expressed. The mechanism by which latent HSV reactivates is unknown. A key question is, what is the mechanism of reactivation in the absence of tegument proteins that enable gene expression in productive infection? Elsewhere we have reported the use of ganglionic organ cultures that enable rapid reactivation in medium containing antibody to NGF or delayed reactivation in medium containing NGF and EGF. We also reported that in the ganglionic organ cultures incubated in medium containing antibody to NGF, all viral genes are derepressed at once without requiring de novo protein synthesis within the time frame of a single replicative cycle. Here we report that latent HSV in ganglia immersed in medium containing NGF and EGF is reactivated by (i) broad spectrum as well as specific histone deacetylase 1 or histone deacetylase 4 inhibitors, (ii) activation of p300/CBP, and (iii) either STAT3 carrying the substitution of tyrosine 705 to phenylalanine or an inhibitor of STAT3. Conversely, reactivation of latent HSV was blocked by p300/CBP inhibitor in medium containing antibody to NGF. The results suggest that (i) STAT3 is required for the maintenance of the latent state and interference with its functions leads to reactivation and (ii) p300/CBP is essential for HSV reactivation.


Assuntos
Gânglios/virologia , Herpesvirus Humano 1/fisiologia , Fator de Transcrição STAT3/fisiologia , Ativação Viral , Fatores de Transcrição de p300-CBP/fisiologia , Humanos , Técnicas de Cultura de Órgãos , Fator de Transcrição STAT3/genética
20.
Proc Natl Acad Sci U S A ; 110(18): E1669-75, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23589852

RESUMO

Herpes simplex virus 1 (HSV-1) encodes an endoribonuclease that is responsible for the shutoff of host protein synthesis [virion host shutoff (VHS)-RNase]. The VHS-RNase released into cells during infection targets differentially four classes of mRNAs. Thus, (a) VHS-RNase degrades stable cellular mRNAs and α (immediate early) viral mRNAs; (b) it stabilizes host stress response mRNAs after deadenylation and subsequent cleavage near the adenylate-uridylate (AU)-rich elements; (c) it does not effectively degrade viral ß or γ mRNAs; and (d) it selectively spares from degradation a small number of cellular mRNAs. Current evidence suggests that several viral and at least one host protein (tristetraprolin) regulate its activity. Thus, virion protein (VP) 16 and VP22 neutralize the RNase activity at late times after infection. By binding to AU-rich elements via its interaction with tristetraprolin, the RNase deadenylates and cleaves the mRNAs in proximity to the AU-rich elements. In this report we show that another virion protein, UL47, brought into the cell during infection, attenuates the VHS-RNase activity with respect to stable host and viral α mRNAs and effectively blocks the degradation of ß and γ mRNAs, but it has no effect on the processing of AU-rich mRNAs. The properties of UL47 suggest that it, along with the α protein infected cell protein 27, attenuates degradation of mRNAs by the VHS-RNase through interaction with the enzyme in polyribosomes. Mutants lacking both VHS-RNase and UL47 overexpress α genes and delay the expression of ß and γ genes, suggesting that overexpression of α genes inhibits the downstream expression of early and late genes.


Assuntos
Herpesvirus Humano 1/metabolismo , Estabilidade de RNA , Ribonucleases/metabolismo , Proteínas Virais de Fusão/metabolismo , Proteínas Virais/metabolismo , Elementos Ricos em Adenilato e Uridilato/genética , Animais , Extratos Celulares , DNA Viral/metabolismo , Deleção de Genes , Células HEK293 , Herpes Simples/metabolismo , Herpes Simples/virologia , Humanos , Proteínas Imediatamente Precoces , Modelos Biológicos , Mutação/genética , RNA Viral/metabolismo , Reprodutibilidade dos Testes , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA