Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.461
Filter
1.
Methods Mol Biol ; 2854: 41-50, 2025.
Article in English | MEDLINE | ID: mdl-39192117

ABSTRACT

The innate immune system relies on a variety of pathogen recognition receptors (PRRs) as the first line of defense against pathogenic invasions. Viruses have evolved multiple strategies to evade the host immune system through coevolution with hosts. The CRISPR-Cas system is an adaptive immune system in bacteria or archaea that defends against viral reinvasion by targeting nucleic acids for cleavage. Based on the characteristics of Cas proteins and their variants, the CRISPR-Cas system has been developed into a versatile gene-editing tool capable of gene knockout or knock-in operations to achieve genetic variations in organisms. It is now widely used in the study of viral immune evasion mechanisms. This chapter will introduce the use of the CRISPR-Cas9 system for editing herpes simplex virus 1 (HSV-1) genes to explore the mechanisms by which HSV-1 evades host innate immunity and the experimental procedures involved.


Subject(s)
CRISPR-Cas Systems , Gene Knockout Techniques , Herpesvirus 1, Human , Immune Evasion , Immunity, Innate , CRISPR-Cas Systems/genetics , Immunity, Innate/genetics , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/genetics , Immune Evasion/genetics , Humans , Gene Editing/methods , Animals , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Herpes Simplex/immunology , Herpes Simplex/virology , Herpes Simplex/genetics
2.
Virol J ; 21(1): 219, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285274

ABSTRACT

BACKGROUND: To investigate the mechanism of Golgi matrix protein 130(GM130) regulating the antiviral immune response of TLR3 after herpes simplex virus type 1(HSV-1) infection of microglia cells. We explored the regulatory effects of berberine on the immune response mediated by GM130 and TLR3. METHODS: An in vitro model of HSV-1 infection was established by infecting BV2 cells with HSV-1. RESULTS: Compared to the uninfected group, the Golgi apparatus (GA) fragmentation and GM130 decreased after HSV-1 infection; TLR3 increased at 6 h and began to decrease at 12 h after HSV-1 infection; the secretion of interferon-beta(IFN-ß), tumour necrosis factor alpha(TNF-α), and interleukin-6(IL-6) increased after infection. Knockdown of GM130 aggravated fragmentation of the GA and caused TLR3 to further decrease, and the virus titer also increased significantly. GM130 knockdown inhibits the increase in TLR3 and inflammatory factors induced by TLR3 agonists and increases the viral titer. Overexpression of GM130 alleviated fragmentation of the GA induced by HSV-1, partially restored the levels of TLR3, and reduced viral titers. GM130 overexpression reversed the reduction in TLR3 and inflammatory cytokine levels induced by TLR3 inhibitors. Therefore, the decrease in GM130 levels caused by HSV-1 infection leads to increased viral replication by inhibiting TLR3-mediated innate immunity. Berberine can protect the GA and reverse the downregulation of GM130, as well as the downregulation of TLR3 and its downstream factors after HSV-1 infection, reducing the virus titer. CONCLUSIONS: In microglia, one mechanism of HSV-1 immune escape is disruption of the GM130/TLR3 pathway. Berberine protects the GA and enhances TLR3-mediated antiviral immune responses.


Subject(s)
Down-Regulation , Herpesvirus 1, Human , Immunity, Innate , Microglia , Toll-Like Receptor 3 , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/genetics , Microglia/virology , Microglia/immunology , Microglia/drug effects , Animals , Mice , Cell Line , Immune Evasion , Berberine/pharmacology , Cytokines/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Herpes Simplex/immunology , Herpes Simplex/virology
4.
J Med Virol ; 96(9): e29865, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39233492

ABSTRACT

Eczema herpeticum (EH) is a disseminated severe herpes simplex virus type 1 (HSV-1) infection that mainly occurs in a subset of patients suffering from atopic dermatitis (AD). EH is complex and multifaceted, involving immunological changes, environmental influences, and genetic aberrations. Certain genetic variants of the thymic stromal lymphopoietin (TSLP) may predispose to develop severe HSV-1-induced eczema. Therefore, we investigated the impact of TSLP on HSV-1 infection. TSLP encodes for two distinct forms: a long-form (lfTSLP), primarily associated with type 2 immunity, and a short-form (sfTSLP) with anti-inflammatory and antimicrobial properties. While sfTSLP reduced HSV-1 infectibility in human primary keratinocytes (HPK), lfTSLP did not. In HPK treated with sfTSLP, HSV-1 gene expression, and replication decreased, while virion binding to cells and targeting of incoming capsids to the nucleus were not diminished compared to untreated cells. sfTSLP caused only minor changes in the expression of innate immunity cytokines, and its inhibition of HSV-1 infection did not require de novo protein synthesis. Time window experiments indicated a different antiviral mechanism than LL-37. sfTSLP showed the strongest antiviral effect when administered to HPK before or after inoculation with HSV-1, and outperformed the inhibitory potential of LL-37 under these conditions. Our data show that sfTSLP has antiviral functions and promotes repression of the HSV-1 infection in HPK.


Subject(s)
Cytokines , Herpesvirus 1, Human , Keratinocytes , Thymic Stromal Lymphopoietin , Humans , Cytokines/metabolism , Keratinocytes/virology , Keratinocytes/immunology , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/genetics , Cells, Cultured , Virus Replication , Kaposi Varicelliform Eruption/virology , Kaposi Varicelliform Eruption/immunology , Herpes Simplex/virology , Herpes Simplex/immunology , Herpes Simplex/genetics , Immunity, Innate
5.
Cells ; 13(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39120287

ABSTRACT

Autophagy engulfs cellular components in double-membrane-bound autophagosomes for clearance and recycling after fusion with lysosomes. Thus, autophagy is a key process for maintaining proteostasis and a powerful cell-intrinsic host defense mechanism, protecting cells against pathogens by targeting them through a specific form of selective autophagy known as xenophagy. In this context, ubiquitination acts as a signal of recognition of the cargoes for autophagic receptors, which direct them towards autophagosomes for subsequent breakdown. Nevertheless, autophagy can carry out a dual role since numerous viruses including members of the Orthoherpesviridae family can either inhibit or exploit autophagy for its own benefit and to replicate within host cells. There is growing evidence that Herpes simplex virus type 1 (HSV-1), a highly prevalent human pathogen that infects epidermal keratinocytes and sensitive neurons, is capable of negatively modulating autophagy. Since the effects of HSV-1 infection on autophagic receptors have been poorly explored, this study aims to understand the consequences of HSV-1 productive infection on the levels of the major autophagic receptors involved in xenophagy, key proteins in the recruitment of intracellular pathogens into autophagosomes. We found that productive HSV-1 infection in human neuroglioma cells and keratinocytes causes a reduction in the total levels of Ub conjugates and decreases protein levels of autophagic receptors, including SQSTM1/p62, OPTN1, NBR1, and NDP52, a phenotype that is also accompanied by reduced levels of LC3-I and LC3-II, which interact directly with autophagic receptors. Mechanistically, we show these phenotypes are the result of xenophagy activation in the early stages of productive HSV-1 infection to limit virus replication, thereby reducing progeny HSV-1 yield. Additionally, we found that the removal of the tegument HSV-1 protein US11, a recognized viral factor that counteracts autophagy in host cells, enhances the clearance of autophagic receptors, with a significant reduction in the progeny HSV-1 yield. Moreover, the removal of US11 increases the ubiquitination of SQSTM1/p62, indicating that US11 slows down the autophagy turnover of autophagy receptors. Overall, our findings suggest that xenophagy is a potent host defense against HSV-1 replication and reveals the role of the autophagic receptors in the delivery of HSV-1 to clearance via xenophagy.


Subject(s)
Autophagy , Herpesvirus 1, Human , Humans , Herpesvirus 1, Human/physiology , Herpes Simplex/virology , Herpes Simplex/immunology , Herpes Simplex/metabolism , Macroautophagy , Virus Replication , Autophagosomes/metabolism , Keratinocytes/virology , Keratinocytes/metabolism , Sequestosome-1 Protein/metabolism , Host-Pathogen Interactions , Animals , Nuclear Proteins , Cell Cycle Proteins , Membrane Transport Proteins
6.
Rev Med Virol ; 34(5): e2574, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39090526

ABSTRACT

Herpes simplex virus (HSV) infections in allogeneic haematopoietic stem cell transplantation (HSCT) recipients pose significant challenges, with higher incidence, severity, and risk of emergence of resistance to antivirals due to impaired T-cell mediated immunity. This literature review focuses on acyclovir-refractory/resistant HSV infections in HSCT recipients. The review addresses the efficacy of antiviral prophylaxis, the incidence of acyclovir-refractory/resistant HSV infections, and the identification of risk factors and potential prognostic impact associated with those infections. Additionally, alternative therapeutic options are discussed. While acyclovir prophylaxis demonstrates a significant benefit in reducing HSV infections in HSCT recipients and, in some cases, overall mortality, concerns arise about the emergence of drug-resistant HSV strains. Our systematic review reports a median incidence of acyclovir-resistant HSV infections of 16.1%, with an increasing trend in recent years. Despite limitations in available studies, potential risk factors of emergence of HSV resistance to acyclovir include human leucocyte antigen (HLA) mismatches, myeloid neoplasms and acute leukaemias, and graft-versus-host disease (GVHD). Limited evidences suggest a potentially poorer prognosis for allogeneic HSCT recipients with acyclovir-refractory/resistant HSV infection. Alternative therapeutic approaches, such as foscarnet, cidofovir, topical cidofovir, optimised acyclovir dosing, and helicase-primase inhibitors offer promising options but require further investigations. Overall, larger studies are needed to refine preventive and therapeutic strategies for acyclovir-refractory/resistant HSV infections in allogeneic HSCT recipients and to identify those at higher risk.


Subject(s)
Acyclovir , Antiviral Agents , Drug Resistance, Viral , Hematopoietic Stem Cell Transplantation , Herpes Simplex , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Herpes Simplex/drug therapy , Herpes Simplex/virology , Herpes Simplex/therapy , Antiviral Agents/therapeutic use , Acyclovir/therapeutic use , Simplexvirus/drug effects , Simplexvirus/physiology , Risk Factors , Transplant Recipients , Incidence
7.
J Clin Microbiol ; 62(9): e0013124, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39133014

ABSTRACT

Bloodstream HSV-1 and HSV-2 infections can cause devastating outcomes with high morbidity and mortality, especially in neonates or immunocompromised individuals. Proper patient management for herpes simplex virus (HSV) bloodstream infections is time-sensitive and requires a rapid, accurate, and definitive diagnosis. The absence of the U.S. Food and Drug Administration (FDA)-approved molecular assays for HSV detection in blood, coupled with a lack of consensus on the optimal sample type, underscores the unmet need for improved diagnostics. We prospectively compared the cycle threshold values in paired samples including whole blood (WB), plasma, serum, and peripheral blood mononuclear cells (PBMCs) from patients with bloodstream HSV infections. This analysis employed a modified use of the FDA-cleared Simplexa HSV-1 & 2 Direct assay. The clinical performance in serum was assessed by comparing the results of 247 remnant specimens on this sample-to-answer platform to established laboratory-developed tests in a blinded fashion. Serum samples exhibited significantly lower cycle thresholds than whole blood samples [2.6 cycle threshold (Ct) bias, P < 0.001]. The modified Simplexa assay demonstrated 100% positive percent agreement for the detection of HSV-1 and HSV-2 DNA in serum samples and yielded an overall agreement of 95% (95% CI, 0.92 to 0.97), with a κ statistic of 0.75 (95% CI, 0.62 to 0.86) compared to the composite reference method. Discordance rates were 5.20% for HSV-1 and 0.81% for HSV-2. This investigation demonstrates that serum is an optimal specimen type for HSV detection when compared to several blood compartments. Serum offers a promising sample type for rapid and accurate diagnosis of HSV bloodstream infections using the modified Simplexa assay. IMPORTANCE: Rapid, accurate, and definitive diagnosis of herpes simplex virus (HSV) infections is crucial in clinical settings for patient management. The absence of FDA-authorized molecular assays for HSV-1/2 detection in blood, coupled with a lack of consensus on the optimal sample type, underscores the need for improved diagnostic methods. Furthermore, rapid diagnosis of HSV bloodstream infections enables timely administration of antiviral treatment, influences patient management decisions for those at high risk, and can contribute to shorter hospital stays, thereby reducing healthcare costs.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Herpesvirus 2, Human , Molecular Diagnostic Techniques , Sensitivity and Specificity , Humans , Herpesvirus 1, Human/isolation & purification , Herpesvirus 1, Human/genetics , Herpes Simplex/diagnosis , Herpes Simplex/virology , Herpesvirus 2, Human/isolation & purification , Herpesvirus 2, Human/genetics , Male , Female , Prospective Studies , Middle Aged , Adult , Molecular Diagnostic Techniques/methods , Molecular Diagnostic Techniques/standards , Young Adult , Aged , Adolescent , Child , Time Factors , Child, Preschool , Infant , Aged, 80 and over
8.
J Virol ; 98(9): e0065624, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39136460

ABSTRACT

The multifunctional tegument protein pUL21 of HSV-2 is phosphorylated in infected cells. We have identified two residues in the unstructured linker region of pUL21, serine 251 and serine 253, as phosphorylation sites. Both phosphorylation sites are absent in HSV-1 pUL21, which likely explains why phosphorylated pUL21 was not detected in cells infected with HSV-1. Cells infected with HSV-2 strain 186 viruses deficient in pUL21 phosphorylation exhibited reductions in both cell-cell spread of virus infection and virus replication. Defects in secondary envelopment of cytoplasmic nucleocapsids were also observed in cells infected with viruses deficient in pUL21 phosphorylation as well as in cells infected with multiple strains of HSV-2 and HSV-1 deleted for pUL21. These results confirm a role for HSV pUL21 in the secondary envelopment of cytoplasmic nucleocapsids and indicate that phosphorylation of HSV-2 pUL21 is required for this activity. Phosphorylation of pUL21 was substantially reduced in cells infected with HSV-2 strain 186 mutants lacking the viral serine/threonine kinase pUL13, indicating a requirement for pUL13 in pUL21 phosphorylation. IMPORTANCE: It is well known that post-translational modification of proteins by phosphorylation can regulate protein function. Here, we determined that phosphorylation of the multifunctional HSV-2 tegument protein pUL21 requires the viral serine/threonine kinase pUL13. In addition, we identified serine residues within HSV-2 pUL21 that can be phosphorylated. Phenotypic analysis of mutant HSV-2 strains with deficiencies in pUL21 phosphorylation revealed reductions in both cell-cell spread of virus infection and virus replication. Deficiencies in pUL21 phosphorylation also compromised the secondary envelopment of cytoplasmic nucleocapsids, a critical final step in the maturation of all herpes virions. Unlike HSV-2 pUL21, phosphorylation of HSV-1 pUL21 was not detected. This fundamental difference between HSV-2 and HSV-1 may underlie our previous observations that the requirements for pUL21 differ between HSV species.


Subject(s)
Herpesvirus 2, Human , Nucleocapsid , Virus Replication , Herpesvirus 2, Human/metabolism , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/physiology , Phosphorylation , Animals , Chlorocebus aethiops , Humans , Vero Cells , Nucleocapsid/metabolism , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Cytoplasm/metabolism , Cytoplasm/virology , Cell Line , Viral Structural Proteins/metabolism , Viral Structural Proteins/genetics , Virus Assembly , Herpes Simplex/virology , Herpes Simplex/metabolism
9.
J Virol ; 98(9): e0059924, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39136459

ABSTRACT

Herpes simplex virus 1 (HSV-1) is an alpha herpesvirus that infects a majority of the world population. The mechanisms and cellular host factors involved in the intracellular transport and exocytosis of HSV-1 particles are not fully understood. To elucidate these late steps in the replication cycle, we developed a live-cell fluorescence microscopy assay of HSV-1 virion intracellular trafficking and exocytosis. This method allows us to track individual virus particles and identify the precise moment and location of particle exocytosis using a pH-sensitive reporter. We show that HSV-1 uses the host cell's post-Golgi secretory pathway during egress. The small GTPase, Rab6, binds to nascent secretory vesicles at the trans-Golgi network and plays important, but non-essential, roles in vesicle traffic and exocytosis at the plasma membrane, therefore making it a useful marker of the Golgi and post-Golgi secretory pathway. We show that HSV-1 particles colocalize with Rab6a in the region of the Golgi, cotraffic with Rab6a to the cell periphery, and undergo exocytosis from Rab6a vesicles. Consistent with previous reports, we find that HSV-1 particles accumulate at preferential egress sites in infected cells. The secretory pathway mediates this preferential/polarized egress, since Rab6a vesicles accumulate near the plasma membrane similarly in uninfected cells. These data suggest that, following particle envelopment, HSV-1 egress follows a pre-existing cellular secretory pathway to exit infected cells rather than novel, virus-induced mechanisms. IMPORTANCE: Herpes simplex virus 1 (HSV-1) infects a majority of people. It establishes a life-long latent infection and occasionally reactivates, typically causing characteristic oral or genital lesions. Rarely in healthy natural hosts, but more commonly in zoonotic infections and in elderly, newborn, or immunocompromised patients, HSV-1 can cause severe herpes encephalitis. The precise cellular mechanisms used by HSV-1 remain an important area of research. In particular, the egress pathways that newly assembled virus particles use to exit from infected cells are unclear. In this study, we used fluorescence microscopy to visualize individual virus particles exiting from cells and found that HSV-1 particles use the pre-existing cellular secretory pathway.


Subject(s)
Exocytosis , Golgi Apparatus , Herpesvirus 1, Human , Secretory Pathway , Virus Release , rab GTP-Binding Proteins , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/metabolism , rab GTP-Binding Proteins/metabolism , Humans , Animals , Golgi Apparatus/metabolism , Golgi Apparatus/virology , Vero Cells , trans-Golgi Network/metabolism , trans-Golgi Network/virology , Chlorocebus aethiops , Herpes Simplex/virology , Herpes Simplex/metabolism , Virion/metabolism , HeLa Cells , Cell Membrane/metabolism , Cell Membrane/virology
10.
Antiviral Res ; 230: 105989, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39154753

ABSTRACT

Recently it was discovered that extracellular 2'-3'cGAMP can activate the STING pathway in a cGAS-independent fashion by being transported across the cell membrane via the folate transporter, SLC19A1, the first identified extracellular antiporter of this critical signaling molecule in cancer cells. We hypothesized that this non-canonical activation of STING pathway would function to establish an antiviral state similar to that seen with the paracrine antiviral activities of interferon. Herein, we report that treatment of the monocytic cell line, THP-1 cells and SH-SY5Y neuronal cell line with exogenous 2'-3'cGAMP induces interferon production and establishes an antiviral state that limits herpes simplex virus-1 (HSV-1), a ubiquitous virus with high seropositivity in the human population. Using either pharmaceutical inhibition or genetic knockout of SLC19A1 blocks the 2'-3'cGAMP-induced inhibition of viral replication. Our data indicate SLC19A1 functions as a newly identified antiviral mediator for extracellular 2'-3'cGAMP. This work presents novel and important findings about an antiviral mechanism which information could aid in the development of better antiviral drugs in the future.


Subject(s)
Herpesvirus 1, Human , Reduced Folate Carrier Protein , Virus Replication , Humans , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/drug effects , Virus Replication/drug effects , Reduced Folate Carrier Protein/metabolism , Reduced Folate Carrier Protein/genetics , Antiviral Agents/pharmacology , Antiviral Agents/metabolism , Cell Line , Herpes Simplex/virology , Herpes Simplex/metabolism , THP-1 Cells , Biological Transport , Membrane Proteins/metabolism , Membrane Proteins/genetics , Interferons/metabolism , Signal Transduction
11.
Microb Pathog ; 195: 106846, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39128643

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is responsible for a wide range of human infections, including skin and mucosal ulcers, encephalitis, and keratitis. The gold standard for treating HSV-1 infections is acyclovir. However, the use of this drug is associated with several limitations such as toxic reactions and the development of drug-resistant strains. So, there is an urgent need to discover and develop novel and effective agents against this virus. For the first time, this study aimed to investigate the antiviral effects of the Thermally Expanded Graphite (TEG)-copper oxide (CuO) nanocomposite against HSV-1 and compare results with its constituent components. After microwave (MW)-assisted synthesis of TEG and CuO nanosheets as well as MW-CuO/TEG nanocomposite and characterization of all these nanomaterials, an MTT assay was used to determine their cytotoxicity. The quantitative real-time PCR was then used to investigate the effects of these nanomaterials on viral load. Three-hour incubation of HSV-1 with TEG nanosheets (500 µg/mL), MW-CuO nanosheets (15 µg/mL), and MW-CuO/TEG nanocomposite (35 µg/mL) resulted in a decrease in viral load with an inhibition rate of 31.4 %, 49.2 %, and 74.4 %, respectively. The results from the post-treatment assay also showed that TEG nanosheets (600 µg/mL), MW-CuO nanosheets (15 µg/mL), and MW-CuO/TEG nanocomposite (10 µg/mL) led to a remarkable decrease in viral load with an inhibition rate of 56.9 %, 63 %, and 99.9 %, respectively. The combination of TEG and MW-CuO nanosheets together and the formation of a nanocomposite structure display strong synergy in their ability to inhibit HSV-1 infection. MW-CuO/TEG nanocomposites can be considered a suitable candidate for the treatment of HSV-1 infection.


Subject(s)
Antiviral Agents , Copper , Graphite , Herpesvirus 1, Human , Nanocomposites , Copper/pharmacology , Copper/chemistry , Herpesvirus 1, Human/drug effects , Graphite/chemistry , Graphite/pharmacology , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Nanocomposites/chemistry , Vero Cells , Chlorocebus aethiops , Animals , Viral Load/drug effects , Microwaves , Drug Synergism , Cell Survival/drug effects , Humans , Herpes Simplex/drug therapy , Herpes Simplex/virology
12.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39126036

ABSTRACT

The periodontal ligament (PDL) is a complex connective tissue that connects the tooth root to the dental alveolar bone and plays crucial mechanical roles. PDL also exhibits regenerative roles and regulatory functions to maintain periodontium integrity and homeostasis. While PDL exposure to oral microbial pathogens is common, virtually nothing is known regarding viral infections of PDL. In particular, human herpes simplex virus type 1 (HSV-1) persistently infects the oral cavity through infections of the oral epithelium, connective tissue and neurons. While the oral spread of HSV-1 is generally asymptomatic, this virus has also been implicated in various oral pathologies. In this study, using a primary cell model derived from PDL (PDL cells), and whole surgical fragments of PDL, we provide evidence supporting the efficient infection of PDL by HSV-1 and the promotion of cytopathic effects. Infection of PDL by HSV-1 was also associated with an acute innate inflammatory response, as illustrated by the production of antiviral interferons and pro-inflammatory cytokines. Furthermore, this inflammatory response to HSV-1 was exacerbated in the presence of bacterial-derived products, such as peptidoglycans. This work therefore highlights the ability of HSV-1 to infect mesenchymal cells from PDL, suggesting that PDL may serve as a viral reservoir for the periodontal spread of HSV-1. Moreover, this raises questions about HSV-1 oral pathogenesis, as HSV-1-associated cytopathic and inflammatory effects may contribute to profound alterations of PDL integrity and functioning.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Periodontal Ligament , Humans , Periodontal Ligament/virology , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/pathogenicity , Herpes Simplex/virology , Cytokines/metabolism , Cells, Cultured
13.
Biosensors (Basel) ; 14(8)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39194630

ABSTRACT

Static well plates remain the gold standard to study viral infections in vitro, but they cannot accurately mimic dynamic viral infections as they occur in the human body. Therefore, we established a dynamic cell culture platform, based on centrifugal microfluidics, to study viral infections in perfusion. To do so, we used human primary periodontal dental ligament (PDL) cells and herpes simplex virus-1 (HSV-1) as a case study. By microscopy, we confirmed that the PDL cells efficiently attached and grew in the chip. Successful dynamic viral infection of perfused PDL cells was monitored using fluorescent imaging and RT-qPCR-based experiments. Remarkably, viral infection in flow resulted in a gradient of HSV-1-infected cells gradually decreasing from the cell culture chamber entrance towards its end. The perfusion of acyclovir in the chip prevented HSV-1 spreading, demonstrating the usefulness of such a platform for monitoring the effects of antiviral drugs. In addition, the innate antiviral response of PDL cells, measured by interferon gene expression, increased significantly over time in conventional static conditions compared to the perfusion model. These results provide evidence suggesting that dynamic viral infections differ from conventional static infections, which highlights the need for more physiologically relevant in vitro models to study viral infections.


Subject(s)
Herpesvirus 1, Human , Periodontal Ligament , Humans , Periodontal Ligament/cytology , Periodontal Ligament/virology , Cell Culture Techniques , Microfluidics , Herpes Simplex/virology , Cells, Cultured , Antiviral Agents/pharmacology
14.
Mol Biomed ; 5(1): 35, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39207577

ABSTRACT

Herpes simplex virus (HSV), an epidemic human pathogen threatening global public health, gains notoriety for its complex pathogenesis that encompasses lytic infection of mucosal cells, latent infection within neurons, and periodic reactivation. This intricate interplay, coupled with HSV's sophisticated immune evasion strategies, gives rise to various diseases, including genital lesions, neonatal encephalitis, and cancer. Despite more than 70 years of relentless research, an effective preventive or therapeutic vaccine against HSV has yet to emerge, primarily due to the limited understanding of virus-host interactions, which in turn impedes the identification of effective vaccine targets. However, HSV's unique pathological features, including its substantial genetic load capacity, high replicability, transmissibility, and neurotropism, render it a promising candidate for various applications, spanning oncolytic virotherapy, gene and immune therapies, and even as an imaging tracer in neuroscience. In this review, we comprehensively update recent breakthroughs in HSV pathogenesis and immune evasion, critically summarize the progress made in vaccine candidate development, and discuss the multifaceted applications of HSV as a biological tool. Importantly, we highlight both success and challenges, emphasizing the critical need for intensified research into HSV, with the aim of providing deeper insights that can not only advance HSV treatment strategies but also broaden its application horizons.


Subject(s)
Herpes Simplex , Vaccine Development , Humans , Herpes Simplex/immunology , Herpes Simplex/prevention & control , Herpes Simplex/virology , Animals , Simplexvirus/pathogenicity , Simplexvirus/immunology , Simplexvirus/physiology , Herpes Simplex Virus Vaccines/immunology , Immune Evasion
15.
Viruses ; 16(8)2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39205282

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-dependent Cl- channel, is closely associated with multiple pathogen infections, such as SARS-CoV-2. However, whether the function of the CFTR is involved in herpes simplex virus (HSV) infection has not been reported. To evaluate the association of CFTR activity with HSV infection, the antiviral effect of CFTR inhibitors in epithelial cells and HSV-infected mice was tested in this study. The data showed that treatment with CFTR inhibitors in different concentrations, Glyh-101 (5-20 µM), CFTRi-172 (5-20 µM) and IOWH-032 (5-20 µM), or the gene silence of the CFTR could suppress herpes simplex virus 1 (HSV-1) and herpes simplex virus 2 (HSV-2) replication in human HaCaT keratinocytes cells, and that a CFTR inhibitor, Glyh-101 (10-20 µM), protected mice from HSV-1 and HSV-2 infection. Intracellular Cl- concentration ([Cl-]i) was decreased after HSV infection via the activation of adenylyl cyclase (AC)-cAMP signaling pathways. CFTR inhibitors (20 µM) increased the reduced [Cl-]i caused by HSV infection in host epithelial cells. Additionally, CFTR inhibitors reduced the activity and phosphorylation of SGK1 in infected cells and tissues (from the eye and vagina). Our study found that CFTR inhibitors can effectively suppress HSV-1 and HSV-2 infection, revealing a previously unknown role of CFTR inhibitors in HSV infection and suggesting new perspectives on the mechanisms governing HSV infection in host epithelial cells, as well as leading to potential novel treatments.


Subject(s)
Antiviral Agents , Cystic Fibrosis Transmembrane Conductance Regulator , Herpes Simplex , Herpesvirus 1, Human , Herpesvirus 2, Human , Virus Replication , Animals , Mice , Antiviral Agents/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Humans , Herpes Simplex/drug therapy , Herpes Simplex/virology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Virus Replication/drug effects , Herpesvirus 2, Human/drug effects , Herpesvirus 2, Human/physiology , Female , Cell Line , Epithelial Cells/virology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , HaCaT Cells , Keratinocytes/virology , Keratinocytes/drug effects , Mice, Inbred BALB C , Chlorocebus aethiops , Simplexvirus/drug effects , Simplexvirus/physiology
16.
Int J Mol Sci ; 25(16)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39201398

ABSTRACT

Protein expression is regulated through multiple mechanisms, including post-translational modifications (PTMs), which can alter protein structure, stability, localization, and function. Among these, citrullination stands out due to its ability to convert arginine residues into citrulline, altering protein charge and mass. This modification is catalyzed by calcium-dependent protein arginine deiminases (PADs), enzymes implicated in various inflammatory diseases. We have recently shown that human cytomegalovirus (HCMV) and herpes simplex virus type 1 (HSV-1) exploit these enzymes to enhance their replication capabilities. Although the role of PADs in HCMV and HSV-1 infections is well documented, their involvement in HSV-2 infection has not yet been thoroughly investigated. Here, we demonstrate that HSV-2 manipulates the overall protein citrullination profile by activating three PAD isoforms: PAD2, PAD3, and PAD4. However, as previously observed during HSV-1 infection, PAD3 is the most significantly upregulated isoform, both at the mRNA and protein levels. Consistently, we demonstrate that inhibiting PAD3, either through the specific inhibitor CAY10727 or via CRISPR/Cas9-mediated gene silencing, markedly reduces HSV-2 replication and viral protein expression. Lastly, we show that CAY10727 displays an IC50 value of 0.3 µM, which is extremely close to what was previously observed for HSV-1. Overall, our findings highlight the crucial role of PAD3 in the life cycle of HSV-2 and suggest that the targeted inhibition of PAD3 may represent a promising approach for treating HSV-2 infections, especially in cases resistant to existing antiviral therapies.


Subject(s)
Herpesvirus 2, Human , Protein-Arginine Deiminase Type 3 , Humans , Herpesvirus 2, Human/physiology , Herpesvirus 2, Human/genetics , Protein-Arginine Deiminase Type 3/metabolism , Citrullination , Herpes Simplex/virology , Herpes Simplex/metabolism , Virus Replication/drug effects , Animals , Herpes Genitalis/metabolism , Herpes Genitalis/virology , Herpes Genitalis/drug therapy , Protein-Arginine Deiminase Type 2/metabolism , Antiviral Agents/pharmacology
17.
Nature ; 632(8024): 383-389, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048823

ABSTRACT

The brain is highly sensitive to damage caused by infection and inflammation1,2. Herpes simplex virus 1 (HSV-1) is a neurotropic virus and the cause of herpes simplex encephalitis3. It is unknown whether neuron-specific antiviral factors control virus replication to prevent infection and excessive inflammatory responses, hence protecting the brain. Here we identify TMEFF1 as an HSV-1 restriction factor using genome-wide CRISPR screening. TMEFF1 is expressed specifically in neurons of the central nervous system and is not regulated by type I interferon, the best-known innate antiviral system controlling virus infections. Depletion of TMEFF1 in stem-cell-derived human neurons led to elevated viral replication and neuronal death following HSV-1 infection. TMEFF1 blocked the HSV-1 replication cycle at the level of viral entry through interactions with nectin-1 and non-muscle myosin heavy chains IIA and IIB, which are core proteins in virus-cell binding and virus-cell fusion, respectively4-6. Notably, Tmeff1-/- mice exhibited increased susceptibility to HSV-1 infection in the brain but not in the periphery. Within the brain, elevated viral load was observed specifically in neurons. Our study identifies TMEFF1 as a neuron-specific restriction factor essential for prevention of HSV-1 replication in the central nervous system.


Subject(s)
Antiviral Restriction Factors , Brain , Herpes Simplex , Herpesvirus 1, Human , Membrane Proteins , Neurons , Virus Internalization , Virus Replication , Animals , Female , Humans , Male , Mice , Antiviral Restriction Factors/metabolism , Brain/cytology , Brain/metabolism , Brain/pathology , Brain/virology , Cell Death , CRISPR-Cas Systems/genetics , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/growth & development , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology , Membrane Proteins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Neurons/virology , Neurons/metabolism , Viral Load , Nectins/metabolism , Nonmuscle Myosin Type IIA/metabolism , Nonmuscle Myosin Type IIB/metabolism , Interferon Type I , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/prevention & control , Neuroinflammatory Diseases/virology
18.
J Virol ; 98(8): e0073724, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39016551

ABSTRACT

Facilitates chromatin transcription (FACT) interacts with nucleosomes to promote gene transcription by regulating the dissociation and reassembly of nucleosomes downstream and upstream of RNA polymerase II (Pol II). A previous study reported that herpes simplex virus 1 (HSV-1) regulatory protein ICP22 interacted with FACT and was required for its recruitment to the viral DNA genome in HSV-1-infected cells. However, the biological importance of interactions between ICP22 and FACT in relation to HSV-1 infection is unclear. Here, we mapped the minimal domain of ICP22 required for its efficient interaction with FACT to a cluster of five basic amino acids in ICP22. A recombinant virus harboring alanine substitutions in this identified cluster led to the decreased accumulation of viral mRNAs from UL54, UL38, and UL44 genes, reduced Pol II occupancy of these genes in MRC-5 cells, and impaired HSV-1 virulence in mice following ocular or intracranial infection. Furthermore, the treatment of mice infected with wild-type HSV-1 with CBL0137, a FACT inhibitor currently being investigated in clinical trials, significantly improved the survival rate of mice. These results suggested that the interaction between ICP22 and FACT was required for efficient HSV-1 gene expression and pathogenicity. Therefore, FACT might be a potential therapeutic target for HSV-1 infection.IMPORTANCEICP22 is a well-known regulatory factor of HSV-1 gene expression, but its mechanism(s) are poorly understood. Although the interaction of FACT with ICP22 was reported previously, its significance in HSV-1 infection is unknown. Given that FACT is involved in gene transcription, it is of interest to investigate this interaction as it relates to HSV-1 gene expression. To determine a direct link between the interaction and HSV-1 infection, we mapped a minimal domain of ICP22 required for its efficient interaction with FACT and generated a recombinant virus carrying mutations in the identified domain. Using the recombinant virus, we obtained evidence suggesting that the interaction between ICP22 and FACT promoted Pol II transcription from HSV-1 genes and viral virulence in mice. In addition, CBL0137, an inhibitor of FACT, effectively protected mice from lethal HSV-1 infection, suggesting FACT might be a potential target for the development of novel anti-HSV drugs.


Subject(s)
Gene Expression Regulation, Viral , Herpes Simplex , Herpesvirus 1, Human , Immediate-Early Proteins , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Animals , Mice , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/genetics , Herpes Simplex/virology , Herpes Simplex/metabolism , Humans , Vero Cells , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Chlorocebus aethiops , Virus Replication , Virulence , Cell Line , Female , Mice, Inbred BALB C , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Transcription, Genetic
19.
Transpl Infect Dis ; 26(4): e14335, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39010324

ABSTRACT

BACKGROUND: Herpes simplex viruses (HSVs) frequently reactivate during immunosuppression and may be a risk factor for adverse outcomes after solid organ transplant (SOT). While suppressive antiviral therapy reduces the risk of symptomatic HSV reactivation, the kinetics of asymptomatic viral shedding with chronic immunosuppression after transplant are not well understood. We report the characteristics of oral HSV shedding among 15 HSV-1 seropositive SOT recipients (n = 8 liver, n = 7 kidney, median age 58.5 years, median 20 months post-transplant) who were not taking daily antiviral suppressive therapy. METHODS: Participants self-collected oral swabs three times daily for 6 weeks for HSV quantification and recorded the presence of oral symptoms or lesions in a diary. RESULTS: Sample collection adherence was high (median 122 swabs/person, range: 85.7%-101.6% of expected swabs). Most participants (n = 12, 80%) experienced at least one shedding episode, with a median shedding rate of 8.9% (range: 0%-33.6%). There were 32 total shedding episodes, 24 (75%) of which occurred without symptoms or lesions. For episodes of known duration, the median length was 21.8 hrs (interquartile range: 10.8-46.1 hrs). CONCLUSION: Most shedding episodes (78.1%) lasted >12 hrs, suggesting that twice-daily sampling may be sufficient to detect most episodes. These data show that self-collection of oral swabs is feasible for patients who have undergone SOTs and can provide insight into the frequency of oral HSV reactivation, which can be used to design future studies in this population.


Subject(s)
Herpesvirus 1, Human , Transplant Recipients , Virus Shedding , Humans , Pilot Projects , Male , Middle Aged , Female , Transplant Recipients/statistics & numerical data , Aged , Herpesvirus 1, Human/isolation & purification , Adult , Organ Transplantation/adverse effects , Herpes Simplex/virology , Virus Activation , Kidney Transplantation/adverse effects , Immunosuppression Therapy/adverse effects , Liver Transplantation/adverse effects
20.
J Virol Methods ; 329: 114994, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38971379

ABSTRACT

Herpes Simplex Virus Type 1 (HSV-1) is a widespread human pathogen known for causing a spectrum of clinical manifestations, ranging from mild cold sores to severe complications like encephalitis. Understanding the strain-specific variations of HSV-1 is crucial for elucidating its pathogenesis and developing targeted therapeutic interventions. In this multifaceted study, we investigated the strain-specific characteristics of HSV-1 using an in vivo rat model. Firstly, a pilot study was conducted to assess the capacity of three HSV-1 strains (Fisher (F), KOS (K), and MacIntyre (M)) to induce cold sores in rats. Remarkably, the F strain exhibited pronounced pathogenicity, inducing erythema, swelling, and disrupted epidermis with ulceration, distinguishing it from the K and M strains. Subsequently, the treatment capability of intravenous acyclovir injection in HSV-1 F strain-infected rats was evaluated. Acyclovir treatment resulted in a significant reduction in HSV-1 viral copy numbers in serum and dissected neuronal tissues, particularly in the spinal cord, brain, and lower lip. Lastly, whole genome sequencing data revealed that high-impact mutations occurred in the K and M strains within the UL49, US2, and US3 genes. These mutations may play a pivotal role in influencing viral replication, dissemination, pathogenesis, and infectivity. In contrast, the moderate missense variant mutations detected in the US12, US8, UL3, UL30, UL31, and UL36 genes appeared to have no effect on viral pathogenesis and infectivity, based on RT-PCR data for spinal cord, trigeminal nerve, brain, and the lower lip. These strain-specific mutations underscore the dynamic nature of HSV-1 evolution. Collectively, our findings contribute to a deeper understanding of HSV-1 strain diversity and pave the way for the development of targeted therapeutic strategies against this medically significant virus.


Subject(s)
Acyclovir , Antiviral Agents , Herpes Simplex , Herpesvirus 1, Human , High-Throughput Nucleotide Sequencing , Animals , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/isolation & purification , Herpesvirus 1, Human/drug effects , Rats , Herpes Simplex/virology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Acyclovir/pharmacology , Acyclovir/therapeutic use , Disease Models, Animal , Pilot Projects , Mutation , Virulence , Genome, Viral , Male
SELECTION OF CITATIONS
SEARCH DETAIL