Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
Viruses ; 16(5)2024 05 08.
Article in English | MEDLINE | ID: mdl-38793628

ABSTRACT

Our current understanding of HSV latency is based on a variety of clinical observations, and in vivo, ex vivo, and in vitro model systems, each with unique advantages and drawbacks. The criteria for authentically modeling HSV latency include the ability to easily manipulate host genetics and biological pathways, as well as mimicking the immune response and viral pathogenesis in human infections. Although realistically modeling HSV latency is necessary when choosing a model, the cost, time requirement, ethical constraints, and reagent availability are also equally important. Presently, there remains a pressing need for in vivo models that more closely recapitulate human HSV infection. While the current in vivo, ex vivo, and in vitro models used to study HSV latency have limitations, they provide further insights that add to our understanding of latency. In vivo models have shed light on natural infection routes and the interplay between the host immune response and the virus during latency, while in vitro models have been invaluable in elucidating molecular pathways involved in latency. Below, we review the relative advantages and disadvantages of current HSV models and highlight insights gained through each.


Subject(s)
Herpes Simplex , Virus Latency , Humans , Herpes Simplex/virology , Animals , Simplexvirus/physiology , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/genetics , Disease Models, Animal
2.
Cell Rep Med ; 5(2): 101417, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38350452

ABSTRACT

Multiple failed herpes simplex virus (HSV) vaccine candidates induce robust neutralizing antibody (Ab) responses in clinical trials, raising the hypothesis that Fc-domain-dependent effector functions may be critical for protection. While neonatal HSV (nHSV) infection results in mortality and lifelong neurological morbidity in humans, it is uncommon among neonates with a seropositive birthing parent, supporting the hypothesis that Ab-based therapeutics could protect neonates from HSV. We therefore investigated the mechanisms of monoclonal Ab (mAb)-mediated protection in a mouse model of nHSV infection. For a panel of glycoprotein D (gD)-specific mAbs, neutralization and effector functions contributed to nHSV-1 protection. In contrast, effector functions alone were sufficient to protect against nHSV-2, exposing a functional dichotomy between virus types consistent with vaccine trial results. Effector functions are therefore crucial for protection by these gD-specific mAbs, informing effective Ab and vaccine design and demonstrating the potential of polyfunctional Abs as therapeutics for nHSV infections.


Subject(s)
Herpes Simplex , Viral Vaccines , Humans , Animals , Mice , Animals, Newborn , Antibodies, Viral , Herpes Simplex/prevention & control , Antibodies, Monoclonal/therapeutic use , Glycoproteins
3.
bioRxiv ; 2023 Aug 31.
Article in English | MEDLINE | ID: mdl-37693377

ABSTRACT

The failure of multiple herpes simplex virus (HSV) vaccine candidates that induce neutralizing antibody responses raises the hypothesis that other activities, such as Fc domain-dependent effector functions, may be critical for protection. While neonatal HSV (nHSV) infection result in mortality and lifelong neurological morbidity in humans, it is uncommon among neonates with a seropositive birthing parent, suggesting the potential efficacy of antibody-based therapeutics to protect neonates. We therefore investigated the mechanisms of monoclonal antibody (mAb)-mediated protection in a mouse model of nHSV infection. Both neutralization and effector functions contributed to robust protection against nHSV-1. In contrast, effector functions alone were sufficient to protect against nHSV-2, exposing a functional dichotomy between virus types that is consistent with vaccine trial results. Together, these results emphasize that effector functions are crucial for optimal mAb-mediated protection, informing effective Ab and vaccine design, and demonstrating the potential of polyfunctional Abs as potent therapeutics for nHSV infections.

4.
mBio ; 14(2): e0034123, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36946726

ABSTRACT

Transplacental transfer of maternal antibodies provides the fetus and newborn with passive protection against infectious diseases. While the role of the highly conserved neonatal Fc receptor (FcRn) in transfer of IgG in mammals is undisputed, recent reports have suggested that a second receptor may contribute to transport in humans. We report poor transfer efficiency of plant-expressed recombinant HIV-specific antibodies, including engineered variants with high FcRn affinity, following subcutaneous infusion into rhesus macaques close to parturition. Unexpectedly, unlike those derived from mammalian tissue culture, plant-derived antibodies were essentially unable to cross macaque placentas. This defect was associated with poor Fcγ receptor binding and altered Fc glycans and was not recapitulated in mice. These results suggest that maternal-fetal transfer of IgG across the three-layer primate placenta may require a second receptor and suggest a means of providing maternal antibody treatments during pregnancy while avoiding fetal harm. IMPORTANCE This study compared the ability of several human HIV envelope-directed monoclonal antibodies produced in plants with the same antibodies produced in mammalian cells for their ability to cross monkey and mouse placentas. We found that the two types of antibodies have comparable transfer efficiencies in mice, but they are differentially transferred across macaque placentas, consistent with a two-receptor IgG transport model in primates. Importantly, plant-produced monoclonal antibodies have excellent binding characteristics for human FcRn receptors, permitting desirable pharmacokinetics in humans. The lack of efficient transfer across the primate placenta suggests that therapeutic plant-based antibody treatments against autoimmune diseases and cancer could be provided to the mother while avoiding transfer and preventing harm to the fetus.


Subject(s)
HIV Infections , Placenta , Pregnancy , Female , Mice , Humans , Animals , Maternal-Fetal Exchange , Macaca mulatta , Immunoglobulin G , Receptors, Fc/metabolism , Antibodies, Monoclonal/metabolism , Histocompatibility Antigens Class I , HIV Infections/metabolism , Mammals/metabolism
5.
J Exp Med ; 219(12)2022 12 05.
Article in English | MEDLINE | ID: mdl-36156707

ABSTRACT

Neonatal herpes simplex virus (nHSV) infections often result in significant mortality and neurological morbidity despite antiviral drug therapy. Maternally transferred herpes simplex virus (HSV)-specific antibodies reduce the risk of clinically overt nHSV, but this observation has not been translationally applied. Using a neonatal mouse model, we tested the hypothesis that passive transfer of HSV-specific human mAbs can prevent mortality and morbidity associated with nHSV. The mAbs were expressed in vivo via vectored immunoprophylaxis or recombinantly. Through these maternally derived routes or through direct administration to pups, diverse mAbs to HSV glycoprotein D protected against neonatal HSV-1 and HSV-2 infection. Using in vivo bioluminescent imaging, both pre- and post-exposure mAb treatment significantly reduced viral load in mouse pups. Together these studies support the notion that HSV-specific mAb-based therapies could prevent or improve HSV infection outcomes in neonates.


Subject(s)
Herpes Simplex , Animals , Animals, Newborn , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral , Antiviral Agents , Glycoproteins , Humans , Mice , Morbidity , Pregnancy Complications, Infectious
6.
Front Immunol ; 13: 959603, 2022.
Article in English | MEDLINE | ID: mdl-36016956

ABSTRACT

The fetal/neonatal period represents both a unique window of opportunity for interventions as well as vulnerability to a number of viral infections. While Herpesviruses such as herpes simplex virus (HSV) are highly prevalent and typically of little consequence among healthy adults, they are among the most consequential infections of early life. Despite treatment with antiviral drugs, neonatal HSV (nHSV) infections can still result in significant mortality and lifelong neurological morbidity. Fortunately, newborns in our pathogen-rich world inherit some of the protection provided by the maternal immune system in the form of transferred antibodies. Maternal seropositivity, resulting in placental transfer of antibodies capable of neutralizing virus and eliciting the diverse effector functions of the innate immune system are associated with dramatically decreased risk of nHSV. Given this clear epidemiological evidence of reduced risk of infection and its sequelae, we present what is known about the ability of monoclonal antibody therapies to treat or prevent HSV infection and explore how effective antibody-based interventions in conjunction with antiviral therapy might reduce early life mortality and long-term morbidity.


Subject(s)
Communicable Diseases , Herpes Simplex , Pregnancy Complications, Infectious , Adult , Antibodies, Monoclonal/therapeutic use , Female , Humans , Infant, Newborn , Placenta , Pregnancy , Simplexvirus
7.
J Infect Dis ; 226(9): 1499-1509, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35451492

ABSTRACT

Herpes simplex virus (HSV) infection of the neonatal brain causes severe encephalitis and permanent neurologic deficits. However, infants infected with HSV at the time of birth follow varied clinical courses, with approximately half of infants experiencing only external infection of the skin rather than invasive neurologic disease. Understanding the cause of these divergent outcomes is essential to developing neuroprotective strategies. To directly assess the contribution of viral variation to neurovirulence, independent of human host factors, we evaluated clinical HSV isolates from neonates with different neurologic outcomes in neurologically relevant in vitro and in vivo models. We found that isolates taken from neonates with encephalitis are more neurovirulent in human neuronal culture and mouse models of HSV encephalitis, as compared to isolates collected from neonates with skin-limited disease. These findings suggest that inherent characteristics of the infecting HSV strain contribute to disease outcome following neonatal infection.


Subject(s)
Communicable Diseases , Encephalitis, Herpes Simplex , Herpes Simplex , Animals , Mice , Infant, Newborn , Humans , Herpesvirus 2, Human , Brain
8.
J Virol ; 94(14)2020 07 01.
Article in English | MEDLINE | ID: mdl-32376626

ABSTRACT

Expression of viral genes and activation of innate antiviral responses during infection result in an increase in reactive oxygen species (ROS) and toxic by-products of energy metabolism which can lead to cell death. The mitochondrion and its associated proteins are crucial regulators of these responses and related pathways such as autophagy and apoptosis. Through a mass spectrometry approach, we have shown that the herpes simplex virus 1 (HSV-1) neurovirulence- and autophagy-modulating protein ICP34.5 interacts with numerous mitochondrion-associated factors. Specifically, we showed that amino acids 68 to 87 of ICP34.5, the domain that binds beclin1 and controls neurovirulence, are necessary for interactions with PGAM5, KEAP1, and other regulators of the antioxidant response, mitochondrial trafficking, and programmed cell death. We further show that while this domain interacts with multiple cellular stress response factors, it does not alter apoptosis or antioxidant gene expression. That said, the attenuated replication of a recombinant virus lacking residues 68 to 87 (termed Δ68-87) in primary human fibroblasts was restored by addition of ferric nitrate. Furthermore, in primary mouse neurons, the perinuclear localization of mitochondria that follows infection with HSV-1 was notably absent following Δ68-87 infection. Through this 20-amino-acid domain, ICP34.5 significantly reduces mitochondrial motility in axons of neurons. We propose the hypothesis that ICP34.5 promotes perinuclear mitochondrial localization by modulating transport of mitochondria through interaction with PGAM5. These data expand upon previous observations of altered mitochondrial dynamics following alphaherpesvirus infections and identify a key determinant of this activity during HSV-1 infections.IMPORTANCE Herpes simplex virus persists lifelong in neurons and can reactivate to cause recurrent lesions in mucosal tissues. A key determinant of virulence is the viral protein ICP34.5, of which residues 68 to 87 significantly contribute to neurovirulence through an unknown mechanism. Our report provides evidence that residues 68 to 87 of ICP34.5 are required for binding mitochondrion-associated factors. These interactions alter mitochondrial dynamics in neurons, thereby facilitating viral replication and pathogenesis.


Subject(s)
Axons/metabolism , Herpes Simplex/metabolism , Herpesvirus 1, Human/metabolism , Mitochondria/metabolism , Viral Proteins/metabolism , Axons/pathology , Axons/virology , HEK293 Cells , Herpes Simplex/pathology , Herpesvirus 1, Human/genetics , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Domains , Protein Transport , Viral Proteins/genetics
9.
J Virol ; 94(11)2020 05 18.
Article in English | MEDLINE | ID: mdl-32188735

ABSTRACT

Herpes simplex virus (HSV) can cause severe infection in neonates leading to mortality and lifelong morbidity. Prophylactic approaches, such as maternal immunization, could prevent neonatal HSV (nHSV) infection by providing protective immunity and preventing perinatal transmission. We previously showed that maternal immunization with a replication-defective HSV vaccine candidate, dl5-29, leads to transfer of virus-specific antibodies into the neonatal circulation and protects against nHSV neurological sequela and mortality (C. D. Patel, I. M. Backes, S. A. Taylor, Y. Jiang, et al., Sci Transl Med, 11:eaau6039, 2019, https://doi.org/10.1126/scitranslmed.aau6039). In this study, we evaluated the efficacy of maternal immunization with an experimental trivalent (gC2, gD2, and gE2) subunit vaccine to protect against nHSV. Using a murine model of nHSV, we demonstrated that maternal immunization with the trivalent vaccine protected offspring against nHSV-disseminated disease and mortality. In addition, offspring of immunized dams were substantially protected from behavioral pathology following HSV infection. This study supports the idea that maternal immunization is a viable strategy for the prevention of neonatal infections.IMPORTANCE Herpes simplex virus is among the most serious infections of newborns. Current antiviral therapies can prevent mortality if infection is recognized early and treated promptly. Most children who survive nHSV develop lifelong neurological and behavioral deficits, despite aggressive antiviral treatment. We propose that maternal immunization could provide protection against HSV for both mother and baby. To this end, we used a trivalent glycoprotein vaccine candidate to demonstrate that offspring are protected from nHSV following maternal immunization. Significantly, this approach protected offspring from long-term behavioral morbidity. Our results emphasize the importance of providing protective immunity to neonates during this window of vulnerability.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human/immunology , Pregnancy Complications, Infectious , Animals , Animals, Newborn , Cell Line , Child , Herpes Simplex/immunology , Herpes Simplex/prevention & control , Humans , Infant, Newborn , Mice , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/prevention & control , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacology
10.
Cancer Immunol Res ; 7(7): 1079-1090, 2019 07.
Article in English | MEDLINE | ID: mdl-31088847

ABSTRACT

Tumor hypoxia is a negative prognostic factor that is implicated in oncogenic signal activation, immune escape, and resistance to treatment. Identifying the mechanistic role of hypoxia in immune escape and resistance to immune-checkpoint inhibitors may aid the identification of therapeutic targets. We and others have shown that V-domain Ig suppressor of T-cell activation (VISTA), a negative checkpoint regulator in the B7 family, is highly expressed in the tumor microenvironment in tumor models and primary human cancers. In this study, we show that VISTA and HIF1α activity are correlated in a cohort of colorectal cancer patients. High VISTA expression was associated with worse overall survival. We used the CT26 colon cancer model to investigate the regulation of VISTA by hypoxia. Compared with less hypoxic tumor regions or draining lymph nodes, regions of profound hypoxia in the tumor microenvironment were associated with increased VISTA expression on tumor-infiltrating myeloid-derived suppressor cells (MDSC). Using chromatin immunoprecipitation and genetic silencing, we show that hypoxia-inducible factor (HIF)-1α binding to a conserved hypoxia response element in the VISTA promoter upregulated VISTA on myeloid cells. Further, antibody targeting or genetic ablation of VISTA under hypoxia relieved MDSC-mediated T-cell suppression, revealing VISTA as a mediator of MDSC function. Collectively, these data suggest that targeting VISTA may mitigate the deleterious effects of hypoxia on antitumor immunity.


Subject(s)
Adenocarcinoma/immunology , B7 Antigens/metabolism , Colorectal Neoplasms/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/physiopathology , Myeloid-Derived Suppressor Cells/immunology , Tumor Microenvironment/immunology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Apoptosis , B7 Antigens/genetics , Case-Control Studies , Cell Proliferation , Cohort Studies , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Prognosis , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Cells, Cultured
11.
Sci Transl Med ; 11(487)2019 04 10.
Article in English | MEDLINE | ID: mdl-30971454

ABSTRACT

Neonatal herpes simplex virus (nHSV) infections cause devastating morbidity and mortality in infants. Most nHSV cases are associated with primary maternal infection, consistent with the hypothesis that maternal immunity is protective. In humans, we found HSV-specific neutralizing antibodies in newborns of immune mothers, indicating that placentally transferred HSV-specific antibody is protective. Using a murine model, we showed that passive administration of HSV-specific antibody to dams prevented disseminated infection and mortality in pups. Maternal immunization with an HSV-2 replication-defective vaccine candidate, dl5-29, led to transfer of HSV-specific antibodies into neonatal circulation that protected against nHSV neurological disease and death. Furthermore, we observed considerable anxiety-like behavior in adult mice that had been infected with low doses of HSV as neonates, despite a notable lack of signs of infection. This phenotype suggests that nHSV infection can have an unsuspected and permanent impact on behavior. These behavioral sequelae of nHSV were prevented by maternal immunization with dl5-29, demonstrating an unexpected benefit of immunization. These findings also support the general concept that maternal immunization can prevent neurotropic neonatal infections and associated morbidity and mortality.


Subject(s)
Behavior, Animal , Herpes Simplex/immunology , Herpes Simplex/prevention & control , Immunization , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/prevention & control , Animals , Animals, Newborn , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Anxiety/etiology , Female , Herpes Simplex/blood , Herpes Simplex/virology , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 1, Human/immunology , Humans , Immunoglobulin G/blood , Mice , Morbidity , Pregnancy , Pregnancy Complications, Infectious/blood , Pregnancy Complications, Infectious/virology , Trigeminal Ganglion/pathology , Trigeminal Ganglion/virology , Vaccination
12.
mBio ; 10(2)2019 03 05.
Article in English | MEDLINE | ID: mdl-30837340

ABSTRACT

Interferons (IFNs) and autophagy are critical neuronal defenses against viral infection. IFNs alter neuronal autophagy by promoting the accumulation of IFN-dependent LC3-decorated autophagic structures, termed LC3 clusters. Here, we analyzed LC3 clusters in sensory ganglia following herpes simplex virus 1 (HSV-1) infection. In the vicinity of acutely infected neurons, antigen-negative neurons contained structures resembling accumulated autophagosomes and autolysosomes that culminated in LC3 clusters. This accumulation reflects a delayed completion of autophagy. The endosomal sorting complexes required for transport (ESCRT) machinery participates in autophagosome closure and is also required for HSV-1 replication. In this study, our results showed that HSV-1 infection in vivo and in primary neurons caused a decrease in Vps4 (a key ESCRT pathway ATPase) RNA and protein with concomitant Stat1 activation and LC3 cluster induction. We also observed that IFNs were sufficient to decrease RNA and protein levels of Vps4 in primary neurons and in other cell types. The accumulation of ubiquitin was also observed at the LC3 cluster sites. Together, our results show that IFNs modulate the ESCRT machinery in neurons in response to HSV-1 infections.IMPORTANCE Neurons rely on IFNs and autophagy as major defenses against viral infections, and HSV must overcome such defenses in order to replicate. In addition to controlling host immunity, HSV must also control host membranes in order to complete its life cycle. HSV uses the host ESCRT membrane scission machinery for viral production and transport. Here we present evidence of a new IFN-dependent mechanism used by the host to prevent ESCRT subversion by HSV. This activity also impacts the dynamics of autophagy, possibly explaining the presence of recently described LC3 clusters in the HSV-infected nervous system. The induced accumulations of ubiquitin observed in these LC3 clusters resembled those observed in certain neurodegenerative diseases, suggesting possible mechanistic parallels between these conditions.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Herpes Simplex/pathology , Herpesvirus 1, Human/growth & development , Host-Pathogen Interactions , Interferons/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Autophagy , Cells, Cultured , Disease Models, Animal , Down-Regulation , Gene Expression Profiling , Mice , Neurons/pathology , Neurons/virology , STAT1 Transcription Factor/metabolism
13.
J Virol ; 93(9)2019 05 01.
Article in English | MEDLINE | ID: mdl-30760571

ABSTRACT

Herpes simplex virus 1 (HSV-1) cycles between phases of latency in sensory neurons and replication in mucosal sites. HSV-1 encodes two key proteins that antagonize the shutdown of host translation, US11 through preventing PKR activation and ICP34.5 through mediating dephosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α). While profound attenuation of ICP34.5 deletion mutants has been repeatedly demonstrated, a role for US11 in HSV-1 pathogenesis remains unclear. We therefore generated an HSV-1 strain 17 US11-null virus and examined its properties in vitro and in vivo In U373 glioblastoma cells, US11 cooperated with ICP34.5 to prevent eIF2α phosphorylation late in infection. However, the effect was muted in human corneal epithelial cells (HCLEs), which did not accumulate phosphorylated eIF2α unless both US11 and ICP34.5 were absent. Low levels of phosphorylated eIF2α correlated with continued protein synthesis and with the ability of virus lacking US11 to overcome antiviral immunity in HCLE and U373 cells. Neurovirulence following intracerebral inoculation of mice was not affected by the deletion of US11. In contrast, the time to endpoint criteria following corneal infection was greater for the US11-null virus than for the wild-type virus. Replication in trigeminal ganglia and periocular tissue was promoted by US11, as was periocular disease. The establishment of latency and the frequency of virus reactivation from trigeminal ganglia were unaffected by US11 deletion, although emergence of the US11-null virus occurred with slowed kinetics. Considered together, the data indicate that US11 facilitates the countering of antiviral response of infected cells and promotes the efficient emergence of virus following reactivation.IMPORTANCE Alphaherpesviruses are ubiquitous DNA viruses and include the human pathogens herpes simplex virus 1 (HSV-1) and HSV-2 and are significant causes of ulcerative mucosal sores, infectious blindness, encephalitis, and devastating neonatal disease. Successful primary infection and persistent coexistence with host immune defenses are dependent on the ability of these viruses to counter the antiviral response. HSV-1 and HSV-2 and other primate viruses within the Simplexvirus genus encode US11, an immune antagonist that promotes virus production by preventing shutdown of protein translation. Here we investigated the impact of US11 deletion on HSV-1 growth in vitro and pathogenesis in vivo This work supports a role for US11 in pathogenesis and emergence from latency, elucidating immunomodulation by this medically important cohort of viruses.


Subject(s)
Epithelium, Corneal/metabolism , Herpesvirus 1, Human , Keratitis, Herpetic/metabolism , RNA-Binding Proteins/metabolism , Trigeminal Ganglion/metabolism , Viral Proteins/metabolism , Virus Activation/physiology , Virus Latency/physiology , Animals , Cell Line, Tumor , Chlorocebus aethiops , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Epithelium, Corneal/pathology , Epithelium, Corneal/virology , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Gene Deletion , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/physiology , Humans , Keratitis, Herpetic/genetics , Keratitis, Herpetic/pathology , Keratitis, Herpetic/virology , Phosphorylation , RNA-Binding Proteins/genetics , Trigeminal Ganglion/pathology , Trigeminal Ganglion/virology , Vero Cells , Viral Proteins/genetics
14.
Virology ; 529: 23-28, 2019 03.
Article in English | MEDLINE | ID: mdl-30648635

ABSTRACT

Herpes simplex virus (HSV)- 1 is the most common cause of sporadic viral encephalitis and accounts for 5-10% of cases worldwide. A key factor in host control of viral infection is the initiation of the interferon (IFN) response, mediated in part by the stimulator of interferon genes (STING) pathway. In these studies, we examined the ability of 5,6-dimethylxanthenone-4-acetic acid (DMXAA), a STING agonist, to protect against HSV-1 infection. DMXAA reduced viral replication through increased production of type I IFN in vitro. Furthermore, administration of DMXAA to HSV-1 infected mice resulted in a reduction of viral burden in the peripheral and central nervous systems. This reduced viral burden also correlated with increased survival of DMXAA-treated infected mice. These results therefore demonstrate the potential of STING agonists for immunotherapy against HSV-1.


Subject(s)
Central Nervous System Viral Diseases/prevention & control , Herpes Simplex , Membrane Proteins/agonists , Simplexvirus , Xanthones/therapeutic use , Animals , Cells, Cultured , Female , Fibroblasts/virology , Gene Expression Regulation/drug effects , Interferon-beta/genetics , Interferon-beta/metabolism , Male , Mice , Mice, Inbred C57BL , Virus Replication/drug effects
15.
J Virol ; 92(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29643250

ABSTRACT

Herpes simplex virus (HSV) latency in neurons remains poorly understood, and the heterogeneity of the sensory nervous system complicates mechanistic studies. In this study, we used primary culture of adult trigeminal ganglion (TG) mouse neurons in microfluidic devices and an in vivo model to examine the subtypes of sensory neurons involved in HSV latency. HSV-infected neurofilament heavy-positive (NefH+) neurons were more likely to express latency-associated transcripts (LATs) than infected neurofilament heavy-negative (NefH-) neurons. This differential expression of the LAT promoter correlated with differences in HSV-1 early infection that manifested as differences in the efficiency with which HSV particles reached the cell body following infection at the distal axon. In vivo, we further identified a specific subset of NefH+ neurons which coexpressed calcitonin gene-related peptide α (NefH+ CGRP+ neurons) as the sensory neuron subpopulation with the highest LAT promoter activity following HSV-1 infection. Finally, an early-phase reactivation assay showed HSV-1 reactivating in NefH+ CGRP+ neurons, although other sensory neuron subpopulations were also involved. Together, these results show that sensory neurons expressing neurofilaments exhibit enhanced LAT promoter activity. We hypothesize that the reduced efficiency of HSV-1 invasion at an early phase of infection may promote efficient establishment of latency in NefH+ neurons due to initiation of the antiviral state preceding arrival of the virus at the neuronal cell body. While the outcome of HSV-1 infection of neurons is determined by a broad variety of factors in vivo, neuronal subtypes are likely to play differential roles in modulating the establishment of latent infection.IMPORTANCE Two pivotal properties of HSV-1 make it a successful pathogen. First, it infects neurons, which are immune privileged. Second, it establishes latency in these neurons. Together, these properties allow HSV to persist for the lifetime of its host. Neurons are diverse and highly organized cells, with specific anatomical, physiological, and molecular characteristics. Previous work has shown that establishment of latency by HSV-1 does not occur equally in all types of neurons. Our results show that the kinetics of HSV infection and the levels of latency-related gene expression differ in certain types of neurons. The neuronal subtype infected by HSV is therefore a critical determinant of the outcome of infection and latency.


Subject(s)
Herpesvirus 1, Human/physiology , MicroRNAs/genetics , Sensory Receptor Cells/cytology , Calcitonin Gene-Related Peptide/metabolism , Cell Culture Techniques , Cells, Cultured , Gene Expression Regulation, Viral , Intermediate Filaments/metabolism , Microfluidic Analytical Techniques , Promoter Regions, Genetic , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/virology , Virus Latency
16.
J Virol ; 91(23)2017 12 01.
Article in English | MEDLINE | ID: mdl-28904192

ABSTRACT

During viral infection, pattern recognition receptors (PRRs) and their associated adaptors recruit TANK-binding kinase 1 (TBK1) to activate interferon regulatory factor 3 (IRF3), resulting in production of type I interferons (IFNs). ICP0 and ICP34.5 are among the proteins encoded by herpes simplex virus 1 (HSV-1) that modulate type I IFN signaling. We constructed a recombinant virus (ΔXX) that lacks amino acids 87 to 106, a portion of the previously described TBK1-binding domain of the γ34.5 gene (D. Verpooten, Y. Ma, S. Hou, Z. Yan, and B. He, J Biol Chem 284:1097-1105, 2009, https://doi.org/10.1074/JBC.M805905200). These 20 residues are outside the γ34.5 beclin1-binding domain (BBD) that interacts with beclin1 and regulates autophagy. Unexpectedly, ΔXX showed no deficit in replication in vivo in a variety of tissues and showed virulence comparable to that of wild-type and marker-rescued viruses following intracerebral infection. ΔXX was fully capable of mediating the dephosphorylation of eIF2α, and the virus was capable of controlling the phosphorylation of IRF3. In contrast, a null mutant in γ34.5 failed to control IRF3 phosphorylation due to an inability of the mutant to sustain expression of ICP0. Our data show that while γ34.5 regulates IRF3 phosphorylation, the TBK1-binding domain itself has no impact on IRF3 phosphorylation or on replication and pathogenesis in mice.IMPORTANCE Interferons (IFNs) are potent activators of a variety of host responses that serve to control virus infections. The Herpesviridae have evolved countermeasures to IFN responses. Herpes simplex virus 1 (HSV-1) encodes the multifunctional neurovirulence protein ICP34.5. In this study, we investigated the biological relevance of the interaction between ICP34.5 and TANK-binding kinase 1 (TBK1), an activator of IFN responses. Here, we establish that although ICP34.5 binds TBK1 under certain conditions through a TBK1-binding domain (TBD), there was no direct impact of the TBD on viral replication or virulence in mice. Furthermore, we showed that activation of IRF3, a substrate of TBK1, was independent of the TBD. Instead, we provided evidence that the ability of ICP34.5 to control IRF3 activation is through its ability to reverse translational shutoff and sustain the expression of other IFN inhibitors encoded by the virus. This work provides new insights into the immunomodulatory functions of ICP34.5.


Subject(s)
Herpesvirus 1, Human/metabolism , Host-Pathogen Interactions , Interferon Regulatory Factor-3/metabolism , Signal Transduction , Viral Proteins/metabolism , Animals , Beclin-1/metabolism , Chlorocebus aethiops , Fibroblasts/drug effects , Fibroblasts/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/pathogenicity , Humans , Immunity, Innate , Interferon-beta/pharmacology , Interferons/metabolism , Mice , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication/genetics
17.
Methods Mol Biol ; 1656: 229-251, 2017.
Article in English | MEDLINE | ID: mdl-28808974

ABSTRACT

Cultured primary neurons have been of extraordinary value for the study of neuronal anatomy, cell biology, and physiology. While use of neuronal cell lines has ease and utility, there are often caveats that arise due to their mitotic nature. This methods article presents detailed methodology for the preparation, purification, and culture of adult murine sensory neurons for the study of herpes simplex virus lytic and latent infections. While virology is the application for our laboratory, these cultures also have broad utility for neurobiologists and cell biologists. While these primary cultures have been highly informative, the methodology is challenging to many investigators. Through publication of this highly detailed protocol, it is our hope that the use of this culture system can spread in the field to allow more rapid progress in furthering our understanding of neurotropic virus infection.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Herpes Simplex/immunology , Sensory Receptor Cells/immunology , Simplexvirus/physiology , Virus Latency/immunology , Animals , Mice , Sensory Receptor Cells/virology
18.
mBio ; 8(4)2017 07 05.
Article in English | MEDLINE | ID: mdl-28679745

ABSTRACT

While antibody responses to neurovirulent pathogens are critical for clearance, the extent to which antibodies access the nervous system to ameliorate infection is poorly understood. In this study on herpes simplex virus 1 (HSV-1), we demonstrate that HSV-specific antibodies are present during HSV-1 latency in the nervous systems of both mice and humans. We show that antibody-secreting cells entered the trigeminal ganglion (TG), a key site of HSV infection, and persisted long after the establishment of latent infection. We also demonstrate the ability of passively administered IgG to enter the TG independently of infection, showing that the naive TG is accessible to antibodies. The translational implication of this finding is that human fetal neural tissue could contain HSV-specific maternally derived antibodies. Exploring this possibility, we observed HSV-specific IgG in HSV DNA-negative human fetal TG, suggesting passive transfer of maternal immunity into the prenatal nervous system. To further investigate the role of maternal antibodies in the neonatal nervous system, we established a murine model to demonstrate that maternal IgG can access and persist in neonatal TG. This maternal antibody not only prevented disseminated infection but also completely protected the neonate from neurological disease and death following HSV challenge. Maternal antibodies therefore have a potent protective role in the neonatal nervous system against HSV infection. These findings strongly support the concept that prevention of prenatal and neonatal neurotropic infections can be achieved through maternal immunization.IMPORTANCE Herpes simplex virus 1 is a common infection of the nervous system that causes devastating neonatal disease. Using mouse and human tissue, we discovered that antiviral antibodies accumulate in neural tissue after HSV-1 infection in adults. Similarly, these antibodies pass to the offspring during pregnancy. We found that antiviral maternal antibodies can readily access neural tissue of the fetus and neonate. These maternal antibodies then protect neonatal mice against HSV-1 neurological infection and death. These results underscore the previously unappreciated role of maternal antibodies in protecting fetal and newborn nervous systems against infection. These data suggest that maternal immunization would be efficacious at preventing fetal/neonatal neurological infections.


Subject(s)
Antibodies, Viral/immunology , Herpes Simplex/prevention & control , Herpesvirus 1, Human/immunology , Immunity, Maternally-Acquired , Nervous System/immunology , Trigeminal Ganglion/immunology , Animals , Animals, Newborn , Antibodies, Viral/biosynthesis , Female , Herpes Simplex/immunology , Humans , Immunization, Passive , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Infant, Newborn , Infectious Disease Transmission, Vertical/prevention & control , Mice , Mothers , Pregnancy , Virus Latency
19.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27795407

ABSTRACT

Neuroinvasive herpesviruses have evolved to efficiently infect and establish latency in neurons. The nervous system has limited capability to regenerate, so immune responses therein are carefully regulated to be nondestructive, with dependence on atypical intrinsic and innate defenses. In this article we review studies of some of these noncanonical defense pathways and how herpesvirus gene products counter them, highlighting the contributions that primary neuronal in vitro models have made to our understanding of this field.


Subject(s)
Gene Silencing , Herpesviridae/growth & development , Immune Evasion , Neurons/virology , Virus Latency/immunology , Autophagy/genetics , Autophagy/immunology , Axonal Transport , Co-Repressor Proteins/genetics , Co-Repressor Proteins/immunology , Herpesviridae/immunology , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Histone Demethylases/genetics , Histone Demethylases/immunology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Immunity, Innate , Interferons/genetics , Interferons/immunology , MicroRNAs/genetics , MicroRNAs/immunology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Neurons/immunology , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology
20.
Methods Mol Biol ; 1656: E3, 2017.
Article in English | MEDLINE | ID: mdl-30350167

ABSTRACT

In chapter 15, section 2.3, the unit "180 µg/ml mouse laminin in HBSS. Prepare 150 µl per coverslip" is corrected to "18 µg/ml mouse laminin in HBSS. Prepare 150 µl per coverslip."

SELECTION OF CITATIONS
SEARCH DETAIL