Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.869
Filter
1.
Arch Microbiol ; 206(6): 269, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38767708

ABSTRACT

Bacteriocins are ribosomally synthesized bacterial peptides endowed with antibacterial, antiprotozoal, anticancer and antiviral activities. In the present study, we evaluated the antiviral activities of two bacteriocins, enterocin DD14 (EntDD14) and lacticaseicin 30, against herpes simplex virus type 1 (HSV-1), human coronavirus 229E (HCoV-229E) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Vero, Huh7 and Vero E6 cells, respectively. In addition, the interactions of these bacteriocins with the envelope glycoprotein D of HSV-1 and the receptor binding domains of HCoV-229E and SARS-CoV-2 have been computationally evaluated using protein-protein docking and molecular dynamics simulations. HSV-1 replication in Vero cells was inhibited by EntDD14 and, to a lesser extent, by lacticaseicin 30 added to cells after virus inoculation. EntDD14 and lacticaseicin 30 had no apparent antiviral activity against HCoV-229E; however, EntDD14 was able to inhibit SARS-CoV-2 in Vero E6 cells. Further studies are needed to elucidate the antiviral mechanism of these bacteriocins.


Subject(s)
Antiviral Agents , Bacteriocins , SARS-CoV-2 , Bacteriocins/pharmacology , Chlorocebus aethiops , Animals , Antiviral Agents/pharmacology , Vero Cells , Humans , SARS-CoV-2/drug effects , Virus Replication/drug effects , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Molecular Docking Simulation , Molecular Dynamics Simulation , Bridged-Ring Compounds
2.
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
3.
Viruses ; 16(5)2024 05 13.
Article in English | MEDLINE | ID: mdl-38793654

ABSTRACT

Based on several clinical observations it was hypothesized that herpesviruses may influence the replication of human bocaviruses, the second known parvoviruses that have been confirmed as human pathogens. While several cell lines support the growth of HSV-1, HBoV-1 was exclusively cultivated on air-liquid interface cultures, the latter being a rather complicated, slow, and low throughput system. One of the cell lines are T84 cells, which are derived from the lung metastasis of a colorectal tumor. In this study, we provide evidence that T84 also supports HBoV replication when cultivated as monolayers, while simultaneously being permissive for HSV-1. The cell culture model thus would enable co-infection studies of both viruses and is worth being optimized for high throughput studies with HBoV-1. Additionally, the study provides evidence for a supporting effect of HSV-1 on the replication and packaging of HBoV-1 progeny DNA into DNase-resistant viral particles.


Subject(s)
Coinfection , Herpesvirus 1, Human , Human bocavirus , Virus Replication , Herpesvirus 1, Human/physiology , Humans , Coinfection/virology , Human bocavirus/physiology , Human bocavirus/genetics , Cell Line , Cell Line, Tumor , Cell Culture Techniques/methods , Herpes Simplex/virology , Parvoviridae Infections/virology , Chlorocebus aethiops , Virus Cultivation/methods
4.
Biomolecules ; 14(5)2024 May 20.
Article in English | MEDLINE | ID: mdl-38786010

ABSTRACT

Cholesterol, a crucial component of cell membranes, influences various biological processes, including membrane trafficking, signal transduction, and host-pathogen interactions. Disruptions in cholesterol homeostasis have been linked to congenital and acquired conditions, including neurodegenerative disorders such as Alzheimer's disease (AD). Previous research from our group has demonstrated that herpes simplex virus type I (HSV-1) induces an AD-like phenotype in several cell models of infection. This study explores the interplay between cholesterol and HSV-1-induced neurodegeneration. The impact of cholesterol was determined by modulating its levels with methyl-beta-cyclodextrin (MßCD) using the neuroblastoma cell lines SK-N-MC and N2a. We have found that HSV-1 infection triggers the intracellular accumulation of cholesterol in structures resembling endolysosomal/autophagic compartments, a process reversible upon MßCD treatment. Moreover, MßCD exhibits inhibitory effects at various stages of HSV-1 infection, underscoring the importance of cellular cholesterol levels, not only in the viral entry process but also in subsequent post-entry stages. MßCD also alleviated several features of AD-like neurodegeneration induced by viral infection, including lysosomal impairment and intracellular accumulation of amyloid-beta peptide (Aß) and phosphorylated tau. In conclusion, these findings highlight the connection between cholesterol, neurodegeneration, and HSV-1 infection, providing valuable insights into the underlying mechanisms of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Cholesterol , Herpes Simplex , Herpesvirus 1, Human , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Cholesterol/metabolism , Humans , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/virology , Alzheimer Disease/pathology , Alzheimer Disease/drug therapy , Herpes Simplex/virology , Herpes Simplex/metabolism , Herpes Simplex/drug therapy , Herpes Simplex/pathology , Cell Line, Tumor , Animals , beta-Cyclodextrins/pharmacology , Lysosomes/metabolism , Lysosomes/drug effects , tau Proteins/metabolism , Phenotype , Mice
5.
Proc Natl Acad Sci U S A ; 121(19): e2401341121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38696466

ABSTRACT

Neurotropic alphaherpesviruses, including herpes simplex virus type 1 (HSV-1), recruit microtubule motor proteins to invade cells. The incoming viral particle traffics to nuclei in a two-step process. First, the particle uses the dynein-dynactin motor to sustain transport to the centrosome. In neurons, this step is responsible for long-distance retrograde axonal transport and is an important component of the neuroinvasive property shared by these viruses. Second, a kinesin-dependent mechanism redirects the particle from the centrosome to the nucleus. We have reported that the kinesin motor used during the second step of invasion is assimilated into nascent virions during the previous round of infection. Here, we report that the HSV-1 pUL37 tegument protein suppresses the assimilated kinesin-1 motor during retrograde axonal transport. Region 2 (R2) of pUL37 was required for suppression and functioned independently of the autoinhibitory mechanism native to kinesin-1. Furthermore, the motor domain and proximal coiled coil of kinesin-1 were sufficient for HSV-1 assimilation, pUL37 suppression, and nuclear trafficking. pUL37 localized to the centrosome, the site of assimilated kinesin-1 activation during infection, when expressed in cells in the absence of other viral proteins; however, pUL37 did not suppress kinesin-1 in this context. These results indicate that the pUL37 tegument protein spatially and temporally regulates kinesin-1 via the amino-terminal motor region in the context of the incoming viral particle.


Subject(s)
Herpesvirus 1, Human , Kinesins , Viral Structural Proteins , Kinesins/metabolism , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/metabolism , Humans , Animals , Axonal Transport/physiology , Chlorocebus aethiops , Centrosome/metabolism , Neurons/metabolism , Neurons/virology , Vero Cells , Cell Nucleus/metabolism , Cell Nucleus/virology
6.
Int J Mol Sci ; 25(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38732185

ABSTRACT

Herpes simplex virus (HSV) infections are highly widespread among humans, producing symptoms ranging from ulcerative lesions to severe diseases such as blindness and life-threatening encephalitis. At present, there are no vaccines available, and some existing antiviral treatments can be ineffective or lead to adverse effects. As a result, there is a need for new anti-HSV drugs. In this report, the in vitro anti-HSV effect of 9,9'-norharmane dimer (nHo-dimer), which belongs to the ß-carboline (ßC) alkaloid family, was evaluated. The dimer exhibited no virucidal properties and did not impede either the attachment or penetration steps of viral particles. The antiviral effect was only exerted under the constant presence of the dimer in the incubation media, and the mechanism of action was found to involve later events of virus infection. Analysis of fluorescence lifetime imaging data showed that the nHo-dimer internalized well into the cells when present in the extracellular incubation medium, with a preferential accumulation into perinuclear organelles including mitochondria. After washing the host cells with fresh medium free of nHo-dimer, the signal decreased, suggesting the partial release of the compound from the cells. This agrees with the observation that the antiviral effect is solely manifested when the alkaloid is consistently present in the incubation media.


Subject(s)
Antiviral Agents , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Chlorocebus aethiops , Humans , Vero Cells , Animals , Simplexvirus/drug effects , Simplexvirus/physiology , Herpes Simplex/drug therapy , Herpes Simplex/virology , Carbolines/pharmacology , Carbolines/chemistry , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Harmine/pharmacology , Harmine/chemistry , Harmine/analogs & derivatives
7.
Nat Commun ; 15(1): 4018, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740820

ABSTRACT

Anti-HSV therapies are only suppressive because they do not eliminate latent HSV present in ganglionic neurons, the source of recurrent disease. We have developed a potentially curative approach against HSV infection, based on gene editing using HSV-specific meganucleases delivered by adeno-associated virus (AAV) vectors. Gene editing performed with two anti-HSV-1 meganucleases delivered by a combination of AAV9, AAV-Dj/8, and AAV-Rh10 can eliminate 90% or more of latent HSV DNA in mouse models of orofacial infection, and up to 97% of latent HSV DNA in mouse models of genital infection. Using a pharmacological approach to reactivate latent HSV-1, we demonstrate that ganglionic viral load reduction leads to a significant decrease of viral shedding in treated female mice. While therapy is well tolerated, in some instances, we observe hepatotoxicity at high doses and subtle histological evidence of neuronal injury without observable neurological signs or deficits. Simplification of the regimen through use of a single serotype (AAV9) delivering single meganuclease targeting a duplicated region of the HSV genome, dose reduction, and use of a neuron-specific promoter each results in improved tolerability while retaining efficacy. These results reinforce the curative potential of gene editing for HSV disease.


Subject(s)
Dependovirus , Gene Editing , Herpes Simplex , Herpesvirus 1, Human , Viral Load , Virus Shedding , Animals , Gene Editing/methods , Female , Dependovirus/genetics , Mice , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Herpes Simplex/genetics , Herpes Simplex/virology , Herpes Simplex/therapy , Disease Models, Animal , Virus Latency/genetics , Humans , Genetic Vectors/genetics , Vero Cells , Genetic Therapy/methods , Herpes Genitalis/therapy , Herpes Genitalis/virology , DNA, Viral/genetics
8.
Virology ; 595: 110096, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38710129

ABSTRACT

Herpes stromal keratitis is the leading cause of infectious blindness in the western world. Infection by HSV1 is most common, but VZV and hCMV also infect the cornea. Multiple models of HSV1 corneal infection exist, but none for VZV and hCMV because of their host specificity. Here, we used commercially available 3D human corneal epithelial equivalents (HCEE) to study infection by these herpesviruses. HCEE was infected by HSV-1 and hCMV without requiring scarification and resulted in spreading infections. Spread of HSV-1 infection was rapid, while that of hCMV was slow. In contrast, infections with VZV required damage to the HCEE and did not spread. Acyclovir dramatically reduced replication of HSV-1 in this model. We conclude that highly quality-controlled, readily available HCEE is a useful model to study human-restricted herpesvirus infection of the human corneal epithelium and for screening of antiviral drugs for treating HSK in an 3D model system.


Subject(s)
Antiviral Agents , Epithelium, Corneal , Herpesvirus 1, Human , Keratitis, Herpetic , Humans , Keratitis, Herpetic/virology , Keratitis, Herpetic/drug therapy , Epithelium, Corneal/virology , Epithelium, Corneal/pathology , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Herpesvirus 3, Human/physiology , Herpesvirus 3, Human/drug effects , Cytomegalovirus/physiology , Cytomegalovirus/drug effects , Virus Replication , Acyclovir/pharmacology , Acyclovir/therapeutic use , Epithelial Cells/virology , Models, Biological
9.
Front Cell Infect Microbiol ; 14: 1383811, 2024.
Article in English | MEDLINE | ID: mdl-38808062

ABSTRACT

Introduction: While astrocytes participate in the CNS innate immunity against herpes simplex virus type 1 (HSV-1) infection, they are the major target for the virus. Therefore, it is of importance to understand the interplay between the astrocyte-mediated immunity and HSV-1 infection. Methods: Both primary human astrocytes and the astrocyte line (U373) were used in this study. RT-qPCR and Western blot assay were used to measure IFNs, the antiviral IFN-stimulated genes (ISGs), IFN regulatory factors (IRFs) and HSV-1 DNA. IRF1 knockout or knockdown was performed with CRISPR/Cas9 and siRNA transfection techniques. Results: Poly(dA:dT) could inhibit HSV-1 replication and induce IFN-ß/IFN-λs production in human astrocytes. Poly(dA:dT) treatment of astrocytes also induced the expression of the antiviral ISGs (Viperin, ISG56 and MxA). Among IRFs members examined, poly(dA:dT) selectively unregulated IRF1 and IRF9, particularly IRF1 in human astrocytes. The inductive effects of poly(dA:dT) on IFNs and ISGs were diminished in the IRF1 knockout cells. In addition, IRF1 knockout attenuated poly(dA:dT)-mediated HSV-1 inhibition in the cells. Conclusion: The DNA sensors activation induces astrocyte intracellular innate immunity against HSV-1. Therefore, targeting the DNA sensors has potential for immune activation-based HSV-1 therapy.


Subject(s)
Astrocytes , Herpesvirus 1, Human , Interferon Regulatory Factor-1 , Virus Replication , Humans , Astrocytes/virology , Astrocytes/metabolism , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Immunity, Innate , Poly dA-dT , Herpes Simplex/immunology , Herpes Simplex/virology , Cytosol/metabolism , Cell Line , Cells, Cultured , DNA, Viral/genetics , Gene Knockout Techniques
10.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731826

ABSTRACT

Although Herpes simplex virus type 1 (HSV-1) has been deeply studied, significant gaps remain in the fundamental understanding of HSV-host interactions: our work focused on studying the Infected Cell Protein 27 (ICP27) as an inhibitor of the Absent-in-melanoma-2 (AIM 2) inflammasome pathway, leading to reduced pro-inflammatory cytokines that influence the activation of a protective innate immune response to infection. To assess the inhibition of the inflammasome by the ICP27, hTert-immortalized Retinal Pigment Epithelial cells (hTert-RPE 1) infected with HSV-1 wild type were compared to HSV-1 lacking functional ICP27 (HSV-1∆ICP27) infected cells. The activation of the inflammasome by HSV-1∆ICP27 was demonstrated by quantifying the gene and protein expression of the inflammasome constituents using real-time PCR and Western blot. The detection of the cleavage of the pro-caspase-1 into the active form was performed by using a bioluminescent assay, while the quantification of interleukins 1ß (IL-1ß) and 18 (IL-18)released in the supernatant was quantified using an ELISA assay. The data showed that the presence of the ICP27 expressed by HSV-1 induces, in contrast to HSV-1∆ICP27 vector, a significant downregulation of AIM 2 inflammasome constituent proteins and, consequently, the release of pro-inflammatory interleukins into the extracellular environment reducing an effective response in counteracting infection.


Subject(s)
Cytokines , Herpesvirus 1, Human , Immediate-Early Proteins , Inflammasomes , Retinal Pigment Epithelium , Humans , Inflammasomes/metabolism , Herpesvirus 1, Human/physiology , Cytokines/metabolism , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Cell Line , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpes Simplex/virology , DNA-Binding Proteins
11.
Virol J ; 21(1): 118, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802860

ABSTRACT

Herpes simplex virus type 1 (HSV-1) infection of the eyes results in herpes simplex keratitis (HSK), which has led to vision loss and even blindness in patients. However, the rate of drug resistance in HSV is on the rise; therefore, new antiviral agents with sufficient safety profiles must be developed. At present, we assessed the anti-HSV-1 activity of 502 natural compounds and their ability to reduce the HSV-1-induced cytopathic effect. We chose harmol for further studies because it exhibited the highest antiviral activity. We found that harmol inhibited both HSV-1 F and HSV-1/153 (a clinical drug-resistant strain) replication, with an EC50 of 9.34 µM and 5.84 µM, respectively. Moreover, harmol reduced HSV-1 replication in corneal tissues and viral progeny production in tears, and also alleviated early corneal surface lesions related to HSK. For example, harmol treatment preserved corneal thickness and nerve density in HSK mice. Interestingly, harmol also showed a promising antiviral effect on HSV-1/153 induced HSK in mouse model. Furthermore, harmol combined with acyclovir (ACV) treatment showed a greater antiviral effect than either one alone in vitro. Therefore, harmol may be a promising therapeutic agent for managing HSK.


Subject(s)
Antiviral Agents , Disease Models, Animal , Herpesvirus 1, Human , Keratitis, Herpetic , Virus Replication , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Keratitis, Herpetic/drug therapy , Keratitis, Herpetic/virology , Mice , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Virus Replication/drug effects , Acyclovir/pharmacology , Acyclovir/therapeutic use , Cornea/virology , Cornea/drug effects , Cornea/pathology , Chlorocebus aethiops , Humans , Female , Vero Cells , Mice, Inbred BALB C
12.
Commun Biol ; 7(1): 494, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658802

ABSTRACT

Inflammatory monocytes (iMO) are recruited from the bone marrow to the brain during viral encephalitis. C-C motif chemokine receptor (CCR) 2 deficiency substantially reduces iMO recruitment for most, but not all encephalitic viruses. Here we show CCR7 acts synergistically with CCR2 to control this process. Following Herpes simplex virus type-1 (HSV-1), or La Crosse virus (LACV) infection, we find iMO proportions are reduced by approximately half in either Ccr2 or Ccr7 knockout mice compared to control mice. However, Ccr2/Ccr7 double knockouts eliminate iMO recruitment following infection with either virus, indicating these receptors together control iMO recruitment. We also find that LACV induces a more robust iMO recruitment than HSV-1. However, unlike iMOs in HSV-1 infection, LACV-recruited iMOs do not influence neurological disease development. LACV-induced iMOs have higher expression of proinflammatory and proapoptotic but reduced mitotic, phagocytic and phagolysosomal transcripts compared to HSV-1-induced iMOs. Thus, virus-specific activation of iMOs affects their recruitment, activation, and function.


Subject(s)
Brain , Herpesvirus 1, Human , La Crosse virus , Mice, Knockout , Monocytes , Receptors, CCR2 , Receptors, CCR7 , Animals , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Mice , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Brain/virology , Brain/metabolism , Brain/immunology , Herpesvirus 1, Human/physiology , La Crosse virus/genetics , La Crosse virus/physiology , Receptors, CCR7/metabolism , Receptors, CCR7/genetics , Encephalitis, California/virology , Encephalitis, California/genetics , Encephalitis, California/metabolism , Encephalitis, California/immunology , Mice, Inbred C57BL , Inflammation/metabolism , Inflammation/virology , Female , Male
13.
PLoS Pathog ; 20(4): e1011829, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38620036

ABSTRACT

Viruses target mitochondria to promote their replication, and infection-induced stress during the progression of infection leads to the regulation of antiviral defenses and mitochondrial metabolism which are opposed by counteracting viral factors. The precise structural and functional changes that underlie how mitochondria react to the infection remain largely unclear. Here we show extensive transcriptional remodeling of protein-encoding host genes involved in the respiratory chain, apoptosis, and structural organization of mitochondria as herpes simplex virus type 1 lytic infection proceeds from early to late stages of infection. High-resolution microscopy and interaction analyses unveiled infection-induced emergence of rough, thin, and elongated mitochondria relocalized to the perinuclear area, a significant increase in the number and clustering of endoplasmic reticulum-mitochondria contact sites, and thickening and shortening of mitochondrial cristae. Finally, metabolic analyses demonstrated that reactivation of ATP production is accompanied by increased mitochondrial Ca2+ content and proton leakage as the infection proceeds. Overall, the significant structural and functional changes in the mitochondria triggered by the viral invasion are tightly connected to the progression of the virus infection.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Mitochondria , Mitochondria/metabolism , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/metabolism , Humans , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpes Simplex/pathology , Animals , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Herpesviridae Infections/pathology , Disease Progression , Chlorocebus aethiops
14.
Cell Rep ; 43(4): 114095, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38613787

ABSTRACT

Interferon (IFN) contributes to the host's antiviral response by inducing IFN-stimulated genes (ISGs). However, their functional targets and the mechanism of action remain elusive. Here, we report that one such ISG, TRIM21, interacts with and degrades the TRPV2 channel in myeloid cells, reducing its expression and providing host protection against viral infections. Moreover, viral infection upregulates TRIM21 in paracrine and autocrine manners, downregulating TRPV2 in neighboring cells to prevent viral spread to uninfected cells. Consistently, the Trim21-/- mice are more susceptible to HSV-1 and VSV infection than the Trim21+/+ littermates, in which viral susceptibility is rescued by inhibition or deletion of TRPV2. Mechanistically, TRIM21 catalyzes the K48-linked ubiquitination of TRPV2 at Lys295. TRPV2K295R is resistant to viral-infection-induced TRIM21-dependent ubiquitination and degradation, promoting viral infection more profoundly than wild-type TRPV2 when reconstituted into Lyz2-Cre;Trpv2fl/fl myeloid cells. These findings characterize targeting the TRIM21-TRPV2 axis as a conducive strategy to control viral spread to bystander cells.


Subject(s)
Ribonucleoproteins , TRPV Cation Channels , Ubiquitination , Virus Diseases , Animals , Humans , Mice , Down-Regulation , HEK293 Cells , Herpesvirus 1, Human/physiology , Interferons/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Ribonucleoproteins/metabolism , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Virus Diseases/metabolism
15.
J Virol ; 98(5): e0003224, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38651900

ABSTRACT

Critical stages of lytic herpes simplex virus type 1 (HSV-1) replication are marked by the sequential expression of immediate early (IE) to early (E), then late (L) viral genes. HSV-1 can also persist in neuronal cells via a non-replicative, transcriptionally repressed infection called latency. The regulation of lytic and latent transcriptional profiles is critical to HSV-1 pathogenesis and persistence. We sought a fluorescence-based approach to observe the outcome of neuronal HSV-1 infection at the single-cell level. To achieve this goal, we constructed and characterized a novel HSV-1 recombinant that enables discrimination between lytic and latent infection. The dual reporter HSV-1 encodes a human cytomegalovirus-immediate early (hCMV-IE) promoter-driven enhanced yellow fluorescent protein (eYFP) to visualize the establishment of infection and an endogenous mCherry-VP26 fusion to report lytic replication. We confirmed that viral gene expression, replication, and spread of infection are not altered by the incorporation of the fluorescent reporters, and fluorescent protein (FP) detection virtuously reports the progression of lytic replication. We demonstrate that the outcome of HSV-1 infection of compartmentalized primary neurons is determined by viral inoculating dose: high-dose axonal inoculation proceeds to lytic replication, whereas low-dose axonal inoculation establishes a latent HSV-1 infection. Interfering with low-dose axonal inoculation via small molecule drugs reports divergent phenotypes of eYFP and mCherry reporter detection, correlating with altered states of viral gene expression. We report that the transcriptional state of neuronal HSV-1 infection is variable in response to changes in the intracellular neuronal environment.IMPORTANCEHerpes simplex virus type 1 (HSV-1) is a prevalent human pathogen that infects approximately 67% of the global human population. HSV-1 invades the peripheral nervous system, where latent HSV-1 infection persists within the host for life. Immunological evasion, viral persistence, and herpetic pathologies are determined by the regulation of HSV-1 gene expression. Studying HSV-1 gene expression during neuronal infection is challenging but essential for the development of antiviral therapeutics and interventions. We used a recombinant HSV-1 to evaluate viral gene expression during infection of primary neurons. Manipulation of cell signaling pathways impacts the establishment and transcriptional state of HSV-1 latency in neurons. The work here provides critical insight into the cellular and viral factors contributing to the establishment of latent HSV-1 infection.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Luminescent Proteins , Neurons , Virus Replication , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Neurons/virology , Neurons/metabolism , Humans , Animals , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Herpes Simplex/virology , Genes, Reporter , Virus Latency/genetics , Gene Expression Regulation, Viral , Chlorocebus aethiops , Vero Cells , Cytomegalovirus/genetics , Cytomegalovirus/physiology
16.
Virology ; 595: 110063, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38564935

ABSTRACT

This experimental study aimed to evaluate the antiviral and synergistic effects of photoenergy irradiation on human herpes simplex virus type I (HSV-1) infection. We assessed viral replication, plaque formation, and relevant viral gene expression to examine the antiviral and synergistic effects of blue light (BL) with acyclovir treatment. Our results showed that daily BL (10 J/cm2) irradiation inhibited plaque-forming ability and decreased viral copy numbers in HSV-1-infected monkey kidney epithelial Vero cells and primary human oral keratinocyte (HOK) cells. Combined treatment with the antiviral agent acyclovir and BL irradiation increased anti-viral activity, reducing viral titers and copy numbers. In particular, accumulated BL irradiation suppressed characteristic viral genes including UL19 and US6, and viral DNA replication-essential genes including UL9, UL30, UL42, and UL52 in HOK cells. Our results suggest that BL irradiation has anti-viral and synergistic properties, making it a promising therapeutic candidate for suppressing viral infections in clinical trials.


Subject(s)
Acyclovir , Antiviral Agents , Herpesvirus 1, Human , Virus Replication , Antiviral Agents/pharmacology , Animals , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/radiation effects , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/genetics , Chlorocebus aethiops , Vero Cells , Humans , Virus Replication/drug effects , Virus Replication/radiation effects , Acyclovir/pharmacology , Light , Herpes Simplex/virology , Herpes Simplex/drug therapy , Keratinocytes/virology , Keratinocytes/radiation effects , Keratinocytes/drug effects , Viral Plaque Assay
17.
Cell Rep ; 43(5): 114135, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38652662

ABSTRACT

Optimal activation of stimulator of interferon genes (STING) protein is crucial for host defenses against pathogens and avoiding detrimental effects. Various post-translational modifications control STING activity. However, the function of interferon (IFN)-stimulated gene (ISG) 15 modification (ISGylation) in controlling STING stability and activation is unclear. Here, we show that the E3 ISGylation ligases HECT domain- and RCC1-like domain-containing proteins (HERCs; HERC5 in humans and HERC6 in mice) facilitate STING activation by mediating ISGylation of STING at K150, preventing its K48-linked ubiquitination and degradation. Concordantly, Herc6 deficiency suppresses herpes simplex virus 1 infection-induced type I IFN responses and facilitates viral replication both in vitro and in vivo. Notably, severe acute respiratory syndrome coronavirus 2 protein papain-like protease cleaves HERC5-mediated ISGylation of STING, suppressing host antiviral responses. These data identify a mechanism by which HERCs-mediated ISGylation controls STING stability and activation and uncover the correlations and interactions of ISGylation and ubiquitination during STING activation.


Subject(s)
Membrane Proteins , Ubiquitin-Protein Ligases , Ubiquitination , Ubiquitins , Membrane Proteins/metabolism , Humans , Animals , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Mice , HEK293 Cells , Cytokines/metabolism , Herpesvirus 1, Human/physiology , SARS-CoV-2/metabolism , Mice, Inbred C57BL , Herpes Simplex/virology , Herpes Simplex/metabolism , Herpes Simplex/immunology , Virus Replication , Interferon Type I/metabolism , Intracellular Signaling Peptides and Proteins
18.
Cell Rep ; 43(5): 114122, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38652659

ABSTRACT

DNA sensing is important for antiviral immunity. The DNA sensor cGAS synthesizes 2'3'-cyclic GMP-AMP (cGAMP), a second messenger that activates STING, which induces innate immunity. cGAMP not only activates STING in the cell where it is produced but cGAMP also transfers to other cells. Transporters, channels, and pores (including SLC19A1, SLC46A2, P2X7, ABCC1, and volume-regulated anion channels (VRACs)) release cGAMP into the extracellular space and/or import cGAMP. We report that infection with multiple human viruses depletes some of these cGAMP conduits. This includes herpes simplex virus 1 (HSV-1) that targets SLC46A2, P2X7, and the VRAC subunits LRRC8A and LRRC8C for degradation. The HSV-1 protein UL56 is necessary and sufficient for these effects that are mediated at least partially by proteasomal turnover. UL56 thereby inhibits cGAMP uptake via VRAC, SLC46A2, and P2X7. Taken together, HSV-1 antagonizes intercellular cGAMP transfer. We propose that this limits innate immunity by reducing cell-to-cell communication via the immunotransmitter cGAMP.


Subject(s)
Herpesvirus 1, Human , Nucleotides, Cyclic , Herpesvirus 1, Human/physiology , Humans , Nucleotides, Cyclic/metabolism , Viral Proteins/metabolism , HEK293 Cells , Animals , Herpes Simplex/virology , Herpes Simplex/metabolism , Herpes Simplex/immunology
19.
Sci Rep ; 14(1): 6859, 2024 03 21.
Article in English | MEDLINE | ID: mdl-38514671

ABSTRACT

Reactivation of latent herpes simplex type 1 results in virus returning to the cornea leading to recurrent herpetic stromal keratitis (rHSK). We compare two competing models to reactivate viruses from latency, UV-B irradiation and cyclophosphamide (CP). Results revealed that while both result in corneal recrudescence, only UV-B irradiation results in rHSK. To better understand the dynamics of reactivation, we analyzed corneas for both the presence of infectious viruses and the dynamics of exposure to multiple reactivations using UV-B. We noted that multiple reactivations result in progressively worse corneal disease. We also noted that expression of IFNα and STING, surragate markers for the presence of virus, are induced by the presence of reactivated virus. Studies to determine the importance of STING to the development of HSK revealed that in the absence of STING, mice do not develop significant HSK and the magnitude of the infiltrate of CD45+ cells in these corneas is significantly reduced. The resulting paucity of CD45+CD11b+GR-1+F4/80-neutrophils, and to a lesser extent CD45+CD11b+GR-1-F4/80+ macrophages in B6-STING KO mice following reactivation is likely the underlying cause for lack of rHSK as has been noted by ourselves and others. These results underscore the critical importance of STING's role in developing rHSK.


Subject(s)
Corneal Diseases , Herpes Simplex , Herpesvirus 1, Human , Keratitis, Herpetic , Mice , Animals , Herpesvirus 1, Human/physiology , Cornea/metabolism , Corneal Diseases/etiology
20.
J Gen Virol ; 105(3)2024 03.
Article in English | MEDLINE | ID: mdl-38471041

ABSTRACT

Many viruses downregulate their cognate receptors, facilitating virus replication and pathogenesis via processes that are not yet fully understood. In the case of herpes simplex virus 1 (HSV1), the receptor binding protein glycoprotein D (gD) has been implicated in downregulation of its receptor nectin1, but current understanding of the process is limited. Some studies suggest that gD on the incoming virion is sufficient to achieve nectin1 downregulation, but the virus-encoded E3 ubiquitin ligase ICP0 has also been implicated. Here we have used the physiologically relevant nTERT human keratinocyte cell type - which we have previously shown to express readily detectable levels of endogenous nectin1 - to conduct a detailed investigation of nectin1 expression during HSV1 infection. In these cells, nectin1, but not nectin2 or the transferrin receptor, disappeared from the cell surface in a process that required virus protein synthesis rather than incoming virus, but did not involve virus-induced host shutoff. Furthermore, gD was not only required but was sufficient for nectin1 depletion, indicating that no other virus proteins are essential. NK cells were shown to be activated in the presence of keratinocytes, a process that was greatly inhibited in cells infected with wild-type virus. However, degranulation of NK cells was also inhibited in ΔgD-infected cells, indicating that blocking of NK cell activation was independent of gD downregulation of nectin1. By contrast, a superinfection time-course revealed that the ability of HSV1 infection to block subsequent infection of a GFP-expressing HSV1 was dependent on gD and occurred in line with the timing of nectin1 downregulation. Thus, the role of gD-dependent nectin1 impairment during HSV infection is important for virus infection, but not immune evasion, which is achieved by other mechanisms.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Superinfection , Humans , Cell Adhesion Molecules/metabolism , Cell Line , Down-Regulation , Herpesvirus 1, Human/physiology , Keratinocytes , Receptors, Virus/metabolism , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...