Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.117
Filter
1.
Mol Biomed ; 5(1): 35, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39207577

ABSTRACT

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


Subject(s)
Herpes Simplex , Vaccine Development , Humans , Herpes Simplex/immunology , Herpes Simplex/prevention & control , Herpes Simplex/virology , Animals , Simplexvirus/pathogenicity , Simplexvirus/immunology , Simplexvirus/physiology , Herpes Simplex Virus Vaccines/immunology , Immune Evasion
2.
J Immunother Cancer ; 12(5)2024 May 31.
Article in English | MEDLINE | ID: mdl-38821716

ABSTRACT

Cytokines are small proteins that regulate the growth and functional activity of immune cells, and several have been approved for cancer therapy. Oncolytic viruses are agents that mediate antitumor activity by directly killing tumor cells and inducing immune responses. Talimogene laherparepvec is an oncolytic herpes simplex virus type 1 (oHSV), approved for the treatment of recurrent melanoma, and the virus encodes the human cytokine, granulocyte-macrophage colony-stimulating factor (GM-CSF). A significant advantage of oncolytic viruses is the ability to deliver therapeutic payloads to the tumor site that can help drive antitumor immunity. While cytokines are especially interesting as payloads, the optimal cytokine(s) used in oncolytic viruses remains controversial. In this review, we highlight preliminary data with several cytokines and chemokines, including GM-CSF, interleukin 12, FMS-like tyrosine kinase 3 ligand, tumor necrosis factor α, interleukin 2, interleukin 15, interleukin 18, chemokine (C-C motif) ligand 2, chemokine (C-C motif) ligand 5, chemokine (C-X-C motif) ligand 4, or their combinations, and show how these payloads can further enhance the antitumor immunity of oHSV. A better understanding of cytokine delivery by oHSV can help improve clinical benefit from oncolytic virus immunotherapy in patients with cancer.


Subject(s)
Cytokines , Immunotherapy , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Oncolytic Viruses/genetics , Cytokines/metabolism , Immunotherapy/methods , Neoplasms/therapy , Neoplasms/immunology , Animals , Simplexvirus/immunology , Simplexvirus/genetics , Herpesvirus 1, Human/immunology
4.
Cell Rep Med ; 5(5): 101528, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38677283

ABSTRACT

Stimulator of interferon genes (STING)-dependent signaling is requisite for effective anti-microbial and anti-tumor activity. STING signaling is commonly defective in cancer cells, which enables tumor cells to evade the immunosurveillance system. We evaluate here whether intrinsic STING signaling in such tumor cells could be reconstituted by creating recombinant herpes simplex viruses (rHSVs) that express components of the STING signaling pathway. We observe that rHSVs expressing STING and/or cGAS replicate inefficiently yet retain in vivo anti-tumor activity, independent of oncolytic activity requisite on the trans-activation of extrinsic STING signaling in phagocytes by engulfed microbial dsDNA species. Accordingly, the in vivo effects of virotherapy could be simulated by nanoparticles incorporating non-coding dsDNA species, which comparably elicit the trans-activation of phagocytes and augment the efficacy of established cancer treatments including checkpoint inhibition and radiation therapy. Our results help elucidate mechanisms of virotherapeutic anti-tumor activity as well as provide alternate strategies to treat cancer.


Subject(s)
DNA , Phagocytes , Animals , Phagocytes/immunology , Phagocytes/metabolism , Humans , Mice , DNA/metabolism , DNA/immunology , DNA/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Signal Transduction , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Cell Line, Tumor , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Neoplasms/genetics , Simplexvirus/genetics , Simplexvirus/immunology , Mice, Inbred C57BL , Oncolytic Virotherapy/methods
6.
Viruses ; 14(1)2022 01 05.
Article in English | MEDLINE | ID: mdl-35062295

ABSTRACT

Primate simplex viruses, including Herpes simplex viruses 1 and 2, form a group of closely related herpesviruses, which establish latent infections in neurons of their respective host species. While neuropathogenic infections in their natural hosts are rare, zoonotic transmission of Macacine alphaherpesvirus 1 (McHV1) from macaques to humans is associated with severe disease. Human infections with baboon-derived Papiine alphaherpesvirus 2 (PaHV2) have not been reported, although PaHV2 and McHV1 share several biological properties, including neuropathogenicity in mice. The reasons for potential differences in PaHV2 and McHV1 pathogenicity are presently not understood, and answering these questions will require mutagenic analysis. Here, we report the development of a recombinant system, which allows rescue of recombinant PaHV2. In addition, we used recombineering to generate viruses carrying reporter genes (Gaussia luciferase or enhanced green fluorescent protein), which replicate with similar efficiency as wild-type PaHV2. We demonstrate that these viruses can be used to analyze susceptibility of cells to infection and inhibition of infection by neutralizing antibodies and antiviral compounds. In summary, we created a recombinant system for PaHV2, which in the future will be invaluable for molecular analyses of neuropathogenicity of PaHV2.


Subject(s)
Cloning, Molecular , Genome, Viral , Recombination, Genetic , Simplexvirus/genetics , Animals , Antibodies, Viral/immunology , Antiviral Agents/pharmacology , Cell Line , Genes, Reporter , Humans , Papio/immunology , Simplexvirus/immunology , Simplexvirus/pathogenicity , Simplexvirus/physiology
7.
Sex Transm Infect ; 98(1): 44-49, 2022 02.
Article in English | MEDLINE | ID: mdl-33436506

ABSTRACT

ObjectivesThe southeastern US is a domestic epicentre for incident HIV with high prevalence of herpes simplex virus (HSV) coinfection. We estimated the incidence rates (IR) of symptomatic herpetic anogenital ulcer disease (HAUD) and assessed its associations with demographic and clinical characteristics, specifically with immunological markers using median, nadir and trajectory CD4 counts. METHODS: Electronic medical records (EMR) of over 7000 people living with HIV (PLWH) attending one of the leading HIV clinics in the southeastern US between 2006 and 2018 were reviewed and analysed. IR of HSV-related HAUD were estimated per 10 000 person years. Joinpoint regressions were performed to examine temporal changes in the trends of IR. All IR and trends were stratified by gender and race. Six CD4 trajectory groups were constructed using the group-based trajectory modelling. Multivariable logistic models were conducted to assess the associations of CD4 counts (nadir, median CD4 and newly defined CD4 trajectory), separately with HAUD. RESULTS: Of the 4484 PLWH eligible individuals (3429 men, 1031 women and 24 transgender), we observed 425 patients with HSV-related HAUD. The mean log10viral load was higher in HAUD than HAUD-free groups, whereas the median nadir CD4 count (cells/uL) was higher in the non-cases than the case groups (p<0.05). HAUD were more frequent in women than men. Median CD4 (<200 cell/uL) was associated with HAUD (OR=2.1), but there were no significant associations with nadir CD4. Significant associations with declining and sustained low CD4 counts trajectory patterns were observed with HAUD. CONCLUSIONS: There were significant differences between men and women with incident HAUD among PLWH. EMR-based studies can provide innovative trajectory models that can potentially be helpful in guiding screening and clinical care of HAUD among high-risk PLWH.


Subject(s)
Electronic Health Records/statistics & numerical data , Fissure in Ano/virology , Genitalia/virology , Herpes Genitalis/epidemiology , Adult , CD4 Lymphocyte Count/statistics & numerical data , Coinfection/epidemiology , Coinfection/virology , Female , Herpes Genitalis/immunology , Humans , Male , Middle Aged , Simplexvirus/genetics , Simplexvirus/immunology , Simplexvirus/pathogenicity , Southeastern United States/epidemiology , Viral Load
8.
Oral Dis ; 28(1): 150-163, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33200485

ABSTRACT

OBJECTIVE: Double-strand (ds) DNA-enveloped viruses can cause oral infection. Our aim is to investigate whether oral mucosal cells participate in immune response against cytosolic dsDNA invasion. METHODS: We examined the response to transfected herpes simplex virus (HSV) dsDNA via intracellular receptors in oral keratinocytes (RT7) and fibroblasts (GT1), and the effect of TNF-α on those responses. RESULTS: Transfected dsDNA increased CXCL10 expression via NF-κB activation in both cell types, while those responses were inhibited by knockdown of RIG-I, an RNA sensor. Although IFI16, a DNA sensor, was expressed in the nuclei of both types, its knockdown decreased transfected dsDNA-induced CXCL10 expression in GT1 but not RT7 cells. IFI16 in GT1 cells was translocated into cytoplasm from nuclei, which was attributed to immune response to cytosolic dsDNA. TNF-α enhanced transfected dsDNA-induced CXCL10, and knockdown of IFI16 decreased TNF-α and dsDNA-driven CXCL10 expression in both RT7 and GT1 cells. Finally, the combination of TNF-α and transfected dsDNA resulted in translocation of IFI16 from nuclei to cytoplasm in RT7 cells. CONCLUSION: RIG-I and IFI16 in oral mucosal cells may play important roles in host immune response against DNA viral infection, while TNF-α contributes to development of an antiviral system via those intracellular receptors.


Subject(s)
DNA, Viral/immunology , Fibroblasts , Keratinocytes , Simplexvirus/immunology , Antiviral Restriction Factors/immunology , Cell Line , Chemokine CXCL10/immunology , Cytoplasm , Fibroblasts/immunology , Humans , Immunity , Keratinocytes/immunology , Nuclear Proteins/immunology , Phosphoproteins/immunology , Receptors, Retinoic Acid/immunology , Tumor Necrosis Factor-alpha/immunology
9.
Viruses ; 13(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34960633

ABSTRACT

The environment of the central nervous system (CNS) represents a double-edged sword in the context of viral infections. On the one hand, the infectious route for viral pathogens is restricted via neuroprotective barriers; on the other hand, viruses benefit from the immunologically quiescent neural environment after CNS entry. Both the herpes simplex virus (HSV) and the rabies virus (RABV) bypass the neuroprotective blood-brain barrier (BBB) and successfully enter the CNS parenchyma via nerve endings. Despite the differences in the molecular nature of both viruses, each virus uses retrograde transport along peripheral nerves to reach the human CNS. Once inside the CNS parenchyma, HSV infection results in severe acute inflammation, necrosis, and hemorrhaging, while RABV preserves the intact neuronal network by inhibiting apoptosis and limiting inflammation. During RABV neuroinvasion, surveilling glial cells fail to generate a sufficient type I interferon (IFN) response, enabling RABV to replicate undetected, ultimately leading to its fatal outcome. To date, we do not fully understand the molecular mechanisms underlying the activation or suppression of the host inflammatory responses of surveilling glial cells, which present important pathways shaping viral pathogenesis and clinical outcome in viral encephalitis. Here, we compare the innate immune responses of glial cells in RABV- and HSV-infected CNS, highlighting different viral strategies of neuroprotection or Neuroinflamm. in the context of viral encephalitis.


Subject(s)
Encephalitis, Viral/immunology , Herpes Simplex/immunology , Immunity, Innate , Inflammation , Rabies virus/immunology , Rabies/immunology , Simplexvirus/immunology , Animals , Astrocytes/immunology , Astrocytes/virology , Blood-Brain Barrier/virology , Central Nervous System/immunology , Central Nervous System/virology , Encephalitis, Viral/virology , Herpes Simplex/virology , Humans , Microglia/immunology , Microglia/virology , Neuroglia/immunology , Neuroglia/virology , Rabies/virology , Signal Transduction
10.
Article in English | MEDLINE | ID: mdl-34429365

ABSTRACT

BACKGROUND AND OBJECTIVES: Neurotropic viruses are suspected to play a role in the pathogenesis of autoimmune diseases of the CNS such as the association between the Epstein-Barr virus (EBV) and multiple sclerosis (MS). A group of autoimmune encephalitis (AE) is linked to antibodies against neuronal cell surface proteins. Because CNS infection with the herpes simplex virus can trigger anti-NMDA receptor (NMDAR) encephalitis, a similar mechanism for EBV and other neurotropic viruses could be postulated. To investigate for previous viral infections of the CNS, intrathecally produced virus-specific antibody synthesis was determined in patients with AE. METHODS: Antibody-specific indices (AIs) against EBV and measles, rubella, varicella zoster, herpes simplex virus, and cytomegalovirus were determined in 27 patients having AE (anti-NMDAR encephalitis, n = 21, and LGI1 encephalitis, n = 6) and in 2 control groups comprising of 30 patients with MS and 21 patients with noninflammatory CNS diseases (NIND), which were sex and age matched. RESULTS: An intrathecal synthesis of antibodies against EBV was found in 5/27 (19%) patients with AE and 2/30 (7%) of the patients with MS. All these patients had also at least 1 additional elevated virus-specific AI. In contrast, in none of the patients with NIND, an elevated virus-specific AI was detected. DISCUSSION: Intrathecally produced antibodies against EBV can be found in patients with AE and MS but only together with antibodies against different neurotropic viruses. Evidence of these antibodies is the result of a polyspecific immune response similar yet distinct from MS response rather than an elapsed infection of the CNS.


Subject(s)
Antibodies, Viral/cerebrospinal fluid , Autoimmune Diseases of the Nervous System/cerebrospinal fluid , Encephalitis, Viral/cerebrospinal fluid , Herpesvirus 4, Human/immunology , Simplexvirus/immunology , Adolescent , Adult , Aged , Antibodies, Viral/blood , Autoimmune Diseases of the Nervous System/blood , Encephalitis, Viral/blood , Female , Humans , Male , Middle Aged , Young Adult
11.
Int J Biol Macromol ; 178: 514-526, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33662419

ABSTRACT

The binding mode to TAP (i.e., the peptide transporter associated with antigen processing) from a viral peptide thus far has been unknown in the field of antiviral immunity, but an interfering mode from a virus-encoded TAP inhibitor has been well documented with respect to blocking the TAP function. In the current study, we predicted the structure of the pig TAP transporter and its inhibition complex by the small viral protein ICP47 of the herpes simplex virus (HSV) encoded by the TAP inhibitor to exploit inhibition of the TAP transporter as the host's immune evasion strategy. We found that the hot spots (residues Leu5, Tyr22, and Leu51) on the ICP47 inhibitor interface tended to prevail over the favored Leu and Tyr, which contributed to significant functional binding at the C-termini recognition principle of the TAP. We further characterized the specificity determinants of the peptide transporter from the pig TAP by the ICP47 inhibitor effects and multidrug TmrAB transporter from the Thermus thermophillus and its immunity regarding its structural homolog of the pig TAP. The specialized structure-function relationship from the pig TAP exporter could provide insight into substrate specificity of the unique immunological properties from the host organism. The TAP disarming capacity from all five viral inhibitors (i.e., the five virus-encoded TAP inhibitors of ICP47, UL49.5, U6, BNLF2a, and CPXV012 proteins) was linked to the infiltration of the TAP functional structure in an unstable conformation and the mounting susceptibility caused by the host's TAP polymorphism. It is anticipated that the functional characterization of the pig TAP transporter based on the pig genomic variants will lead to additional insights into the genotype and single nucleotide polymorphism (SNP) in relation to antiviral resistance and disease susceptibility.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/chemistry , Immediate-Early Proteins/chemistry , Immune Evasion , Simplexvirus/chemistry , ATP-Binding Cassette Transporters/immunology , Animals , Humans , Immediate-Early Proteins/immunology , Simplexvirus/immunology , Structure-Activity Relationship , Swine
13.
Ann Clin Transl Neurol ; 8(2): 395-405, 2021 02.
Article in English | MEDLINE | ID: mdl-33410595

ABSTRACT

OBJECTIVE: Cerebral microbleeds (CMBs) is a subtype of cerebral small vessel disease. Their underlying pathogenesis remains unclear. The aim of this study was to investigate the association between infectious burden (IB) and CMBs. METHODS: Seven hundred and seventy-three consecutive patients who were hospitalized in the Department of Neurology in General Hospital of Western Theater Command without severe neurological symptoms were recruited and selected in this pilot cross-sectional study. CMBs were assessed using the susceptibility-weighted imaging sequence of magnetic resonance imaging. Immunoglobulin G antibodies against common pathogens, including herpes simplex virus (HSV)-1, HSV-2, cytomegalovirus (CMV), Chlamydia pneumoniae (C. pneumoniae), Mycoplasma pneumoniae (M. pneumoniae), Epstein-Barr virus (EBV), Helicobacter pylori (HP), and Borrelia burgdorferi (B. burgdorferi), were measured by commercial ELISA assays. IB was defined as a composite serologic measure of exposure to these common pathogens. RESULTS: Patients with and without CMBs were defined as the CMBs group (n = 76) and the non-CMBs group (n = 81), respectively. IB was significantly different between the CMBs and non-CMBs groups. After adjusted for other risk factors, the increased IB was independently associated with the presence of CMBs (P = 0.031, OR = 3.00, 95% CI [1.11-8.15]). IB was significantly positively associated with the number of CMBs (Spearman ρ = 0.653, P < 0.001). The levels of serum inflammatory markers were significantly different between the CMBs and non-CMBs groups and among the categories of IB. INTERPRETATION: IB consisting of HSV-1, HSV-2, CMV, C. pneumoniae, M. pneumoniae, EBV, HP, and B. burgdorferi was associated with CMBs. All the findings suggested that pathogen infection could be involved in the pathogenesis of CMBs.


Subject(s)
Biomarkers/blood , Cerebral Hemorrhage , Cerebral Small Vessel Diseases , Aged , Borrelia burgdorferi/immunology , Cerebral Hemorrhage/diagnostic imaging , Cerebral Hemorrhage/microbiology , Cerebral Hemorrhage/physiopathology , Cerebral Hemorrhage/virology , Cerebral Small Vessel Diseases/diagnostic imaging , Cerebral Small Vessel Diseases/microbiology , Cerebral Small Vessel Diseases/physiopathology , Cerebral Small Vessel Diseases/virology , Chlamydophila pneumoniae/immunology , Cross-Sectional Studies , Cytomegalovirus/immunology , Female , Helicobacter pylori/immunology , Herpesvirus 4, Human/immunology , Humans , Immunoglobulin G/blood , Magnetic Resonance Imaging , Male , Middle Aged , Mycoplasma pneumoniae/immunology , Pilot Projects , Risk Factors , Simplexvirus/immunology
14.
Curr Issues Mol Biol ; 41: 381-468, 2021.
Article in English | MEDLINE | ID: mdl-32938804

ABSTRACT

The therapeutic promise of oncolytic viruses (OVs) rests on their ability to both selectively kill tumor cells and induce anti-tumor immunity. The potential of tumors to be recognized and eliminated by an effective anti-tumor immune response has been spurred on by the discovery that immune checkpoint inhibition can overcome tumor-specific cytotoxic T cell (CTL) exhaustion and provide durable responses in multiple tumor indications. OV-mediated tumor destruction is now recognized as a powerful means to assist in the development of anti-tumor immunity for two important reasons: (i) OVs, through the elicitation of an anti-viral response and the production of type I interferon, are potent stimulators of inflammation and can be armed with transgenes to further enhance anti-tumor immune responses; and (ii) lytic activity can promote the release of tumor-associated antigens (TAAs) and tumor neoantigens that function as in situ tumor-specific vaccines to elicit adaptive immunity. Oncolytic herpes simplex viruses (oHSVs) are among the most widely studied OVs for the treatment of solid malignancies, and Amgen's oHSV Imlygic® for the treatment of melanoma is the only OV approved in major markets. Here we describe important biological features of HSV that make it an attractive OV, clinical experience with HSV-based vectors, and strategies to increase applicability to cancer treatment.


Subject(s)
Immune Checkpoint Inhibitors/immunology , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Viruses/immunology , Simplexvirus/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Animals , Humans , Immune Checkpoint Inhibitors/pharmacology , T-Lymphocytes, Cytotoxic/immunology
15.
Cell Rep ; 33(13): 108567, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33378682

ABSTRACT

Concurrent infection with multiple pathogens occurs frequently in individuals and can result in exacerbated infections and altered immunity. However, the impact of such coinfections on immune responses remains poorly understood. Here, we reveal that systemic infection results in an inflammation-induced suppression of local immunity. During localized infection or vaccination in barrier tissues including the skin or respiratory tract, concurrent systemic infection induces a type I interferon-dependent lymphopenia that impairs lymphocyte recruitment to the draining lymph node (dLN) and induces sequestration of lymphocytes in non-draining LN. This contributes to suppressed fibroblastic reticular cell and endothelial cell expansion and dLN remodeling and impairs induction of B cell responses and antibody production. Our data suggest that contemporaneous systemic inflammation constrains the induction of regional immunity.


Subject(s)
Coinfection/immunology , Herpes Simplex/immunology , Inflammation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphoid Tissue/immunology , Simplexvirus/immunology , Animals , Antibody Formation , Fibroblasts/immunology , Herpes Simplex/virology , Interferon Type I/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytic Choriomeningitis/virology , Lymphoid Tissue/metabolism , Lymphopenia/immunology , Mice , Mice, Inbred C57BL , Stromal Cells/immunology , Stromal Cells/metabolism
16.
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
17.
Int J Infect Dis ; 101: 269-275, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33011282

ABSTRACT

OBJECTIVES: The aim of this study was to investigate whether the seroprevalence of IgG antibodies against seven viruses (cytomegalovirus, herpes simplex virus 1&2, measles morbillivirus, parvovirus B19, rubella, and varicella-zoster virus), which can potentially compromise maternal and fetal wellbeing, differs based on country of origin among women with chronic hepatitis B (CHB). METHOD: This study was a single-center, hospital-based cross-sectional study. The study included women with CHB 15-45 years of age, included in the Danish Database for Hepatitis B and C. Seroprevalence estimates were calculated with a 95% confidence interval and were compared between age groups, regions of origin, and to the general population. RESULTS: 177 women were included in the study. Overall, the seroprevalences of antibodies were similar among women with CHB with origin outside Denmark and compared to the general population in Denmark, but there was a notable difference in the seroprevalence of antibodies against herpes simplex 2 between women from Africa (37.1% CI 95% 22.0;55.1) and women from the Middle East (2.5% CI 95% 0.1;14.7). CONCLUSION: Women with CHB whose origin is outside Denmark do not appear to differ, based on origin, or be at greater risk of acquiring these viruses during pregnancy than their Danish counterparts.


Subject(s)
Antibodies, Viral/blood , Hepatitis B, Chronic/blood , Hepatitis B/immunology , Herpesvirus 3, Human/immunology , Measles virus/immunology , Parvovirus B19, Human/immunology , Rubella virus/immunology , Simplexvirus/immunology , Adolescent , Adult , Cross-Sectional Studies , Cytomegalovirus/immunology , Denmark/epidemiology , Female , Hepatitis B, Chronic/epidemiology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Humans , Immunoglobulin G/blood , Middle Aged , Pregnancy , Prevalence , Seroepidemiologic Studies , Young Adult
18.
Cells ; 9(10)2020 09 25.
Article in English | MEDLINE | ID: mdl-32992967

ABSTRACT

To investigate whether and how cerebrospinal fluid (CSF) findings can contribute to distinguish tick-borne encephalitis (TBE) from herpes simplex virus (HSV) and varicella zoster virus (VZV) induced central nervous system (CNS) infections (HSV-I, VZV-I). Chart review and identification of TBE, HSV- I, and VZV-I was carried out, fulfilling the following criteria: (1) clinical signs of encephalitis and/or meningitis, (2) complete CSF analysis and confirmed viral etiology by either PCR or antibody testing in CSF, (3) hospitalized patients, and (4) available brain magnetic resonance imaging (MRI). Fifty-nine patients with 118 CSF/serum pairs were included. These comprised 21 with TBE (35 CSF/serum pairs), 20 (40 CSF/serum pairs) with HSV-I, and 18 (43 CSF/serum pairs) with VZV-I. In contrast to HSV-I and VZV-I, CSF cell differentiation in TBE showed more often an increased (>20%) proportion of granulocytes (p < 0.01) and a more frequent quantitative intrathecal IgM synthesis (p = 0.001 and p < 0.01, respectively), while the second was even more pronounced when follow-up CSF analyses were included (p < 0.001). CSF findings help to distinguish TBE from other viral infections. In cases with CSF pleocytosis and a positive history for a stay in or near an endemic area, TBE antibodies in CSF and serum should be determined, especially if granulocytes in CSF cell differentiation and/or an intrathecal IgM synthesis is present.


Subject(s)
Central Nervous System Infections/diagnosis , Diagnosis, Differential , Encephalitis, Tick-Borne/diagnosis , Meningitis/diagnosis , Adult , Aged , Central Nervous System Infections/blood , Central Nervous System Infections/cerebrospinal fluid , Central Nervous System Infections/virology , Encephalitis, Tick-Borne/blood , Encephalitis, Tick-Borne/cerebrospinal fluid , Encephalitis, Tick-Borne/virology , Female , Herpesvirus 3, Human/pathogenicity , Humans , Immunity, Humoral/genetics , Immunity, Humoral/immunology , Immunoglobulin M/blood , Leukocytosis/blood , Leukocytosis/cerebrospinal fluid , Leukocytosis/diagnosis , Leukocytosis/virology , Magnetic Resonance Imaging , Male , Meningitis/blood , Meningitis/cerebrospinal fluid , Meningitis/virology , Middle Aged , Simplexvirus/immunology , Simplexvirus/pathogenicity
19.
Sci Immunol ; 5(50)2020 08 14.
Article in English | MEDLINE | ID: mdl-32817296

ABSTRACT

Herpes simplex virus (HSV) glycoprotein D (gD) not only is required for virus entry and cell-to-cell spread but also binds the host immunomodulatory molecule, HVEM, blocking interactions with its ligands. Natural infection primarily elicits neutralizing antibodies targeting gD, but subunit protein vaccines designed to induce this response have failed clinically. In contrast, preclinical studies demonstrate that an HSV-2 single-cycle strain deleted in gD, ΔgD-2, induces primarily non-neutralizing antibodies that activate Fcγ receptors (FcγRs) to mediate antibody-dependent cellular cytotoxicity (ADCC). These studies were designed to test the hypothesis that gD interferes with ADCC through engagement of HVEM. Immunization of Hvem-/- mice with ΔgD-2 resulted in significant reduction in HSV-specific IgG2 antibodies, the subclass associated with FcγR activation and ADCC, compared with wild-type controls. This translated into a parallel reduction in active and passive vaccine protection. A similar decrease in ADCC titers was observed in Hvem-/- mice vaccinated with an alternative HSV vaccine candidate (dl5-29) or an unrelated vesicular stomatitis virus-vectored vaccine. Unexpectedly, not only did passive transfer of immune serum from ΔgD-2-vaccinated Hvem-/- mice fail to protect wild-type mice but transfer of immune serum from ΔgD-2-vaccinated wild-type mice failed to protect Hvem-/- mice. Immune cells isolated from Hvem-/- mice were impaired in FcγR activation, and, conversely, addition of gD protein or anti-HVEM antibodies to in vitro murine or human FcγR activation assays inhibited the response. These findings uncover a previously unrecognized role for HVEM signaling in generating and mediating ADCC and an additional HSV immune evasion strategy.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Herpes Simplex/immunology , Receptors, Tumor Necrosis Factor, Member 14/immunology , Simplexvirus/immunology , Viral Vaccines/administration & dosage , Animals , Female , Herpes Simplex/prevention & control , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 14/genetics , Signal Transduction
20.
United European Gastroenterol J ; 8(3): 303-313, 2020 04.
Article in English | MEDLINE | ID: mdl-32529821

ABSTRACT

Background: The magnitude and drivers of the risk of serious viral infections in Inflammatory Bowel diseases (IBD) are unclear. Objective: The objective of this study was to assess the incidence and risk factors for systemic serious viral infections in IBD patients. Methods: Using MICISTA, a database detailing prospective characteristics and complications of IBD, we identified patients that were followed for IBD in 2005-2014 outside the context of organ transplantation, HIV infection or chronic viral hepatitis. We estimated incidences of systemic serious viral infections, defined by the need for hospitalization or permanent organ damage. Standardized incidence ratios (SIRs) were calculated using the French hospital database. We performed a case-control study nested in MICISTA for assessing the role of exposure to IBD drugs and IBD clinical activity in the risk of developing infection. Results: We identified 31 patients with serious viral infections among 2645 patients followed for 15,383 person-years. We observed 13 cases of cytomegalovirus, 10 Epstein-Barr virus, 5 varicella zoster virus and 3 herpes simplex virus infections. No deaths occurred. The incidence rate of infections in patients with IBD was 2.02/1000 person-years, and the SIR was 3.09 (95% confidence interval (CI), 1.98-4.20; p = 0.0002) in the study population. By multivariate analysis, increased risk of infection was associated with exposure to thiopurines (odds ratio (OR), 3.48; 95% CI, 1.36-8.90; p = 0.009), and clinically active IBD at onset of infection (OR, 3.35; 95% CI, 1.23-9.23; p = 0.02). Conclusions: The incidence of systemic serious viral infections in patients with IBD is tripled compared to general population. Clinically active IBD and exposure to thiopurines are the main drivers of the risk.


Subject(s)
Azathioprine/adverse effects , Herpesviridae Infections/epidemiology , Immunosuppressive Agents/adverse effects , Inflammatory Bowel Diseases/drug therapy , Adolescent , Adult , Aged , Case-Control Studies , Cost of Illness , Cytomegalovirus/immunology , Cytomegalovirus/isolation & purification , Female , France/epidemiology , Herpesviridae Infections/diagnosis , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesvirus 3, Human/immunology , Herpesvirus 3, Human/isolation & purification , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/isolation & purification , Hospitalization/statistics & numerical data , Humans , Incidence , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/immunology , Male , Middle Aged , Prospective Studies , Risk Assessment/statistics & numerical data , Risk Factors , Severity of Illness Index , Simplexvirus/immunology , Simplexvirus/isolation & purification , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL