Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 249
Filter
1.
Virol Sin ; 39(2): 251-263, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38219860

ABSTRACT

Viral encephalitis continues to be a significant public health concern. In our previous study, we discovered a lower expression of antiviral factors, such as IFN-ß, STING and IFI16, in the brain tissues of patients with Rasmussen's encephalitis (RE), a rare chronic neurological disorder often occurred in children, characterized by unihemispheric brain atrophy. Furthermore, a higher cumulative viral score of human herpes viruses (HHVs) was also found to have a significant positive correlation with the unihemispheric atrophy in RE. Type I IFNs (IFN-I) signaling is essential for innate anti-infection response by binding to IFN-α/ß receptor (IFNAR). In this study, we infected WT mice and IFNAR-deficient A6 mice with herpes simplex virus 1 (HSV-1) via periocular injection to investigate the relationship between IFN-I signaling and HHVs-induced brain lesions. While all mice exhibited typical viral encephalitis lesions in their brains, HSV-induced epilepsy was only observed in A6 mice. The gene expression matrix, functional enrichment analysis and protein-protein interaction network revealed four gene models that were positively related with HSV-induced epilepsy. Additionally, ten key genes with the highest scores were identified. Taken together, these findings indicate that intact IFN-I signaling can effectively limit HHVs induced neural symptoms and brain lesions, thereby confirming the positive correlation between IFN-I signaling repression and brain atrophy in RE and other HHVs encephalitis.


Subject(s)
Brain , Epilepsy , Herpesvirus 1, Human , Interferon Type I , Signal Transduction , Animals , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/immunology , Interferon Type I/metabolism , Interferon Type I/immunology , Mice , Brain/pathology , Brain/virology , Epilepsy/virology , Epilepsy/pathology , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/deficiency , Disease Models, Animal , Mice, Knockout , Mice, Inbred C57BL , Female , Protein Interaction Maps , Herpes Simplex/virology , Herpes Simplex/pathology , Herpes Simplex/immunology , Encephalitis, Herpes Simplex/virology , Encephalitis, Herpes Simplex/immunology , Encephalitis, Herpes Simplex/pathology , Humans
3.
Sci Rep ; 11(1): 21171, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34707143

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is the predominant cause of herpes simplex encephalitis (HSE), a condition characterized by acute inflammation and viral replication in the brain. Host genetics contribute to HSE onset, including monogenic defects in type I interferon signaling in cases of childhood HSE. Mouse models suggest a further contribution of immune cell-mediated inflammation to HSE pathogenesis. We have previously described a truncating mutation in the c-Rel transcription factor (RelC307X) that drives lethal HSE in 60% of HSV-1-infected RelC307X mice. In this study, we combined dual host-virus RNA sequencing with flow cytometry to explore cell populations and mechanisms involved in RelC307X-driven HSE. At day 5 postinfection, prior to HSE clinical symptom onset, elevated HSV-1 transcription was detected together with augmented host interferon-stimulated and inflammatory gene expression in the brainstems of high-responding RelC307X mice, predictive of HSE development. This early induction of host gene expression preceded pathological infiltration of myeloid and T cells in RelC307X mice at HSE onset by day 7. Thus, we establish c-Rel as an early regulator of viral and host responses during mouse HSE. These data further highlight the importance of achieving a balanced immune response and avoiding excess interferon-driven inflammation to promote HSE resistance.


Subject(s)
Encephalitis, Herpes Simplex/metabolism , Interferon Type I/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Animals , Encephalitis, Herpes Simplex/virology , Female , Male , Mice , Mice, Inbred C57BL , Mutation , Proto-Oncogene Proteins c-rel/genetics , Signal Transduction , Simplexvirus/genetics , Simplexvirus/pathogenicity , Simplexvirus/physiology , T-Lymphocytes/metabolism , T-Lymphocytes/virology
4.
Sci Rep ; 11(1): 18688, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34548521

ABSTRACT

Herpes simplex virus 1 (HSV-1) is responsible for herpes simplex virus encephalitis (HSE), associated with a 70% mortality rate in the absence of treatment. Despite intravenous treatment with acyclovir, mortality remains significant, highlighting the need for new anti-herpetic agents. Herein, we describe a novel neurovirulent recombinant HSV-1 (rHSV-1), expressing the fluorescent tdTomato and Gaussia luciferase (Gluc) enzyme, generated by the Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) (CRISPR-Cas9) system. The Gluc activity measured in the cell culture supernatant was correlated (P = 0.0001) with infectious particles, allowing in vitro monitoring of viral replication kinetics. A significant correlation was also found between brain viral titers and Gluc activity in plasma (R2 = 0.8510, P < 0.0001) collected from BALB/c mice infected intranasally with rHSV-1. Furthermore, evaluation of valacyclovir (VACV) treatment of HSE could also be performed by analyzing Gluc activity in mouse plasma samples. Finally, it was also possible to study rHSV-1 dissemination and additionally to estimate brain viral titers by in vivo imaging system (IVIS). The new rHSV-1 with reporter proteins is not only as a powerful tool for in vitro and in vivo antiviral screening, but can also be used for studying different aspects of HSE pathogenesis.


Subject(s)
Encephalitis, Herpes Simplex/physiopathology , Herpesvirus 1, Human/isolation & purification , Animals , Antiviral Agents/therapeutic use , Base Sequence , Blood-Brain Barrier , Brain/virology , Chlorocebus aethiops , Encephalitis, Herpes Simplex/drug therapy , Encephalitis, Herpes Simplex/virology , Genes, Reporter , Genes, Viral , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Luminescence , Mice , Mice, Inbred BALB C , Multiplex Polymerase Chain Reaction/methods , Valacyclovir/therapeutic use , Vero Cells , Viral Load , Virus Replication/genetics
5.
Neurosci Lett ; 765: 136172, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34433098

ABSTRACT

BACKGROUND: Herpes simplex virus encephalitis (HSE) is an acute central nervous system infectious disease caused by herpes simplex virus (HSV). Currently, there is no effective treatment for HSE infection, which produces many pro-inflammatory factors. Kaempferol-3-O-rhamnoside (K-3-rh) is a plant flavonoid. This study was investigated the anti-inflammatory effect of K-3-rh on encephalitis induced by HSV-1. METHODS: HSV-1 was co-cultured with VERO cells. Cells were divided into four groups, including the control group, virus group, K-3-rh group, Astragalus polysaccharide (APS) group and dexamethasone group. Flow cytometry were utilized to determine cell apoptosis, respectively. Proteins and mRNAs were estimated by western blot and qRT-PCR, respectively. RESULTS: After viral infection, the cytokines were significantly increased. After K-3-rh intervention, the expression of tumor necrosis factor-α (TNF-α), interleukin-1 beta (IL-1ß), and nitric oxide (NO) in microglia were reduced contrast with those in the virus group, and the expression of interleukin-10 (IL-10) did not change. After viral infection, the apoptotic rate increased significantly, and K-3-rh could inhibit viral-induced apoptosis in the microglial cell line. The induction of microglia apoptosis was achieved by cytochrome c and caspase-9-mediated mitochondrial pathway. Also, the pathological changes of brain tissue in mice of each drug intervention group were alleviated. CONCLUSIONS: In conclusion, K-3-rh had the potential to reduce HSV-1-induced brain injury by reducing the secretion of microglial pro-inflammatory factors, inducing apoptosis of microglia cells, and through cytochrome C and caspase-3 pathway.


Subject(s)
Encephalitis, Herpes Simplex/drug therapy , Glycosides/pharmacology , Herpesvirus 1, Human/immunology , Kaempferols/pharmacology , Microglia/drug effects , Animals , Apoptosis/drug effects , Apoptosis/immunology , Caspase 3/metabolism , Cell Line , Chlorocebus aethiops , Cytochromes c/metabolism , Disease Models, Animal , Encephalitis, Herpes Simplex/immunology , Encephalitis, Herpes Simplex/pathology , Encephalitis, Herpes Simplex/virology , Glycosides/therapeutic use , Humans , Kaempferols/therapeutic use , Mice , Microglia/immunology , Microglia/pathology , Microglia/virology , Vero Cells
6.
Neurosci Lett ; 763: 136170, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34391869

ABSTRACT

To explore the differently expressed cytokines and chemokines to understand the pathways that lead to herpes simplex encephalitis (HSE). Mice in the experimental group were inoculated intracranially with HSV-1. A high-throughput cytokine chip assay was employed to assess the expression of cytokines/chemokines in the mice brain. GO, KEGG, and PPIs analyses were used to investigate the biological process (BP), pathways and interaction network of the differently expressed proteins (DEPs) in HSE. 13 DEPs and various proteins-related signal pathways were identified in HSE, including three new factors (IL-1α, MIP-1γ, and sTNF RI). The proteins were mainly implicated in leukocyte activation and chemotaxis. Additionally, the DEPs constituted a pivotal protein interaction network where IL-6 might be a mediator. 13 DEPs and a series of related signal pathways were associated with the pathophysiological mechanisms responsible for HSE. IL-6 might be a key mediator in the inflammatory responses to the disease.


Subject(s)
Brain/pathology , Cytokines/metabolism , Encephalitis, Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Animals , Brain/immunology , Brain/virology , Chlorocebus aethiops , Cytokines/analysis , Disease Models, Animal , Encephalitis, Herpes Simplex/pathology , Encephalitis, Herpes Simplex/virology , Gene Expression Profiling , Humans , Male , Mice , Protein Interaction Maps/genetics , Protein Interaction Maps/immunology , Vero Cells
7.
J Neurovirol ; 27(3): 493-497, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33788139

ABSTRACT

Herpes simplex virus encephalitis (HSE) is the most common sporadic fatal encephalitis. Although timely administered acyclovir treatment decreases mortality, neuropsychiatric sequelae is still common among survivors. Magnetic resonance imaging is frequently utilized for the diagnosis of HSE, which typically involves temporal lobe(s) and can be mixed with brain tumors involving the same area. Here, we report a case of HSE, who received acyclovir with a delay of 90 days because of presumptive tumor diagnosis and survived with minimal sequelae.


Subject(s)
Acyclovir/therapeutic use , Antiviral Agents/therapeutic use , Encephalitis, Herpes Simplex/drug therapy , Herpesvirus 1, Human/drug effects , Temporal Lobe/drug effects , Adult , Delayed Diagnosis , Encephalitis, Herpes Simplex/diagnostic imaging , Encephalitis, Herpes Simplex/pathology , Encephalitis, Herpes Simplex/virology , Female , Herpesvirus 1, Human/growth & development , Herpesvirus 1, Human/pathogenicity , Humans , Magnetic Resonance Imaging , Temporal Lobe/diagnostic imaging , Temporal Lobe/pathology , Temporal Lobe/virology , Treatment Outcome
8.
Nat Commun ; 11(1): 4894, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32994400

ABSTRACT

Identification of the complete set of translated genes of viruses is important to understand viral replication and pathogenesis as well as for therapeutic approaches to control viral infection. Here, we use chemical proteomics, integrating bio-orthogonal non-canonical amino acid tagging and high-resolution mass spectrometry, to characterize the newly synthesized herpes simplex virus 1 (HSV-1) proteome in infected cells. In these infected cells, host cellular protein synthesis is shut-off, increasing the chance to preferentially detect viral proteomes. We identify nine previously cryptic orphan protein coding sequences whose translated products are expressed in HSV-1-infected cells. Functional characterization of one identified protein, designated piUL49, shows that it is critical for HSV-1 neurovirulence in vivo by regulating the activity of virally encoded dUTPase, a key enzyme that maintains accurate DNA replication. Our results demonstrate that cryptic orphan protein coding genes of HSV-1, and probably other large DNA viruses, remain to be identified.


Subject(s)
Encephalitis, Herpes Simplex/virology , Herpesvirus 1, Human/pathogenicity , Pyrophosphatases/metabolism , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Brain/pathology , Brain/virology , Chlorocebus aethiops , DNA Replication , Disease Models, Animal , Encephalitis, Herpes Simplex/pathology , Female , Genes, Viral/genetics , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/genetics , Humans , Mice , Protein Biosynthesis , Proteomics/methods , Vero Cells , Viral Proteins/genetics , Virulence Factors/genetics , Virus Replication
9.
Rinsho Shinkeigaku ; 60(7): 461-465, 2020 Jul 31.
Article in Japanese | MEDLINE | ID: mdl-32536661

ABSTRACT

In January 2008, a 59-year-old man with a history of diabetes mellitus was admitted to our hospital with herpes simplex virus (HSV) encephalitis of his right temporal lobe, which was diagnosed by PCR testing of his cerebrospinal fluid (CSF). He was treated with intravenous acyclovir for three weeks and made a full recovery. On discharge, his CSF was negative for HSV on PCR testing. Seven years later, in March 2015, the man was readmitted to our hospital with fever, disorientation, and nominal dysphasia. Diffusion-weighted MRI of his head revealed a high-intensity area in his left temporal lobe. Testing of his CSF revealed a moderately increased monocyte count and HSV on PCR testing, so he was diagnosed with recurrent HSV encephalitis. He was treated with intravenous acyclovir for three weeks. On discharge, his CSF was negative for HSV on PCR testing, but he had mild residual amnesia. There have been few reports of HSV encephalitis with viral reactivation recurring after a long remission period in adults. This case illustrates the need for prolonged follow up of individuals with HSV encephalitis in order to detect recurrences.


Subject(s)
Acyclovir/therapeutic use , Encephalitis, Herpes Simplex/diagnosis , Encephalitis, Herpes Simplex/drug therapy , Aged , Cerebrospinal Fluid/virology , Encephalitis, Herpes Simplex/virology , Humans , Male , Polymerase Chain Reaction , Recurrence , Simplexvirus/isolation & purification , Time Factors , Treatment Outcome
10.
J Infect Chemother ; 26(7): 736-740, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32201195

ABSTRACT

BACKGROUND: Febrile neonates and young infants presenting with seizure require immediate evaluation and treatment. Herein we experienced two young infants with parechovirus-A3 (PeV-A3) encephalitis, initially presented with focal seizure suspecting herpes simplex virus (HSV) encephalitis. CASES: We have experienced 2 infantile cases, initially presented with focal seizure. At presentation, HSV encephalitis was strongly suspected and empiric acyclovir therapy was started; however, serum and/or cerebrospinal fluid (CSF) PCR for HSV were negative. Instead, serum and/or CSF PCR for parechovirus-A was positive. PeV-A3 infection was confirmed by genetic sequence analyses. Both cases required multiple anticonvulsant therapy and intensive care for intractable seizure. Diffusion-weighted imaging of brain magnetic resonance imaging (MRI) showed distinct findings; high-intensity lesions in the gray matter of parietal and occipital lobes in Case 1, and bilateral decreased diffusion of the deep white matter and corpus callosum in Case 2. We have followed two cases more than four years; Case 1 developed epilepsy, has been on an anticonvulsant to control her seizure. Case 2 has significant neurodevelopmental delay, unable to stand or communicate with language. CONCLUSIONS: PeV-A3 encephalitis needs to be in differential diagnosis when neonates and young infants present with focal seizure, mimicking HSV encephalitis. Special attention may be necessary in patients with PeV-A3 encephalitis given it could present with intractable seizure with high morbidity in a long-term.


Subject(s)
Encephalitis, Herpes Simplex/diagnosis , Encephalitis, Viral/diagnosis , Parechovirus/isolation & purification , Picornaviridae Infections/diagnosis , Seizures/virology , Brain/diagnostic imaging , DNA, Viral/isolation & purification , Diagnosis, Differential , Diffusion Magnetic Resonance Imaging , Encephalitis, Herpes Simplex/virology , Encephalitis, Viral/cerebrospinal fluid , Encephalitis, Viral/complications , Encephalitis, Viral/virology , Epilepsy/drug therapy , Epilepsy/virology , Female , Humans , Infant , Infant, Premature , Male , Neurodevelopmental Disorders/virology , Parechovirus/genetics , Picornaviridae Infections/cerebrospinal fluid , Picornaviridae Infections/complications , Picornaviridae Infections/virology , Polymerase Chain Reaction , RNA, Viral/blood , RNA, Viral/cerebrospinal fluid , RNA, Viral/isolation & purification , Seizures/blood , Seizures/cerebrospinal fluid , Seizures/diagnosis , Simplexvirus/genetics , Simplexvirus/isolation & purification
11.
Hum Genet ; 139(6-7): 911-918, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32040615

ABSTRACT

Herpes simplex virus 1 (HSV-1) encephalitis (HSE) is the most common sporadic viral encephalitis in Western countries. Over the last 15 years, human genetic and immunological studies have provided proof-of-principle that childhood HSE can result from inborn errors of central nervous system (CNS)-specific, cell-intrinsic immunity to HSV-1. HSE-causing mutations of eight genes disrupt known (TLR3-dependent IFN-α/ß immunity) and novel (dependent on DBR1 or snoRNA31) antiviral mechanisms. Monogenic inborn errors confer susceptibility to forebrain (TLR3-IFN or snoRNA31) or brainstem (DBR1) HSE. Most of these disorders display incomplete clinical penetrance, with the possible exception of DBR1 deficiency. They account for a small, but non-negligible proportion of cases (about 7%). These findings pave the way for the gradual definition of the genetic and immunological architecture of childhood HSE, with both biological and clinical implications.


Subject(s)
Central Nervous System Diseases/genetics , Encephalitis, Herpes Simplex/genetics , Genetic Predisposition to Disease , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Host-Pathogen Interactions/genetics , Immunity, Cellular/immunology , Central Nervous System Diseases/epidemiology , Central Nervous System Diseases/immunology , Central Nervous System Diseases/virology , Child , Encephalitis, Herpes Simplex/epidemiology , Encephalitis, Herpes Simplex/immunology , Encephalitis, Herpes Simplex/virology , Herpes Simplex/complications , Herpes Simplex/virology , Herpesvirus 1, Human/pathogenicity , Host-Pathogen Interactions/immunology , Humans , Mutation
12.
J Infect Dis ; 221(8): 1295-1303, 2020 03 28.
Article in English | MEDLINE | ID: mdl-31268141

ABSTRACT

BACKGROUND: Herpes zoster ophthalmicus occurs primarily in elderly or immunocompromised individuals after reactivation of varicella zoster virus (VZV). Recurrences of zoster ophthalmicus are uncommon because the reactivation efficiently boosts anti-VZV immunity. A 28-year-old female presented to our clinic with a history of multiple recurrences of zoster ophthalmicus. METHODS: Whole-exome sequencing (WES), analyses of VZV T-cell immunity, and pathogen recognition receptor function in primary antigen-presenting cells (APCs) and fibroblasts were performed. RESULTS: Normal VZV-specific T-cell immunity and antibody response were detected. Whole-exome sequencing identified a heterozygous nonsynonymous variant (c.2324C > T) in the Toll-like receptor 3 (TLR3) gene resulting in formation of a premature stop-codon. This alteration could potentially undermine TLR3 signaling in a dominant-negative fashion. Therefore, we investigated TLR3 signaling responses in APCs and fibroblasts from the patient. The APCs responded efficiently to stimulation with TLR3 ligands, whereas the responses from the fibroblasts were compromised. CONCLUSIONS: We report a novel TLR3 variant associated with recurrent zoster ophthalmicus. Toll-like receptor 3 responses that were unaffected in APCs but diminished in fibroblasts are in line with previous reports linking TLR3 deficiency with herpes simplex virus encephalitis. Mechanisms involving compromised viral sensing in infected cells may thus be central to the described immunodeficiency.


Subject(s)
Herpes Zoster Ophthalmicus/virology , Herpesvirus 3, Human/pathogenicity , Mutation/genetics , Toll-Like Receptor 3/genetics , Adult , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/virology , Female , Fibroblasts/virology , Herpes Zoster/genetics , Herpes Zoster/virology , Herpes Zoster Ophthalmicus/genetics , Humans , Immunocompromised Host/genetics
13.
Nat Med ; 25(12): 1873-1884, 2019 12.
Article in English | MEDLINE | ID: mdl-31806906

ABSTRACT

Herpes simplex virus-1 (HSV-1) encephalitis (HSE) is typically sporadic. Inborn errors of TLR3- and DBR1-mediated central nervous system cell-intrinsic immunity can account for forebrain and brainstem HSE, respectively. We report five unrelated patients with forebrain HSE, each heterozygous for one of four rare variants of SNORA31, encoding a small nucleolar RNA of the H/ACA class that are predicted to direct the isomerization of uridine residues to pseudouridine in small nuclear RNA and ribosomal RNA. We show that CRISPR/Cas9-introduced bi- and monoallelic SNORA31 deletions render human pluripotent stem cell (hPSC)-derived cortical neurons susceptible to HSV-1. Accordingly, SNORA31-mutated patient hPSC-derived cortical neurons are susceptible to HSV-1, like those from TLR3- or STAT1-deficient patients. Exogenous interferon (IFN)-ß renders SNORA31- and TLR3- but not STAT1-mutated neurons resistant to HSV-1. Finally, transcriptome analysis of SNORA31-mutated neurons revealed normal responses to TLR3 and IFN-α/ß stimulation but abnormal responses to HSV-1. Human SNORA31 thus controls central nervous system neuron-intrinsic immunity to HSV-1 by a distinctive mechanism.


Subject(s)
Encephalitis, Herpes Simplex/genetics , Herpesvirus 1, Human/genetics , Neurons/immunology , RNA, Small Nucleolar/genetics , Adult , Central Nervous System/immunology , Central Nervous System/virology , Child, Preschool , Encephalitis, Herpes Simplex/immunology , Encephalitis, Herpes Simplex/pathology , Encephalitis, Herpes Simplex/virology , Female , Genetic Predisposition to Disease , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Humans , Immunity/genetics , Infant , Male , Metagenome/genetics , Metagenome/immunology , Middle Aged , Neurons/virology , RNA, Small Nucleolar/immunology
16.
Crit Rev Microbiol ; 45(5-6): 581-594, 2019.
Article in English | MEDLINE | ID: mdl-31512533

ABSTRACT

Microglia, as brain-resident macrophages, are the first line of defense against brain invading pathogens. Further, their dysfunction has been recognized to be closely associated with mounting CNS diseases. Of note, chronic HSV-1 infection leads to the persistent activation of microglia, which elicit a comprehensive response by generating certain factors with neurotoxic and neuroprotective effects. CNS infection with HSV-1 results in herpes simplex encephalitis and herpes simplex keratitis. Microglial immune response plays a crucial role in the development of these diseases. Moreover, HSV-1 infection is strongly associated with several CNS diseases, especially Alzheimer's disease and schizophrenia. These CNS diseases can be effectively ameliorated by eliciting an appropriate immune response, such as inhibition of microglial proliferation and activation. Therefore, it is crucial to reassess the positive and negative roles of microglia in HSV-1 CNS infection for a more comprehensive and detailed understanding of the relationship between microglia and CNS diseases. Hence, the present review focuses on the dual roles of microglia in mediating HSV-1 CNS infection, as well as on the strategy of targeting microglia to ameliorate CNS diseases. Further research in this field can help comprehensively elucidate the dual role of the microglial immune response in HSV-1 CNS infection, providing a theoretical basis for identifying therapeutic targets against overactive microglia in CNS diseases and HSV-1 infection.


Subject(s)
Encephalitis, Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Microglia/immunology , Animals , Encephalitis, Herpes Simplex/immunology , Herpesvirus 1, Human/genetics , Humans , Microglia/virology
17.
J Neurovirol ; 25(4): 525-539, 2019 08.
Article in English | MEDLINE | ID: mdl-31144288

ABSTRACT

Herpes simplex virus 1 (HSV-1) is a predominant cause of herpes simplex encephalitis (HSE), leading to a high mortality rate and severe neurological sequelae worldwide. HSE is typically accompanied by the blood-brain barrier (BBB) disruption, but the underlying mechanisms are unclear. To explore the disruption mechanisms of the BBB, quantitative analysis of the cellular proteome was carried out to investigate the proteomic changes that occur after infection. In this study, bEnd.3 cells were infected with HSV-1, followed by liquid chromatography-tandem mass spectrometry. A total of 6761 proteins were identified in three independent mass spectrometry analyses. Compared to the uninfected cells, 386 and 293 differentially expressed proteins were markedly upregulated or downregulated, respectively. Bioinformatic analysis showed that the activator protein-1 factor, including Fos, Jun, and ATF family proteins and cell adhesion molecules were significantly changed. Further validation of the changes observed for these proteins was carried out by western blotting and quantitative real-time PCR. Transendothelial electrical resistance (TEER) studies were performed to explore the effects of ATF3, Fra1, or JunB overexpression on the function of bEnd.3 cells. Characterization of the differential expression of these proteins in bEnd.3 cells will facilitate further exploration of BBB disruption upon HSV-1 infection.


Subject(s)
Activating Transcription Factor 3/genetics , Encephalitis, Herpes Simplex/genetics , Endothelial Cells/metabolism , Herpesvirus 1, Human/physiology , Proto-Oncogene Proteins c-fos/genetics , Transcription Factors/genetics , Activating Transcription Factor 3/metabolism , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/virology , Brain/metabolism , Brain/virology , Cell Line , Encephalitis, Herpes Simplex/metabolism , Encephalitis, Herpes Simplex/virology , Endothelial Cells/virology , Gene Expression Regulation , Gene Ontology , Herpesvirus 1, Human/pathogenicity , Host-Pathogen Interactions/genetics , Humans , Mice , Models, Biological , Molecular Sequence Annotation , Proteome/classification , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Virus Replication
18.
BMJ Case Rep ; 12(5)2019 May 27.
Article in English | MEDLINE | ID: mdl-31138593

ABSTRACT

A 60-year-old man with a history of severe herpes simplex virus type 1 (HSV-1) encephalitis 2 years prior presented with acute onset of visual loss in the left eye. Dilated funduscopic examination showed retinitis and occlusive vasculitis with retinal necrosis. PCR of the vitreous fluid was positive for HSV-1, and he was diagnosed with acute retinal necrosis (ARN) due to HSV-1. The patient was treated with intravenous acyclovir and intravitreous foscarnet for 2 weeks, followed by high dose oral valacyclovir for 2 weeks. He was subsequently placed on planned life-long suppressive valacyclovir. His case demonstrates that acute visual loss concomitant with or subsequent to HSV-1 encephalitis warrants suspicion of ARN. Prompt therapy with effective antiviral medication is necessary to reduce the risk of sight-threatening complications. Chronic suppression with oral antiviral therapy after ARN is recommended to prevent involvement of the contralateral eye, though there is no consensus on the duration and dosage of antivirals.


Subject(s)
Encephalitis, Herpes Simplex/complications , Encephalitis, Herpes Simplex/diagnosis , Retinal Necrosis Syndrome, Acute/diagnostic imaging , Retinal Necrosis Syndrome, Acute/etiology , Acute Disease , Acyclovir/administration & dosage , Acyclovir/therapeutic use , Antiviral Agents/therapeutic use , Diagnosis, Differential , Encephalitis, Herpes Simplex/virology , Eye Infections, Viral/complications , Eye Infections, Viral/diagnosis , Foscarnet/administration & dosage , Foscarnet/therapeutic use , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/isolation & purification , Humans , Intravitreal Injections , Male , Middle Aged , Ophthalmoscopes , Rare Diseases , Retinal Necrosis Syndrome, Acute/drug therapy , Retinal Necrosis Syndrome, Acute/virology , Treatment Outcome , Valacyclovir/administration & dosage , Valacyclovir/therapeutic use
19.
Nat Commun ; 10(1): 2153, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31089128

ABSTRACT

The gut commensal Bacteroides fragilis or its capsular polysaccharide A (PSA) can prevent various peripheral and CNS sterile inflammatory disorders. Fatal herpes simplex encephalitis (HSE) results from immune pathology caused by uncontrolled invasion of the brainstem by inflammatory monocytes and neutrophils. Here we assess the immunomodulatory potential of PSA in HSE by infecting PSA or PBS treated 129S6 mice with HSV1, followed by delayed Acyclovir (ACV) treatment as often occurs in the clinical setting. Only PSA-treated mice survived, with dramatically reduced brainstem inflammation and altered cytokine and chemokine profiles. Importantly, PSA binding by B cells is essential for induction of regulatory CD4+ and CD8+ T cells secreting IL-10 to control innate inflammatory responses, consistent with the lack of PSA mediated protection in Rag-/-, B cell- and IL-10-deficient mice. Our data reveal the translational potential of PSA as an immunomodulatory symbiosis factor to orchestrate robust protective anti-inflammatory responses during viral infections.


Subject(s)
Bacteroides fragilis/immunology , Encephalitis, Herpes Simplex/immunology , Gastrointestinal Microbiome/immunology , Herpesvirus 1, Human/immunology , Polysaccharides, Bacterial/immunology , Acyclovir/therapeutic use , Animals , Antiviral Agents/therapeutic use , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bacteroides fragilis/metabolism , Chlorocebus aethiops , Disease Models, Animal , Encephalitis, Herpes Simplex/drug therapy , Encephalitis, Herpes Simplex/virology , Female , Herpesvirus 1, Human/pathogenicity , Host Microbial Interactions/immunology , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Male , Mice , Mice, Knockout , Polysaccharides, Bacterial/metabolism , Symbiosis/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vero Cells
20.
Jpn J Ophthalmol ; 63(4): 304-309, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31054049

ABSTRACT

PURPOSE: Acute retinal necrosis (ARN) is a severe necrotizing retinitis caused by varicella-zoster virus or herpes simplex virus (HSV) that often results in blindness. Occasionally, HSV-caused ARN develops after herpes simplex encephalitis (HSE). It remains unknown, however, when and how often ARN develops after HSE. To investigate the actual conditions of patients with ARN following HSE and the interval period between the prior HSE and the onset of ARN, a retrospective nationwide survey of the Japanese population was performed. STUDY DESIGN: Retrospective. METHODS: Questionnaires were sent out to the neurology and ophthalmology departments of teaching hospitals in Japan. They inquired about HSE patients in neurology departments and ARN patients diagnosed with HSV in ophthalmology departments. The proportion of the HSV-ARN patients with a history of HSE and their interval periods were obtained from the questionnaires returned by the ophthalmology departments. RESULTS: Neurology departments of 324 hospitals responded to the questionnaires (response proportion: 40.9%), and 53 HSE cases were reported. Ophthalmology departments of 535 hospitals responded the questionnaires (response proportion: 54.3%), and 67 HSV-ARN cases were reported. Among the 67 HSV-ARN cases, 16 (23.9%) had histories of prior HSE. Although the interval periods from the prior HSE to the onset of HSV-ARN varied among cases, nearly half developed HSV-ARN within 2 years. CONCLUSIONS: This nationwide survey of the Japanese population showed that HSV-ARN develops after HSE in higher frequency within 2 years. Neurologists and ophthalmologists should be aware that HSE survivors have a risk of contracting HSV-ARN.


Subject(s)
Encephalitis, Herpes Simplex/complications , Eye Infections, Viral/etiology , Retinal Necrosis Syndrome, Acute/etiology , Surveys and Questionnaires , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Viral/analysis , DNA, Viral/analysis , Encephalitis, Herpes Simplex/epidemiology , Encephalitis, Herpes Simplex/virology , Eye Infections, Viral/diagnosis , Eye Infections, Viral/epidemiology , Female , Humans , Incidence , Japan/epidemiology , Magnetic Resonance Imaging , Male , Middle Aged , Retinal Necrosis Syndrome, Acute/diagnosis , Retinal Necrosis Syndrome, Acute/epidemiology , Retrospective Studies , Simplexvirus/genetics , Simplexvirus/immunology , Tomography, X-Ray Computed , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...