Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
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
2.
J Immunol ; 207(7): 1824-1835, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34470854

ABSTRACT

This report evaluates how HSV enters the brain to cause herpes simplex encephalitis following infection at a peripheral site. We demonstrate that encephalitis regularly occurred when BALB/c mice were infected with HSV and treated daily with 2-deoxy-d-glucose (2DG), which inhibits glucose use via the glycolysis pathway. The outcome of infection in the trigeminal ganglion (TG), the site to which the virus spreads, replicates, and establishes latency, showed marked differences in viral and cellular events between treated and untreated animals. In control-untreated mice, the replicating virus was present only during early time points, whereas in 2DG recipients, replicating virus remained for the 9-d observation period. This outcome correlated with significantly reduced numbers of innate inflammatory cells as well as T cells in 2DG-treated animals. Moreover, T cells in the TG of treated animals were less activated and contained a smaller fraction of expressed IFN-γ production compared with untreated controls. The breakdown of latency was accelerated when cultures of TG cells taken from mice with established HSV latency were cultured in the presence of 2DG. Taken together, the results of both in vivo and in vitro investigations demonstrate that the overall effects of 2DG therapy impaired the protective effects of one or more inflammatory cell types in the TG that normally function to control productive infection and prevent spread of virus to the brain.


Subject(s)
Brain/pathology , Encephalitis, Herpes Simplex/metabolism , Glucose/metabolism , Simplexvirus/physiology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Deoxyglucose/administration & dosage , Humans , Immunity, Innate , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Virus Latency
3.
Front Immunol ; 12: 714821, 2021.
Article in English | MEDLINE | ID: mdl-34526992

ABSTRACT

The Fas/FasL pathway plays a key role in immune homeostasis and immune surveillance. In the central nervous system (CNS) Fas/FasL is involved in axonal outgrowth and adult neurogenesis. However, little is known about the role of the Fas/FasL pathway in herpes encephalitis. In this study, we used a neuropathogenic clinical strain of herpes simplex virus type 1 (HSV-1) to explore infection-induced inflammation and immune responses in the mouse brain and the role of Fas/FasL in antiviral CNS immunity. HSV-1 CNS infection induced the infiltration of Fas- FasL-bearing monocytes and T cells in the brain and also to an up-regulation of Fas and FasL expression on resident astrocytes and microglia within infected sites. Upon infection, Fas- and FasL-deficient mice (lpr and gld) were partially protected from encephalitis with a decreased morbidity and mortality compared to WT mice. Fas/FasL deficiency promoted cell-mediated immunity within the CNS. Fas receptor stimulation abrogated HSV-1 induced activation and inflammatory reactions in microglia from WT mice, while lack of Fas or FasL led to a more pronounced activation of monocytes and microglia and also to an enhanced differentiation of these cells into a pro-inflammatory M1 phenotype. Furthermore, the specific immune system was more efficient in Fas- and FasL-deficient mice with significantly higher numbers of infiltrating HSV-1-specific cytotoxic T cells in the brain. Our data indicate that the Fas/FasL pathway leads to excessive neuroinflammation during HSV-1 infection, which is associated with a diminished anti-viral response and an excessive neuroinflammation.


Subject(s)
Encephalitis, Herpes Simplex/etiology , Encephalitis, Herpes Simplex/metabolism , Fas Ligand Protein/metabolism , Herpesvirus 1, Human/physiology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , fas Receptor/metabolism , Animals , Biomarkers , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility/immunology , Encephalitis, Herpes Simplex/diagnosis , Fas Ligand Protein/genetics , Humans , Mice , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Neuroinflammatory Diseases/diagnosis , fas Receptor/genetics
4.
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
5.
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
6.
J Virol ; 94(23)2020 11 09.
Article in English | MEDLINE | ID: mdl-32938766

ABSTRACT

The role of a signaling pathway through macrophage colony-stimulating factor (MCSF) and its receptor, macrophage colony-stimulating factor 1 receptor (CSF1R), during experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE) was studied by two different approaches. First, we evaluated the effect of stimulation of the MCSF/CSF1R axis before infection. Exogenous MCSF (40 µg/kg of body weight intraperitoneally [i.p.]) was administered once daily to BALB/c mice on days 4 and 2 before intranasal infection with 2,500 PFU of HSV-1. MCSF treatment significantly increased mouse survival compared to saline (50% versus 10%; P = 0.0169). On day 6 postinfection (p.i.), brain viral titers were significantly decreased, whereas beta interferon (IFN-ß) was significantly increased in mice treated with MCSF compared to mice treated with saline. The number of CD68+ (a phagocytosis marker) microglial cells was significantly increased in MCSF-treated mice compared to the saline-treated group. Secondly, we conditionally depleted CSF1R on microglial cells of CSF1R-loxP-CX3CR1-cre/ERT2 mice (in a C57BL/6 background) through induction with tamoxifen. The mice were then infected intranasally with 600,000 PFU of HSV-1. The survival rate of mice depleted of CSF1R (knockout [KO] mice) was significantly lower than that of wild-type (WT) mice (0% versus 67%). Brain viral titers and cytokine/chemokine levels were significantly higher in KO than in WT animals on day 6 p.i. Furthermore, increased infiltration of monocytes into the brains of WT mice was seen on day 6 p.i., but not in KO mice. Our results suggest that microglial cells are essential to control HSE at early stages of the disease and that the MCSF/CSF1R axis could be a therapeutic target to regulate their response to infection.IMPORTANCE Microglia appear to be one of the principal regulators of neuroinflammation in the central nervous system (CNS). An increasing number of studies have demonstrated that the activation of microglia could result in either beneficial or detrimental effects in different CNS disorders. Hence, the role of microglia during herpes simplex virus encephalitis (HSE) has not been fully characterized. Using experimental mouse models, we showed that an early activation of the MCSF/CSF1R axis improved the outcome of the disease, possibly by inducing a proliferation of microglia. In contrast, depletion of microglia before HSV-1 infection worsened the prognosis of HSE. Thus, an early microglial response followed by sustained infiltration of monocytes and T cells into the brain seem to be key components for a better clinical outcome. These data suggest that microglia could be a potential target for immunomodulatory strategies combined with antiviral therapy to better control the outcome of this devastating disease.


Subject(s)
Encephalitis, Herpes Simplex/metabolism , Herpesvirus 1, Human/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Microglia/metabolism , Microglia/virology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Brain/virology , Central Nervous System/metabolism , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Macrophage Colony-Stimulating Factor/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Phagocytosis , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Viral Load
7.
Neurosci Lett ; 736: 135295, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32800922

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is the main etiological agent of acute and sporadic encephalitis. Proteins of the suppressor of cytokine signaling (SOCS) family have shown to regulate the inflammation during HSV-1 infection in the brain. However, the effects of SOCS2 and SOCS3 in viral encephalitis remain unclear. The aim of the current study is to investigate the potential association between SOCS2, SOCS3, cytokines, and hippocampal damage, especially neuronal apoptosis, during acute intracranial HSV-1 infection in mice. Male C57BL/6 mice were infected by intracranial route with 102 plaque-forming units (PFU) inoculum of purified HSV-1. At three days post-infection (3 d.p.i.), mice were euthanized and their hippocampi were collected for histopathological analysis, immunohistochemical reaction against active caspase-3 and quantification of SOCS2, SOCS3 and cytokines (tumoral necrosis factor (TNF), interleukin (IL) 1ß, IL-6, IL-10; interferon (IFN) -α, IFN-ß, IFN-γ) mRNA expression. Infected mice exhibited neuronal loss and hemorrhagic focus in Cornu Ammonis (CA) region. The apoptotic index was higher in infected mice compared to controls. HSV-1 infection was associated with increased hippocampal expression of TNF, IL1-ß, IL-6 and IFNα/IFNß and decreased expression of IL-10, IFN-γ, SOCS2 and SOCS3. Our results suggest that down regulation of SOCS2 and SOCS3 contributes to a pro-inflammatory environment associated with hippocampal damage and neuronal apoptosis during acute HSV-1 infection in mice.


Subject(s)
Encephalitis, Herpes Simplex/metabolism , Hippocampus/virology , Inflammation/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Apoptosis/physiology , Chlorocebus aethiops , Cytokines/metabolism , Hippocampus/metabolism , Male , Mice , Neurons/metabolism , Neurons/virology , Vero Cells
8.
Brain Behav Immun ; 89: 480-490, 2020 10.
Article in English | MEDLINE | ID: mdl-32717399

ABSTRACT

The incidence of infectious diseases affecting the central nervous system (CNS) has been increasing over the last several years. Among the reasons for the expansion of these diseases and the appearance of new neuropathogens are globalization, global warming, and the increased proximity between humans and wild animals due to human activities such as deforestation. Neurotropism affecting normal brain function is shared by organisms such as viruses, bacteria, fungi, and parasites. Neuroinfections caused by these agents activate immune responses, inducing neuroinflammation, excitotoxicity, and neurodegeneration. Purinergic signaling is an evolutionarily conserved signaling pathway associated with these neuropathologies. During neuroinfections, host cells release ATP as an extracellular danger signal with pro-inflammatory activities. ATP is metabolized to its derivatives by ectonucleotidases such as CD39 and CD73; ATP and its metabolites modulate neuronal and immune mechanisms through P1 and P2 purinergic receptors that are involved in pathophysiological mechanisms of neuroinfections. In this review we discuss the beneficial or deleterious effects of various components of the purinergic signaling pathway in infectious diseases that affect the CNS, including human immunodeficiency virus (HIV-1) infection, herpes simplex virus type 1 (HSV-1) infection, bacterial meningitis, sepsis, cryptococcosis, toxoplasmosis, and malaria. We also provide a description of this signaling pathway in emerging viral infections with neurological implications such as Zika and SARS-CoV-2.


Subject(s)
Central Nervous System Infections/metabolism , Receptors, Purinergic P1/metabolism , Receptors, Purinergic P2X/metabolism , Receptors, Purinergic P2Y/metabolism , AIDS Dementia Complex/metabolism , Betacoronavirus , COVID-19 , Coronavirus Infections/metabolism , Encephalitis, Herpes Simplex/metabolism , Humans , Malaria/metabolism , Meningitis, Bacterial/metabolism , Meningitis, Cryptococcal/metabolism , Pandemics , Pneumonia, Viral/metabolism , SARS-CoV-2 , Sepsis/metabolism , Signal Transduction , Toxoplasmosis, Cerebral/metabolism , Zika Virus Infection/metabolism
9.
PLoS Pathog ; 15(12): e1008168, 2019 12.
Article in English | MEDLINE | ID: mdl-31869396

ABSTRACT

We report here two cases of Herpes simplex virus encephalitis (HSE) in adult patients with very rare, previously uncharacterized, non synonymous heterozygous G634R and R203W substitution in mannan-binding lectin serine protease 2 (MASP2), a gene encoding a key protease of the lectin pathway of the complement system. None of the 2 patients had variants in genes involved in the TLR3-interferon signaling pathway. Both MASP2 variants induced functional defects in vitro, including a reduced (R203W) or abolished (G634R) protein secretion, a lost capability to cleave MASP-2 precursor into its active form (G634R) and an in vivo reduced antiviral activity (G634R). In a murine model of HSE, animals deficient in mannose binding lectins (MBL, the main pattern recognition molecule associated with MASP-2) had a decreased survival rate and an increased brain burden of HSV-1 compared to WT C57BL/6J mice. Altogether, these data suggest that MASP-2 deficiency can increase susceptibility to adult HSE.


Subject(s)
Encephalitis, Herpes Simplex/metabolism , Mannose-Binding Protein-Associated Serine Proteases/deficiency , Adult , Animals , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/immunology , Humans , Immunity, Innate/genetics , Lectins/genetics , Lectins/metabolism , Male , Mannose-Binding Lectin/metabolism , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mannose-Binding Protein-Associated Serine Proteases/immunology , Mice, Inbred C57BL , Mice, Transgenic
10.
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
12.
Neuron ; 99(1): 56-63.e3, 2018 07 11.
Article in English | MEDLINE | ID: mdl-30001512

ABSTRACT

Amyloid-ß peptide (Aß) fibrilization and deposition as ß-amyloid are hallmarks of Alzheimer's disease (AD) pathology. We recently reported Aß is an innate immune protein that protects against fungal and bacterial infections. Fibrilization pathways mediate Aß antimicrobial activities. Thus, infection can seed and dramatically accelerate ß-amyloid deposition. Here, we show Aß oligomers bind herpesvirus surface glycoproteins, accelerating ß-amyloid deposition and leading to protective viral entrapment activity in 5XFAD mouse and 3D human neural cell culture infection models against neurotropic herpes simplex virus 1 (HSV1) and human herpesvirus 6A and B. Herpesviridae are linked to AD, but it has been unclear how viruses may induce ß-amyloidosis in brain. These data support the notion that Aß might play a protective role in CNS innate immunity, and suggest an AD etiological mechanism in which herpesviridae infection may directly promote Aß amyloidosis.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloidosis/metabolism , Brain/metabolism , Encephalitis, Viral/metabolism , Herpesviridae , Alzheimer Disease/virology , Amyloidosis/virology , Animals , Brain/virology , Cells, Cultured , Disease Models, Animal , Encephalitis, Herpes Simplex/metabolism , Encephalitis, Herpes Simplex/virology , Encephalitis, Viral/virology , Herpesvirus 1, Human , Herpesvirus 6, Human , Humans , Mice , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Neurons , Plaque, Amyloid/metabolism , Roseolovirus Infections/metabolism , Roseolovirus Infections/virology
13.
J Neuroimmunol ; 316: 65-73, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29305044

ABSTRACT

Herpes simplex encephalitis (HSE) is a severe neurological disease in children and adults caused by herpes simplex virus. This review discusses recent findings on the role of Toll-like receptor 3 (TLR3) deficiencies in the HSE development. Critical checkpoints in the TLR3 signaling that contribute to innate response are discussed, including the importance of TLR3 ligand recognition site and transportation in the cell. We also indicate unresolved issues in the TLR3 functioning that might lead to thorough understanding of immunity during HSE. Such a knowledge base will lead to discovery and design of a rationale therapeutic and preventive approach against HSE.


Subject(s)
Encephalitis, Herpes Simplex/immunology , Immunity, Innate/immunology , Signal Transduction/immunology , Toll-Like Receptor 3/immunology , Encephalitis, Herpes Simplex/metabolism , Humans , Toll-Like Receptor 3/metabolism
14.
Front Immunol ; 9: 3047, 2018.
Article in English | MEDLINE | ID: mdl-30671054

ABSTRACT

The induction and action of type I interferon (IFN) is of fundamental importance in human immune defenses toward microbial pathogens, particularly viruses. Basic discoveries within the molecular and cellular signaling pathways regulating type I IFN induction and downstream actions have shown the essential role of the IFN regulatory factor (IRF) and the signal transducer and activator of transcription (STAT) families, respectively. However, the exact biological and immunological functions of these factors have been most clearly revealed through the study of inborn errors of immunity and the resultant infectious phenotypes in humans. The spectrum of human inborn errors of immunity caused by mutations in IRFs and STATs has proven very diverse. These diseases encompass herpes simplex encephalitis (HSE) and severe influenza in IRF3- and IRF7/IRF9 deficiency, respectively. They also include Mendelian susceptibility to mycobacterial infection (MSMD) in STAT1 deficiency, through disseminated measles infection associated with STAT2 deficiency, and finally staphylococcal abscesses and chronic mucocutaneous candidiasis (CMC) classically described with Hyper-IgE syndrome (HIES) in the case of STAT3 deficiency. More recently, increasing focus has been on aspects of autoimmunity and autoinflammation playing an important part in many primary immunodeficiency diseases (PID)s, as exemplified by STAT1 gain-of-function causing CMC and autoimmune thyroiditis, as well as a recently described autoinflammatory syndrome with hypogammaglobulinemia and lymphoproliferation as a result of STAT3 gain-of-function. Here I review the infectious, inflammatory, and autoimmune disorders arising from mutations in IRF and STAT transcription factors in humans, highlightning the underlying molecular mechanisms and immunopathogenesis as well as the clinical/therapeutic perspectives of these new insights.


Subject(s)
Candidiasis, Chronic Mucocutaneous/metabolism , Encephalitis, Herpes Simplex/metabolism , Influenza, Human/metabolism , Interferon Regulatory Factors/metabolism , Job Syndrome/metabolism , Mycobacterium Infections/metabolism , STAT Transcription Factors/metabolism , Autoimmunity , Candidiasis, Chronic Mucocutaneous/genetics , Candidiasis, Chronic Mucocutaneous/immunology , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/immunology , Humans , Immunity, Innate , Influenza, Human/genetics , Influenza, Human/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Janus Kinases/metabolism , Job Syndrome/genetics , Job Syndrome/immunology , Mutation , Mycobacterium Infections/genetics , Mycobacterium Infections/immunology , Receptor, Interferon alpha-beta/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology
15.
J Pharm Pract ; 30(3): 324-328, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27067742

ABSTRACT

BACKGROUND: Dosing of intravenous acyclovir for herpes encephalitis in obese patients is recommended to be based on ideal body weight. However, limited data support this recommendation, and recent data suggest this may lead to underdosing. OBJECTIVE: To determine national dosing practices of intravenous acyclovir across a range of patient weights. METHODS: A survey was distributed to members of the American College of Clinical Pharmacy Critical Care and Infectious Diseases Practice & Research Networks listservs. Data collected included demographic information and dosing of acyclovir, given consistent patient cases with varying patient weight. RESULTS: A total of 264 pharmacists participated in the survey, with 240 (90.9%) participants completing the survey. Participants were predominately clinical pharmacists. As patient weight increased, respondents were more apt to dose based on an adjusted body weight, with dosing in the obese and morbidly obese showing a clear lack of consistency. CONCLUSIONS: Intravenous dosing of acyclovir for herpes encephalitis is variable, especially in obese patients, and does not reflect recommendations. Limited data provide conflicting recommendations for dosing in obese patients, and future studies are necessary to optimize patient outcomes and prevent toxicity.


Subject(s)
Acyclovir/administration & dosage , Antiviral Agents/administration & dosage , Encephalitis, Herpes Simplex/drug therapy , Obesity/drug therapy , Pharmacists , Surveys and Questionnaires , Acyclovir/pharmacokinetics , Administration, Intravenous , Antiviral Agents/pharmacokinetics , Body Weight/drug effects , Body Weight/physiology , Encephalitis, Herpes Simplex/epidemiology , Encephalitis, Herpes Simplex/metabolism , Female , Humans , Male , Obesity/epidemiology , Obesity/metabolism
16.
J Virol ; 90(15): 6738-6745, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27170756

ABSTRACT

UNLABELLED: p53 is a critical host cell factor in the cellular response to a broad range of stress factors. We recently reported that p53 is required for efficient herpes simplex virus 1 (HSV-1) replication in cell culture. However, a defined role for p53 in HSV-1 replication and pathogenesis in vivo remains elusive. In this study, we examined the effects of p53 on HSV-1 infection in vivo using p53-deficient mice. Following intracranial inoculation, p53 knockout reduced viral replication in the brains of mice and led to significantly reduced rates of mortality due to herpes simplex encephalitis. These results suggest that p53 is an important host cell regulator of HSV-1 replication and pathogenesis in the central nervous system (CNS). IMPORTANCE: HSV-1 causes sporadic cases of encephalitis, which, even with antiviral therapy, can result in severe neurological defects and even death. Many host cell factors involved in the regulation of CNS HSV-1 infection have been investigated using genetically modified mice. However, most of these factors are immunological regulators and act via immunological pathways in order to restrict CNS HSV-1 infection. They therefore provide limited information on intrinsic host cell regulators that may be involved in the facilitation of CNS HSV-1 infection. Here we demonstrate that a host cell protein, p53, which has generally been considered a host cell restriction factor for various viral infections, is required for efficient HSV-1 replication and pathogenesis in the CNS of mice. This is the first report showing that p53 positively regulates viral replication and pathogenesis in vivo and provides insights into its molecular mechanism, which may suggest novel clinical treatment options for herpes simplex encephalitis.


Subject(s)
Brain/pathology , Encephalitis, Herpes Simplex/metabolism , Encephalitis, Herpes Simplex/mortality , Simplexvirus/pathogenicity , Tumor Suppressor Protein p53/physiology , Virus Replication , Animals , Brain/metabolism , Brain/virology , Encephalitis, Herpes Simplex/pathology , Encephalitis, Herpes Simplex/virology , Female , Mice , Mice, Inbred ICR , Mice, Knockout , Survival Rate
17.
Trends Mol Med ; 22(6): 511-527, 2016 06.
Article in English | MEDLINE | ID: mdl-27211305

ABSTRACT

The importance of TANK binding kinase-1 (TBK1), a multimeric kinase that modulates inflammation and autophagy, in human health has been highlighted for the first time by the recent discoveries of mutations in TBK1 that underlie amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), normal tension glaucoma (NTG) or childhood herpes simplex encephalitis (HSE). Gain-of-function of TBK1 are associated with NTG, whereas loss-of-function mutations result in ALS/FTD or in HSE. In light of these new findings, we review the role of TBK1 in these seemingly unrelated, yet allelic diseases, and discuss the role of TBK1 in neuroinflammatory diseases. This discovery has the potential to significantly increase our understanding of the molecular basis of these poorly understood diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Central Nervous System/metabolism , Encephalitis, Herpes Simplex/metabolism , Frontotemporal Dementia/metabolism , Inflammation/metabolism , Low Tension Glaucoma/metabolism , Protein Serine-Threonine Kinases/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/immunology , Autophagy , Disease Progression , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/immunology , Frontotemporal Dementia/genetics , Gene Expression , Humans , Interferons/immunology , Interferons/metabolism , Low Tension Glaucoma/genetics , Low Tension Glaucoma/immunology , Mutation , Protein Serine-Threonine Kinases/genetics
18.
J Nucl Med ; 57(5): 785-91, 2016 May.
Article in English | MEDLINE | ID: mdl-26823567

ABSTRACT

(11)C-PBR28 is a second-generation translocator protein (TSPO) tracer with characteristics supposedly superior to the most commonly used tracer for neuroinflammation, (R)-(11)C-PK11195. Despite its use in clinical research, no studies on the imaging properties and pharmacokinetic analysis of (11)C-PBR28 in rodent models of neuroinflammation have been published yet. Therefore, this study aimed to evaluate (11)C-PBR28 as a tool for detection and quantification of neuroinflammation in preclinical research and to compare its imaging properties with (R)-(11)C-PK11195. The herpes simplex encephalitis (HSE) model was used for induction of neuroinflammation in male Wistar rats. Six or 7 d after virus inoculation, a dynamic (11)C-PBR28 or (R)-(11)C-PK11195 PET scan with arterial blood sampling was obtained. Pharmacokinetic modeling was performed on the PET data and analyzed using volumes of interest and a voxel-based approach. Volume-of-interest- and voxel-based analysis of (11)C-PBR28 images showed overexpression of TSPO in brain regions known to be affected in the HSE rat model. (11)C-PBR28 was metabolized faster than (R)-(11)C-PK11195, with a metabolic half-life in plasma of 5 and 21 min, respectively. Overall, (11)C-PBR28 was more sensitive than (R)-(11)C-PK11195 in detecting neuroinflammation. The binding potential (BPND) of (11)C-PBR28 was significantly higher (P < 0.05) in the medulla (176%), pons (146%), midbrain (101%), hippocampus (85%), thalamus (73%), cerebellum (54%), and hypothalamus (49%) in HSE rats than in control rats, whereas (R)-(11)C-PK11195 showed a higher BPND only in the medulla (32%). The BPND in control animals was not significantly different between tracers, suggesting that the nonspecific binding of both tracers is similar. (11)C-PBR28 was more sensitive than (R)-(11)C-PK11195 in the detection of TSPO overexpression in the HSE rat model, because more brain regions with significantly increased tracer uptake could be found, irrespective of the data analysis method used. These results suggest that (11)C-PBR28 should be able to detect more subtle changes in microglial activation in preclinical models of neuroinflammation.


Subject(s)
Encephalitis, Herpes Simplex/metabolism , Isoquinolines/pharmacokinetics , Pyrimidines/pharmacokinetics , Animals , Biological Transport , Disease Models, Animal , Encephalitis, Herpes Simplex/diagnostic imaging , Image Processing, Computer-Assisted , Isoquinolines/metabolism , Kinetics , Male , Positron-Emission Tomography , Pyrimidines/metabolism , Rats , Rats, Wistar
19.
J Neurovirol ; 22(3): 293-306, 2016 06.
Article in English | MEDLINE | ID: mdl-26501779

ABSTRACT

Herpes Simplex Virus type I (HSV-1) latently infects peripheral nervous system (PNS) sensory neurons, and its reactivation leads to recurring cold sores. The reactivated HSV-1 can travel retrograde from the PNS into the central nervous system (CNS) and is known to be causative of Herpes Simplex viral encephalitis. HSV-1 infection in the PNS is well documented, but little is known on the fate of HSV-1 once it enters the CNS. In the murine model, HSV-1 genome persists in the CNS once infected through an ocular route. To gain more details of HSV-1 infection in the CNS, we characterized HSV-1 infection of the tree shrew (Tupaia belangeri chinensis) brain following ocular inoculation. Here, we report that HSV-1 enters the tree shrew brain following ocular inoculation and HSV-1 transcripts, ICP0, ICP4, and LAT can be detected at 5 days post-infection (p.i.), peaking at 10 days p.i. After 2 weeks, ICP4 and ICP0 transcripts are reduced to a basal level, but the LAT intron region continues to be expressed. Live virus could be recovered from the olfactory bulb and brain stem tissue. Viral proteins could be detected using anti-HSV-1 antibodies and anti-ICP4 antibody, during the acute stage but not beyond. In situ hybridization could detect LAT during acute infection in most brain regions and in olfactory bulb and brain stem tissue well beyond the acute stage. Using a homogenate from these tissues' post-acute infection, we did not recover live HSV-1 virus, supporting a latent infection, but using a modified explant cocultivation technique, we were able to recover reactivated virus from these tissues, suggesting that the HSV-1 virus latently infects the tree shrew CNS. Compared to mouse, the CNS acute infection of the tree shrew is delayed and the olfactory bulb contains most latent virus. During the acute stage, a portion of the infected tree shrews exhibit symptoms similar to human viral encephalitis. These findings, together with the fact that tree shrews are closely related to primates, provided a valuable alternative model to study HSV-1 infection and pathogenesis in the CNS.


Subject(s)
Encephalitis, Herpes Simplex/virology , Gene Expression Regulation, Viral , Herpesvirus 1, Human/genetics , Trigeminal Ganglion/virology , Virus Activation , Virus Latency , Animals , Brain Stem/metabolism , Brain Stem/pathology , Brain Stem/virology , Disease Models, Animal , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/metabolism , Encephalitis, Herpes Simplex/pathology , Female , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/pathogenicity , Host Specificity , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/metabolism , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Olfactory Bulb/virology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Culture Techniques , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/pathology , Tupaiidae , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
J Exp Med ; 212(9): 1371-9, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26216125

ABSTRACT

Herpes simplex encephalitis (HSE) in children has previously been linked to defects in type I interferon (IFN) production downstream of Toll-like receptor 3. Here, we describe a novel genetic etiology of HSE by identifying a heterozygous loss-of-function mutation in the IFN regulatory factor 3 (IRF3) gene, leading to autosomal dominant (AD) IRF3 deficiency by haploinsufficiency, in an adolescent female patient with HSE. IRF3 is activated by most pattern recognition receptors recognizing viral infections and plays an essential role in induction of type I IFN. The identified IRF3 R285Q amino acid substitution results in impaired IFN responses to HSV-1 infection and particularly impairs signaling through the TLR3-TRIF pathway. In addition, the R285Q mutant of IRF3 fails to become phosphorylated at S386 and undergo dimerization, and thus has impaired ability to activate transcription. Finally, transduction with WT IRF3 rescues the ability of patient fibroblasts to express IFN in response to HSV-1 infection. The identification of IRF3 deficiency in HSE provides the first description of a defect in an IFN-regulating transcription factor conferring increased susceptibility to a viral infection in the CNS in humans.


Subject(s)
Encephalitis, Herpes Simplex/genetics , Fibroblasts/metabolism , Haploinsufficiency , Herpesvirus 1, Human/metabolism , Interferon Regulatory Factor-3/deficiency , Mutation, Missense , Adolescent , Amino Acid Substitution , Encephalitis, Herpes Simplex/metabolism , Encephalitis, Herpes Simplex/pathology , Female , Fibroblasts/pathology , Fibroblasts/virology , Herpesvirus 1, Human/genetics , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/biosynthesis , Interferon Type I/genetics , Phosphorylation , Protein Multimerization/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...