Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 278
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
2.
Brain ; 147(4): 1130-1148, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38092513

ABSTRACT

Herpes simplex virus encephalitis (HSE) is the leading cause of non-epidemic encephalitis in the developed world and, despite antiviral therapy, mortality and morbidity is high. The emergence of post-HSE autoimmune encephalitis reveals a new immunological paradigm in autoantibody-mediated disease. A reductionist evaluation of the immunobiological mechanisms in HSE is crucial to dissect the origins of post-viral autoimmunity and supply rational approaches to the selection of immunotherapeutics. Herein, we review the latest evidence behind the phenotypic progression and underlying immunobiology of HSE including the cytokine/chemokine environment, the role of pathogen-recognition receptors, T- and B-cell immunity and relevant inborn errors of immunity. Second, we provide a contemporary review of published patients with post-HSE autoimmune encephalitis from a combined cohort of 110 patients. Third, we integrate novel mechanisms of autoimmunization in deep cervical lymph nodes to explore hypotheses around post-HSE autoimmune encephalitis and challenge these against mechanisms of molecular mimicry and others. Finally, we explore translational concepts where neuroglial surface autoantibodies have been observed with other neuroinfectious diseases and those that generate brain damage including traumatic brain injury, ischaemic stroke and neurodegenerative disease. Overall, the clinical and immunological landscape of HSE is an important and evolving field, from which precision immunotherapeutics could soon emerge.


Subject(s)
Autoimmune Diseases of the Nervous System , Brain Ischemia , Encephalitis, Herpes Simplex , Neurodegenerative Diseases , Stroke , Humans , Autoimmunity , Encephalitis, Herpes Simplex/drug therapy , Encephalitis, Herpes Simplex/pathology , Autoantibodies , Simplexvirus
3.
Infect Dis (Lond) ; 55(10): 694-705, 2023 10.
Article in English | MEDLINE | ID: mdl-37395107

ABSTRACT

OBJECTIVES: We examined the temporal changes of the CSF proteome in patients with herpes simplex encephalitis (HSE) during the course of the disease, in relation to anti-N-methyl-D-aspartate receptor (NMDAR) serostatus, corticosteroid treatment, brain MRI and neurocognitive performance. METHODS: Patients were retrospectively included from a previous prospective trial with a pre-specified CSF sampling protocol. Mass spectrometry data of the CSF proteome were processed using pathway analysis. RESULTS: We included 48 patients (110 CSF samples). Samples were grouped based on time of collection relative to hospital admission - T1: ≤ 9 d, T2: 13-28 d, T3: ≥ 68 d. At T1, a strong multi-pathway response was seen including acute phase response, antimicrobial pattern recognition, glycolysis and gluconeogenesis. At T2, most pathways activated at T1 were no longer significantly different from T3. After correction for multiplicity and considering the effect size threshold, 6 proteins were significantly less abundant in anti-NMDAR seropositive patients compared to seronegative: procathepsin H, heparin cofactor 2, complement factor I, protein AMBP, apolipoprotein A1 and polymeric immunoglobulin receptor. No significant differences in individual protein levels were found in relation to corticosteroid treatment, size of brain MRI lesion or neurocognitive performance. CONCLUSIONS: We show a temporal change in the CSF proteome in HSE patients during the course of the disease. This study provides insight into quantitative and qualitative aspects of the dynamic pathophysiology and pathway activation patterns in HSE and prompts for future studies on the role of apolipoprotein A1 in HSE, which has previously been associated with NMDAR encephalitis.


Subject(s)
Encephalitis, Herpes Simplex , Nervous System Diseases , Humans , Encephalitis, Herpes Simplex/complications , Encephalitis, Herpes Simplex/pathology , Proteome , Apolipoprotein A-I , Retrospective Studies
4.
Virol Sin ; 38(1): 108-118, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36436797

ABSTRACT

Herpes simplex virus-1 (HSV-1) is a widespread neurotropic virus that can reach the brain and cause a rare but acute herpes simplex encephalitis (HSE) with a high mortality rate. Most patients present with changes in neurological and behavioral status, and survivors suffer long-term neurological sequelae. To date, the pathogenesis leading to brain damage is still not well understood. HSV-1 induced encephalitis in the central nervous system (CNS) in animals are usually very diffuse and progressing rapidly, and mostly fatal, making the analysis difficult. Here, we established a mouse model of HSE via intracerebral inoculation of modified version of neural-attenuated strains of HSV-1 (deletion of ICP34.5 and inserting a strong promoter into the latency-associated transcript region), in which the LMR-αΔpA strain initiated moderate productive infection, leading to strong host immune and inflammatory response characterized by persistent microglia activation. This viral replication activity and prolonged inflammatory response activated signaling pathways in neuronal damage, amyloidosis, Alzheimer's disease, and neurodegeneration, eventually leading to neuronal loss and behavioral changes characterized by hypokinesia. Our study reveals detailed pathogenic processes and persistent inflammatory responses in the CNS and provides a controlled, mild and non-lethal HSE model for studying long-term neuronal injury and increased risk of neurodegenerative diseases due to HSV-1 infection.


Subject(s)
Encephalitis, Herpes Simplex , Herpes Simplex , Herpesvirus 1, Human , Mice , Animals , Herpesvirus 1, Human/physiology , Encephalitis, Herpes Simplex/complications , Encephalitis, Herpes Simplex/pathology , Brain/pathology , Inflammation
5.
Autophagy ; 19(2): 734-736, 2023 02.
Article in English | MEDLINE | ID: mdl-35849507

ABSTRACT

Herpes simplex encephalitis (HSE), mainly caused by herpes simplex virus type 1 (HSV-1), is a severe central nervous system disease commonly followed by cognitive impairment, behavioral changes, and focal neurological signs. Although increasing evidence implicates the central role of microglia in HSE progression, the intrinsic restrictors or the acquired environmental factors that balance the beneficial or detrimental immune responses in microglia remain unclear. In a recent study, we find that a gut microbial metabolite activates mitophagy to regulate microglia-mediated neuroinflammation and to mitigate HSE progression. HSV-1 neurotropic infection causes gut microbiota dysbiosis and microglial antiviral immune response, whereas depletion of gut microbiota by oral antibiotics treatment further results in hyperactivated microglia and exacerbated HSE pathology. Notably, exogenous administration of nicotinamide n-oxide (NAMO), an oxidative product of nicotinamide mainly produced by intestinal neomycin-sensitive bacteria, especially Lactobacillus gasseri and Lactobacillus reuteri, can significantly suppress HSE progression. Mechanistically, HSV-1 infection causes mitochondrial dysfunction and impairs mitophagy to activate microglia and promote proinflammatory cytokine production, whereas NAMO restores NAD+-dependent mitophagy to restrain microglial over-activation and to prevent HSV-1 early infection in neuronal cells. This work reveals a novel function of gut microbial metabolites as intrinsic regulators of microglia homeostasis and neuroinflammation via mitophagy.Abbreviations: AD: Alzheimer disease; ABX: antibiotics; HSE: herpes simplex encephalitis; HSV-1: herpes simplex virus type 1; NAD+: nicotinamide adenine dinucleotide; NAMO: nicotinamide n-oxide; SCFAs: short-chain fatty acids.


Subject(s)
Encephalitis, Herpes Simplex , Herpesvirus 1, Human , Humans , Encephalitis, Herpes Simplex/pathology , Microglia/metabolism , Neuroinflammatory Diseases , Brain-Gut Axis , Mitophagy , Autophagy
6.
Sci Immunol ; 7(77): eabq4531, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36399538

ABSTRACT

Herpes simplex virus 1 (HSV-1) infects several billion people worldwide and can cause life-threatening herpes simplex encephalitis (HSE) in some patients. Monogenic defects in components of the type I interferon system have been identified in patients with HSE, emphasizing the role of inborn errors of immunity underlying HSE pathogenesis. Here, we identify compound heterozygous loss-of-function mutations in the gene GTF3A encoding for transcription factor IIIA (TFIIIA), a component of the RNA polymerase III complex, in a patient with common variable immunodeficiency and HSE. Patient fibroblasts and GTF3A gene-edited cells displayed impaired HSV-1-induced innate immune responses and enhanced HSV-1 replication. Chromatin immunoprecipitation sequencing analysis identified the 5S ribosomal RNA pseudogene 141 (RNA5SP141), an endogenous ligand of the RNA sensor RIG-I, as a transcriptional target of TFIIIA. GTF3A mutant cells exhibited diminished RNA5SP141 expression and abrogated RIG-I activation upon HSV-1 infection. Our work unveils a crucial role for TFIIIA in transcriptional regulation of a cellular RIG-I agonist and shows that GTF3A genetic defects lead to impaired cell-intrinsic anti-HSV-1 responses and can predispose to HSE.


Subject(s)
Encephalitis, Herpes Simplex , Herpesvirus 1, Human , Humans , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/pathology , Pseudogenes , RNA , Ligands , Transcription Factor TFIIIA/genetics , Herpesvirus 1, Human/genetics , Mutation
7.
Gut Microbes ; 14(1): 2096989, 2022.
Article in English | MEDLINE | ID: mdl-35793266

ABSTRACT

Herpes simplex encephalitis (HSE), a complication of herpes simplex virus type I (HSV-1) infection causes neurological disorder or even death in immunocompromised adults and newborns. However, the intrinsic factors controlling the HSE outcome remain unclear. Here, we show that HSE mice exhibit gut microbiota dysbiosis and altered metabolite configuration and tryptophan-nicotinamide metabolism. HSV-1 neurotropic infection activated microglia, with changed immune properties and cell numbers, to stimulate antiviral immune response and contribute substantially to HSE. In addition, depletion of gut microbiota by oral antibiotics (ABX)-treatment triggered the hyper-activation of microglia, which in turn enhanced inflammatory immune response, and cytokine production, resulting in aggregated viral burden and HSE pathology. Furthermore, exogenous administration of nicotinamide n-oxide (NAMO), an oxidative product of nicotinamide derived from gut microbiota, to ABX-treated or untreated HSE mice significantly diminished microglia-mediated proinflammatory response and limited HSV-1 infection in CNS. Mechanistic study revealed that HSV-1 activates microglia by increasing mitochondrial damage via defective mitophagy, whereas microbial metabolite NAMO restores NAD+-dependent mitophagy to inhibit microglia activation and HSE progression. NAMO also prevented neuronal cell death triggered by HSV-1 infection or microglia-mediated microenvironmental toxicity. Finally, we show that NAMO is mainly generated by neomycin-sensitive bacteria, especially Lactobacillus_gasseri and Lactobacillus_reuteri. Together, these data demonstrate that gut microbial metabolites act as intrinsic restrictive factors against HSE progression via regulating mitophagy in microglia, implying further exploration of bacterial or nutritional approaches for treating neurotropic virus-related neurodegenerative diseases.


Subject(s)
Encephalitis, Herpes Simplex , Gastrointestinal Microbiome , Animals , Encephalitis, Herpes Simplex/pathology , Mice , Microglia/metabolism , Mitophagy , Niacinamide/analogs & derivatives , Niacinamide/metabolism
8.
Zhongguo Zhong Yao Za Zhi ; 47(11): 3023-3028, 2022 Jun.
Article in Chinese | MEDLINE | ID: mdl-35718526

ABSTRACT

This study aims to explore the protective effect of Forsythiae Fructus extract(FFE) against herpes simplex virus encephalitis(HSE) in mice. To be specific, life extension rate of mice, viral load in mouse brain, levels of tumor necrosis factor-α(TNF-α), interleukin-1ß(IL-1ß), and interferon-α(IFN-α), and nitric oxide(NO) content in mouse brain were determined. Mice were classified into normal group, model group, acyclovir(ACV) group, and high-dose, medium-dose, and low-dose(100, 50, 25 mg·kg~(-1), respectively) FFE groups. HSE was induced in mice in corresponding groups. Then, the life extension rate was compared among groups. Viral load in brain was detected by real-time fluorescent quantitative PCR, the changes of TNF-α, IL-1ß, and IFN-α in brain by enzyme-linked immunosorbent assay(ELISA), NO content in brain with nitrate reduction method, and pathological changes by hematoxylin-eosin(HE) staining. The result showed that the life extension rate in the high-dose, medium-dose, and low-dose FFE groups was 27.93%, 19.94%, and 10.66%, respectively, and the difference between the high-dose group and the model group was statistically significant(P<0.05). FFE decreased the viral load in brains of HSE mice. The levels of TNF-α, IL-1ß, and IFN-α in ACV group and high-dose and medium-dose FFE groups were lower than those in the model group(P<0.01,P<0.05), and NO content in the three FFE groups was lower than that in the model group(P<0.01). In conclusion, FFE can improve the survival rate of HSE mice, reduce the load of herpes simplex virus type Ⅰ(HSV-1) in the brains of HSE mice, decrease the levels of inflammatory factors and NO content, and alleviate inflammation and pathological damage, thereby protecting the central nervous system.


Subject(s)
Encephalitis, Herpes Simplex , Herpesvirus 1, Human , Acyclovir/pharmacology , Animals , Encephalitis, Herpes Simplex/drug therapy , Encephalitis, Herpes Simplex/pathology , Herpesvirus 1, Human/physiology , Mice , Mice, Inbred BALB C , Nitric Oxide , Plant Extracts/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology
9.
J Med Imaging Radiat Oncol ; 66(5): 609-617, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34519419

ABSTRACT

INTRODUCTION: We aim to assess the imaging manifestations of brain involvement in paediatric immunocompromised patients with haematological malignancies on chemotherapy presenting with encephalitis and positive HSV CSF PCR. We also aim to determine whether our findings are similar or different to those described in literature for paediatric patients in general. METHODS: A retrospective study was performed in paediatric ALL/lymphoma patients on chemotherapy, and cases with positive CSF HSV-PCR with at least one head MRI scan were included. On imaging, location(typical/atypical), post-contrast enhancement and haemorrhage/diffusivity/gliosis were evaluated. RESULT: A total of 28 scans were included in study from 16 patients fulfilling inclusion criteria, 12 (75%) HSV-1 and 4 (25%) HSV-2. Of the 16 initial scans (CT n = 10, MRI n = 6), 11 were normal (CT = 7, MRI = 4). Fourteen patients had follow-up MRI, of which two had normal scans. On the abnormal initial scan (5/16), only 20% had typical medial temporal/inferomedial frontal/cingulate involvement. Most had frontal (80%), parietal (60%) and non-medial temporal (40%) lesions. Abnormal diffusivity/haemorrhage was absent in all, and postcontrast enhancement was seen in 20%. On follow-up, more patients (33%) had typical medial temporal/inferomedial frontal/cingulate involvement. Widespread atypical site involvement of frontoparietal (75%), thalamus (58%), non-medial temporal (50%), occipital/basal ganglia (33%) and cerebellum (8%) was noted. Most lesions were cortical (91%)/subcortical (75%) with few periventricular lesions (58%). Few showed abnormal diffusivity (16%), post-contrast parenchymal enhancement/haemorrhage (8%), post-contrast meningeal enhancement (25%) and gliosis (16%). CONCLUSION: Immunocompromised paediatric patients with haematological malignancies have widespread atypical brain involvement in herpes simplex encephalitis with lack of diffusion restriction and post-contrast enhancement, likely due to haematogenous spread of HSV across the blood-brain barrier, lack of cellular immunity and limited inflammatory cytokine response.


Subject(s)
Encephalitis, Herpes Simplex , Hematologic Neoplasms , Neoplasms , Child , Encephalitis, Herpes Simplex/diagnostic imaging , Encephalitis, Herpes Simplex/drug therapy , Encephalitis, Herpes Simplex/pathology , Gliosis , Humans , Immunocompromised Host , Magnetic Resonance Imaging , Retrospective Studies , Simplexvirus
10.
Brain Pathol ; 32(3): e13031, 2022 05.
Article in English | MEDLINE | ID: mdl-34709694

ABSTRACT

Herpes simplex encephalitis (HSE) is one of the most serious diseases of the nervous system in humans. However, its pathogenesis is still only poorly understood. Although several mouse models of predominantly herpes simplex virus 1 (HSV-1) infections mimic different crucial aspects of HSE, central questions remain unanswered. They comprise the specific temporofrontal tropism, viral spread within the central nervous system (CNS), as well as potential molecular and immunological barriers that drive virus into latency while only rarely resulting in severe HSE. We have recently proposed an alternative mouse model by using a pseudorabies virus (PrV) mutant that more faithfully represents the striking features of human HSE: temporofrontal meningoencephalitis with few severely, but generally only moderately to subclinically affected mice as well as characteristic behavioral abnormalities. Here, we characterized this animal model using 6- to 8-week-old female CD-1 mice in more detail. Long-term investigation over 6 months consistently revealed a biphasic course of infection accompanied by recurring clinical signs including behavioral alterations and mainly mild meningoencephalitis restricted to the temporal and frontal lobes. By histopathological and immunological analyses, we followed the kinetics and spatial distribution of inflammatory lesions as well as the underlying cytokine expression in the CNS over 21 days within the acute phase of infection. Affecting the temporal lobes, the inflammatory infiltrate was composed of lymphocytes and macrophages showing a predominantly lymphocytic shift 15 days after infection. A strong increase was observed in cytokines CXCL10, CCL2, CCL5, and CXCL1 recruiting inflammatory cells to the CNS. Unlike the majority of infected mice, strongly affected animals demonstrated extensive temporal lobe edema, which is typically present in severe human HSE cases. In summary, these results support the validity of our animal model for in-depth investigation of HSE pathogenesis.


Subject(s)
Encephalitis, Herpes Simplex , Meningoencephalitis , Animals , Central Nervous System/pathology , Cytokines , Disease Models, Animal , Encephalitis, Herpes Simplex/diagnosis , Encephalitis, Herpes Simplex/pathology , Female , Humans , Mice , Neuropathology
11.
J Neurovirol ; 27(5): 774-781, 2021 10.
Article in English | MEDLINE | ID: mdl-34546546

ABSTRACT

Herpes simplex encephalitis (HSE) is a very severe infection of the central nervous system (CNS) caused mainly by herpes simplex virus type 1 (HSV-1) and occasionally by herpes simplex virus type 2 (HSV-2). After relapse or drug-resistant to chemotherapy, whole-brain radiation therapy (WBRT) is a mainstay of treatment in patients with both identifiable brain metastases and CNS lymphoma. Although HSV-1 encephalitis predominantly affects immunocompetent host, HSV encephalitis may be more common in immune-suppressed patients than is currently recognized. Disease presentation may be atypical including lack of pleocytosis in cerebrospinal fluid (CSF). We report four patients diagnosed with HSE following chemotherapy and WBRT. The occurrence of HSE in patients with cancer seems not to be increased compared to the general population, but as our case series shows, a high level of suspicion is needed by the treating physician to diagnose HSE early in patients presenting with new neurological symptoms following WBRT.


Subject(s)
Brain Neoplasms , Encephalitis, Herpes Simplex , Herpes Simplex , Herpesvirus 1, Human , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Cranial Irradiation/adverse effects , Encephalitis, Herpes Simplex/pathology , Herpes Simplex/pathology , Humans , Neoplasm Recurrence, Local , Simplexvirus
12.
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
13.
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
14.
J Neurovirol ; 27(4): 601-608, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34398444

ABSTRACT

The pathophysiology of the memory impairment following Herpes Simplex virus encephalitis is not yet established and understood. This study attempts to elucidate the role of white matter injury and its impact on neuropsychological outcome in patients with history of Herpes Simplex virus encephalitis. This is a single-institution prospective study assessing 9 patients and 15 matched controls utilizing a combination of MRI with diffusion tensor imaging and neuropsychological testing. Tract-based spatial statistics analysis was performed and correlated with neuropsychological outcomes. Significantly decreased fractional anisotropy (FA) values were noted in corpus callosum, corona radiata, left posterior thalamic radiation, cingulum, superior longitudinal fasciculus, fornix, inferior longitudinal fasciculus, inferior fronto-occipital fasciculus, and uncinated fasciculus. Impaired facial recognition significantly correlated with reduction in FA of right uncinate fasciculus, right inferior longitudinal fasciculus, and splenium genu of corpus callosum. FA value of left cingulum significantly correlated with logical memory, auditory verbal learning. FA value of fornix correlated with visual recognition; FA value of left uncinate fasciculus with auditory verbal learning and delayed recall. In conclusion, this study demonstrates microstructural abnormalities involving several white matter tracts corresponding to neuropsychological deficits.


Subject(s)
Encephalitis, Herpes Simplex/pathology , Memory Disorders/pathology , White Matter/pathology , Adolescent , Adult , Aged , Diffusion Tensor Imaging/methods , Encephalitis, Herpes Simplex/complications , Encephalitis, Herpes Simplex/diagnostic imaging , Female , Humans , Learning , Male , Memory Disorders/diagnostic imaging , Memory Disorders/virology , Middle Aged , Neuroimaging/methods , White Matter/diagnostic imaging , Young Adult
15.
Am J Hum Genet ; 108(6): 1012-1025, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34015270

ABSTRACT

The human genetic dissection of clinical phenotypes is complicated by genetic heterogeneity. Gene burden approaches that detect genetic signals in case-control studies are underpowered in genetically heterogeneous cohorts. We therefore developed a genome-wide computational method, network-based heterogeneity clustering (NHC), to detect physiological homogeneity in the midst of genetic heterogeneity. Simulation studies showed our method to be capable of systematically converging genes in biological proximity on the background biological interaction network, and capturing gene clusters harboring presumably deleterious variants, in an efficient and unbiased manner. We applied NHC to whole-exome sequencing data from a cohort of 122 individuals with herpes simplex encephalitis (HSE), including 13 individuals with previously published monogenic inborn errors of TLR3-dependent IFN-α/ß immunity. The top gene cluster identified by our approach successfully detected and prioritized all causal variants of five TLR3 pathway genes in the 13 previously reported individuals. This approach also suggested candidate variants of three reported genes and four candidate genes from the same pathway in another ten previously unstudied individuals. TLR3 responsiveness was impaired in dermal fibroblasts from four of the five individuals tested, suggesting that the variants detected were causal for HSE. NHC is, therefore, an effective and unbiased approach for unraveling genetic heterogeneity by detecting physiological homogeneity.


Subject(s)
Computational Biology/methods , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/pathology , Fibroblasts/immunology , Gene Regulatory Networks , Genetic Heterogeneity , Genetic Predisposition to Disease , Case-Control Studies , Encephalitis, Herpes Simplex/immunology , Fibroblasts/metabolism , Humans , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Toll-Like Receptor 3/metabolism , Exome Sequencing
16.
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
17.
Neurol Res ; 43(4): 307-313, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33274693

ABSTRACT

Background and purpose: To investigate the effect of Emodin on the inflammatory response of brain tissue and the expression of the TLR3 pathway in mice with herpes virus encephalitis.Method: Twenty male BALB/c mice were randomly divided into the NS group, HSV-1 group, HSV-1 + Emodin group and HSV-1 + ACV group. The histopathological features and the effect of TLR3 expression were observed by staining and immunohistochemistry (IHC) respectively. The gene expression of TLR3, trif, TRADD, TRAF6, traf3, p38, Nemo and IRF3 was detected by polymerase chain reaction (PCR). The protein production of TLR3 and its downstream molecules was detected by Western blot. The expression of IL-6, TNF-α and IFN-ß in the brain tissues was detected by ELISA.Result: Compared to the HSV-1 group, the pathological changes (inflammatory cell infiltration, necrotic temporal lobe and massive hemorrhage) were not as obvious as those in the HSV-1+emodin and HSV-1+ACV groups. The TLR3 staining increased significantly in the HSV-1 groups and decreased in the HSV-1 + emodin group. Compared with the NS group, the mRNA expression of TLR3, TRIF, TRADD, TRAF6, traf3, p38, NEMO and IRF3 decreased by 20%-60% in the HSV-1 + emodin group and 30% in the HSV-1 + ACV group, respectively. The expression of IL-6, TNF-α and IFN-ß decreased by 30%-50% in the HSV-1 + emodin group and showed no significant change in the HSV-1 + ACV group, respectively.Conclusion: Emodin could inhibit the inflammatory response in the brain of mice with herpes virus encephalitis. The inhibition of TLR3 expression may play an important role in this process.


Subject(s)
Brain/metabolism , Emodin/therapeutic use , Encephalitis, Herpes Simplex/metabolism , Herpes Simplex/metabolism , Herpesvirus 1, Human , Toll-Like Receptor 3/biosynthesis , Animals , Brain/drug effects , Brain/pathology , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Emodin/pharmacology , Encephalitis, Herpes Simplex/drug therapy , Encephalitis, Herpes Simplex/pathology , Herpes Simplex/drug therapy , Herpes Simplex/pathology , Male , Mice , Mice, Inbred BALB C , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 3/antagonists & inhibitors
18.
J Clin Invest ; 131(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-32960813

ABSTRACT

Inborn errors of TLR3-dependent IFN-α/ß- and IFN-λ-mediated immunity in the CNS can underlie herpes simplex virus 1 (HSV-1) encephalitis (HSE). The respective contributions of IFN-α/ß and IFN-λ are unknown. We report a child homozygous for a genomic deletion of the entire coding sequence and part of the 3'-UTR of the last exon of IFNAR1, who died of HSE at the age of 2 years. An older cousin died following vaccination against measles, mumps, and rubella at 12 months of age, and another 17-year-old cousin homozygous for the same variant has had other, less severe, viral illnesses. The encoded IFNAR1 protein is expressed on the cell surface but is truncated and cannot interact with the tyrosine kinase TYK2. The patient's fibroblasts and EBV-B cells did not respond to IFN-α2b or IFN-ß, in terms of STAT1, STAT2, and STAT3 phosphorylation or the genome-wide induction of IFN-stimulated genes. The patient's fibroblasts were susceptible to viruses, including HSV-1, even in the presence of exogenous IFN-α2b or IFN-ß. HSE is therefore a consequence of inherited complete IFNAR1 deficiency. This viral disease occurred in natural conditions, unlike those previously reported in other patients with IFNAR1 or IFNAR2 deficiency. This experiment of nature indicates that IFN-α/ß are essential for anti-HSV-1 immunity in the CNS.


Subject(s)
Encephalitis, Herpes Simplex , Herpesvirus 1, Human/metabolism , Receptor, Interferon alpha-beta/deficiency , Adolescent , Child, Preschool , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/metabolism , Encephalitis, Herpes Simplex/pathology , HEK293 Cells , Herpesvirus 1, Human/genetics , Humans , Interferons/genetics , Interferons/metabolism , Male , Receptor, Interferon alpha-beta/metabolism
19.
Rinsho Shinkeigaku ; 60(12): 840-845, 2020 Dec 26.
Article in Japanese | MEDLINE | ID: mdl-33229829

ABSTRACT

An 89-year-old man was admitted because of persistent fever and impaired consciousness. On admission, his consciousness level was E3V3M4 according to the Glasgow Coma Scale. MRI of the brain showed high intensity lesions in the bilateral cingulate gyri. In the cerebrospinal fluid, both cell counts and glucose level were in the normal ranges. He had received antibiotics and intravenous isotonic saline. On the fifth day of hospitalization, blood examination revealed elevation of anti-herpes simplex virus (HSV) immunoglobulin M antibody, and herpes simplex encephalitis (HSE) was diagnosed. Despite treatment with acyclovir, his respiratory function and consciousness level deteriorated rapidly. On the eighth day, he died of respiratory failure. At autopsy, the brain showed multiple softenings of the gray and white matter in the hippocampus, amygdala, and temporal, insular, and cingulate cortices. Some of these lesions were hemorrhagic. Microscopic examination revealed that the lesions were necrotic and associated with perivascular inflammatory cell infiltration in the limbic system, hypothalamus, brainstem tegmentum area, and medulla. Eosinophilic intranuclear inclusions were rarely found in the astrocytes in the medulla. Immunohistochemistry revealed anti-HSV-1 antibody positive neurons in the brainstem tegmentum including reticular formation and the raphe nuclei. HSV-DNA was also detected in the postmortem cerebrospinal fluid. This was a rare case of HSE in which inflammation in the brainstem proved to be the cause of lethal respiratory failure.


Subject(s)
Brain Stem/pathology , Encephalitis, Herpes Simplex/complications , Encephalitis, Herpes Simplex/pathology , Respiratory Insufficiency/etiology , Respiratory Insufficiency/pathology , Acute Disease , Acyclovir/administration & dosage , Age of Onset , Aged, 80 and over , Antibodies, Viral/blood , Autopsy , Biomarkers/blood , Brain Stem/diagnostic imaging , Diffusion Magnetic Resonance Imaging , Encephalitis, Herpes Simplex/diagnosis , Encephalitis, Herpes Simplex/drug therapy , Fatal Outcome , Humans , Immunoglobulin M/blood , Male , Simplexvirus/immunology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...