Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
J Virol ; 94(13)2020 06 16.
Article in English | MEDLINE | ID: mdl-32295924

ABSTRACT

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.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/metabolism , Herpesvirus 1, Human/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Animals , Chlorocebus aethiops , Exosomes/metabolism , HEK293 Cells , Herpes Simplex/virology , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/pathogenicity , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , MicroRNAs/genetics , RNA, Messenger/genetics , Vero Cells , Viral Proteins/metabolism , Virus Replication
3.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31413129

ABSTRACT

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.


Subject(s)
Antiviral Agents/metabolism , Herpes Simplex/transmission , Herpesvirus 1, Human/physiology , Interferon-gamma/metabolism , MicroRNAs/genetics , RNA, Messenger/metabolism , Virus Replication/drug effects , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Cell Communication , HEK293 Cells , Herpes Simplex/metabolism , Herpes Simplex/virology , Humans , Interferon-gamma/genetics , Laryngeal Neoplasms/immunology , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/pathology , Laryngeal Neoplasms/virology , RNA, Messenger/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
4.
J Virol ; 93(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30700604

ABSTRACT

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.


Subject(s)
Cell Cycle Proteins/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Immunity, Innate/immunology , Nuclear Proteins/immunology , Animals , Chlorocebus aethiops , Histone Deacetylases/immunology , Membrane Proteins/immunology , RNA Helicases/immunology , RNA, Small Interfering/immunology , Vero Cells , Virus Replication/immunology
5.
Proc Natl Acad Sci U S A ; 115(14): E3230-E3237, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29559532

ABSTRACT

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.


Subject(s)
Biomarkers/analysis , Gene Regulatory Networks , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Immunity, Innate/genetics , Laryngeal Neoplasms/immunology , Virus Replication/genetics , Gene Expression Regulation , Gene Knockout Techniques , Herpes Simplex/genetics , Herpes Simplex/virology , Humans , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/virology , Tumor Cells, Cultured
6.
Mol Ther ; 26(4): 1032-1039, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29526650

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Viral , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Immediate-Early Proteins/genetics , MicroRNAs/genetics , RNA Interference , Virus Replication , Cells, Cultured , Exosomes/metabolism , Genetic Vectors/genetics , Herpes Simplex/metabolism , Humans , MicroRNAs/metabolism , RNA Transport
7.
Proc Natl Acad Sci U S A ; 114(33): E6902-E6911, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28760986

ABSTRACT

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.


Subject(s)
Gene Expression Regulation , Immunity, Innate/genetics , RNA Helicases/genetics , RNA-Binding Proteins/genetics , Cell Line, Tumor , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , RNA Helicases/metabolism , RNA Interference , RNA-Binding Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 114(19): E3823-E3829, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28439026

ABSTRACT

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.


Subject(s)
Antigens, Nuclear/metabolism , Autoantigens/metabolism , Cell Nucleus Structures/metabolism , Promyelocytic Leukemia Protein/metabolism , Simplexvirus/physiology , Virus Assembly/physiology , Animals , Antigens, Nuclear/genetics , Autoantigens/genetics , Cell Nucleus Structures/genetics , Cell Nucleus Structures/virology , Chlorocebus aethiops , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/genetics , Vero Cells
9.
Proc Natl Acad Sci U S A ; 113(21): E3022-8, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27162364

ABSTRACT

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.


Subject(s)
Herpesvirus 1, Human/physiology , Immediate-Early Proteins/metabolism , Intranuclear Inclusion Bodies/metabolism , Promyelocytic Leukemia Protein/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Chlorocebus aethiops , Co-Repressor Proteins , Humans , Immediate-Early Proteins/genetics , Interferon-beta/genetics , Interferon-beta/metabolism , Intranuclear Inclusion Bodies/genetics , Intranuclear Inclusion Bodies/virology , Mice , Molecular Chaperones , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/genetics , Ubiquitin-Protein Ligases/genetics , Vero Cells
10.
Proc Natl Acad Sci U S A ; 113(7): E894-901, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26831114

ABSTRACT

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.


Subject(s)
Herpesvirus 1, Human/physiology , MicroRNAs/genetics , Virus Replication/genetics , Cell Line , Humans
11.
Proc Natl Acad Sci U S A ; 112(39): E5420-6, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26305977

ABSTRACT

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.


Subject(s)
Activating Transcription Factor 3/metabolism , Ganglia/virology , Herpesvirus 1, Human/physiology , Virus Latency/physiology , Animals , Antibodies, Monoclonal , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , HEK293 Cells , Humans , Immunoblotting , Mice , Plasmids/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological/physiology , Stress, Psychological/virology
12.
J Virol ; 89(10): 5643-50, 2015 May.
Article in English | MEDLINE | ID: mdl-25762736

ABSTRACT

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.


Subject(s)
Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleases/metabolism , Tristetraprolin/metabolism , Viral Proteins/metabolism , 3' Untranslated Regions , AU Rich Elements , Amino Acid Sequence , Amino Acid Substitution , Apoptosis Regulatory Proteins/genetics , Base Sequence , HEK293 Cells , Host-Pathogen Interactions , Humans , Membrane Proteins/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribonucleases/chemistry , Ribonucleases/genetics , Tristetraprolin/chemistry , Tristetraprolin/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Virion/genetics , Virion/metabolism
13.
Virology ; 479-480: 562-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25771487

ABSTRACT

On entry into the body herpes simplex viruses (HSV) replicate in a series of steps that involves derepression of viral DNA activated by VP16, a virion protein, and sequential transcription of viral genes in a cascade fashion. HSV also enters into neurons in which viral DNA maintained as heterochromatin and with few exceptions viral gene expression is silenced. A third face of the interaction of HSV with its host cells takes place at the moment when the silenced viral genome in neurons is abruptly derepressed. The available data do no reveal evidence that HSV encodes different regulatory programs for each facet of its interaction with its host cells. Rather the data point to significant gaps in our knowledge of the mechanisms by which each facet is initiated and the roles of the infected cells at each facet of the interaction of viral gene products with the host cell.


Subject(s)
Gene Expression Regulation, Viral , Simplexvirus/genetics , Simplexvirus/physiology , Virus Latency , Virus Replication
14.
Annu Rev Virol ; 2(1): 1-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26958904

ABSTRACT

I was shaped by World War II, years of near starvation as a war refugee, postwar chaos, life in several countries, and relative affluence in later life. The truth is that as I was growing up I wanted to be a writer. My aspirations came to an end when, in order to speed up my graduation from college, I took courses in microbiology. It was my second love at first sight-that of my wife preceded it. I view science as an opportunity to discover the designs in the mosaics of life. What initiates my search of discovery is an observation that makes no sense unless there exists a novel design. Once the design is revealed there is little interest in filling all the gaps. I was fortunate to understand that what lasts are not the scientific reports but rather the generations of scientists whose education I may have influenced.


Subject(s)
Laboratory Personnel/history , Science/history , Virology/history , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , History, 20th Century , History, 21st Century , Humans , Male , Romania , Workforce
15.
Proc Natl Acad Sci U S A ; 112(1): E49-55, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25535379

ABSTRACT

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.


Subject(s)
Herpes Simplex/genetics , Herpes Simplex/virology , MicroRNAs/metabolism , Simplexvirus/physiology , Virus Activation/genetics , Virus Latency/genetics , Animals , Female , Mice , MicroRNAs/genetics , Models, Biological , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/pathology , Trigeminal Ganglion/virology
16.
Proc Natl Acad Sci U S A ; 111(46): E4991-6, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368198

ABSTRACT

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.


Subject(s)
Exosomes/metabolism , Fibroblasts/metabolism , Fibroblasts/virology , Herpesvirus 1, Human/pathogenicity , Host-Pathogen Interactions/physiology , Membrane Proteins/metabolism , MicroRNAs/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Animals , Biological Transport , Cell Communication/physiology , Cell Line, Transformed , Cell Line, Tumor , Cellular Microenvironment , Chlorocebus aethiops , Exosomes/chemistry , Herpes Simplex/transmission , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Humans , Immunoprecipitation , Membrane Proteins/deficiency , Tetraspanin 29/analysis , Vero Cells , Viral Proteins/genetics , Viral Proteins/physiology , Virion/isolation & purification , Virulence , Virus Activation , Virus Replication
17.
J Virol ; 88(10): 5437-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24574411

ABSTRACT

UNLABELLED: Herpes simplex virus 1 (HSV-1)-infected cell protein 0 (ICP0) is a multifunctional protein that plays a key role in overcoming numerous facets of host innate immunity. A key function of ICP0 that requires an intact RING finger domain is that of an ubiquitin E3 ligase: ICP0 interacts with at least three ubiquitin-conjugating enzymes of which one, UbcH5a, is required for degradation of PML and SP100. A preceding report showed that ICP0 is highly unstable at very early times after infection but becomes stable at later times. We report here that (i) the degradation of ICP0 is not infected cell specific, (ii) the degradation does not require the interaction of ICP0 with either UbcH5a, UbcH6, or UbcH9, (iii) ICP0 is degraded both early and late in cells infected with a mutant lacking the UL13 protein kinase, (iv) ICP0 encoded by wild-type virus or the ΔUL13 mutant is stable in cells transfected with a plasmid encoding UL13 before infection, (v) ICP0 carrying mutations in the RING finger domain is stable both early and late in infection, and, finally, (vi) in cells infected with both wild type and RING finger mutant only the wild-type ICP0 is rapidly degraded at early times. The results suggest that the stability of ICP0 is mediated by the UL13 protein kinase and that the target of proteolysis is a site at or near the RING domain of ICP0. IMPORTANCE: ICP0, a major regulatory protein of HSV-1, turns over rapidly early in infection but becomes stable at late times. We report that stabilization requires the presence of UL13 protein kinase and that an ICP0 with mutations in RING finger is stable. In mixed infections mutant ICP0 is stable, whereas the wild-type ICP0 is degraded. Our findings suggest that the lifestyle of HSV-1 requires an ICP0 that turns over rapidly if late proteins are absent.


Subject(s)
Herpesvirus 1, Human/physiology , Immediate-Early Proteins/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication , Animals , Cell Line , Humans , Proteolysis , RING Finger Domains
18.
Proc Natl Acad Sci U S A ; 111(5): E611-7, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449861

ABSTRACT

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.


Subject(s)
Herpesvirus 1, Human/metabolism , Immediate-Early Proteins/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , Animals , HEK293 Cells , Herpesvirus 1, Human/physiology , Humans , Mice , Mutation/genetics , Protein Kinases/metabolism , Protein Stability , Virus Replication
19.
Proc Natl Acad Sci U S A ; 111(4): E484-91, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24434553

ABSTRACT

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.


Subject(s)
Cell Survival/physiology , DEAD-box RNA Helicases/physiology , Neoplasms, Experimental/pathology , RNA Helicases/physiology , Radiation, Ionizing , Animals , Apoptosis , Brain Neoplasms/pathology , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , Glioblastoma/pathology , Humans , Interferon Type I/biosynthesis , Mice , Mice, Knockout , Neoplasms, Experimental/metabolism , RNA Helicases/metabolism , Tumor Cells, Cultured
20.
J Virol ; 87(24): 13569-78, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24109211

ABSTRACT

The herpes simplex virus host shutoff RNase (VHS-RNase) is the major early block of host responses to infection. VHS-RNase is introduced into cells during infection and selectively degrades stable mRNAs made before infection and the normally short-lived AU-rich stress response mRNAs induced by sensors of innate immunity. Through its interactions with pUL47, another tegument protein, it spares from degradation viral mRNAs. Analyses of embedded motifs revealed that VHS-RNase contains a nuclear export signal (NES) but not a nuclear localization signal. To reconcile the potential nuclear localization with earlier studies showing that VHS-RNase degrades mRNAs in polyribosomes, we constructed a mutant in which NES was ablated. Comparison of the mutant and wild-type VHS-RNases revealed the following. (i) On infection, VHS-RNase is transported to the nucleus, but only the wild-type protein shuttles between the nucleus and cytoplasm. (ii) Both VHS-RNases localized in the cytoplasm following transfection. On cotransfection with pUL47, a fraction of VHS-RNase was translocated to the nucleus, suggesting that pUL47 may enable nuclear localization of VHS-RNase. (iii) In infected cells, VHS-RNase lacking NES degraded the short-lived AU-rich mRNAs but not the stable mRNAs. In transfected cells, both wild-type and NES mutant VHS-RNases effectively degraded cellular mRNAs. Our results suggest that the stable mRNAs are degraded in the cytoplasm, whereas the AU-rich mRNAs may be degraded in both cellular compartments. The selective sparing of viral mRNAs may take place during the nuclear phase in the course of interaction of pUL47, VHS-RNase, and nascent viral mRNAs.


Subject(s)
Cell Nucleus/virology , Cytoplasm/virology , Herpes Simplex/genetics , Herpesvirus 1, Human/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleases/metabolism , Viral Fusion Proteins/metabolism , Viral Proteins/metabolism , AT Rich Sequence , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/chemistry , Herpesvirus 1, Human/genetics , Humans , Nuclear Export Signals , Protein Transport , RNA Stability , RNA, Messenger/chemistry , Ribonucleases/chemistry , Ribonucleases/genetics , Viral Fusion Proteins/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...