Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Trends Immunol ; 40(9): 842-856, 2019 09.
Article in English | MEDLINE | ID: mdl-31439415

ABSTRACT

Interferons (IFNs) comprise a pleiotropic family of signaling molecules that are often the first line of defense against viral infection. Inflammatory responses induced by IFN are generally well tolerated during peripheral infections; yet, the same degree of inflammation during infection of the central nervous system (CNS) could be catastrophic. Thus, IFN responses must be modified within the CNS to ensure host survival. In this review, we discuss emerging principles highlighting unique aspects of antiviral effects of IFN protection following neurotropic viral infection, chiefly using new techniques in rodent models. Evaluation of these unique responses provides insights into how the immune system eradicates or controls pathogens, while avoiding host damage. Defining these principles may have direct impact on the development of novel clinical approaches.


Subject(s)
Antiviral Agents/immunology , Interferons/immunology , Virus Diseases/immunology , Animals , Humans
2.
J Natl Compr Canc Netw ; 20(2): 160-166, 2022 02.
Article in English | MEDLINE | ID: mdl-35130494

ABSTRACT

BACKGROUND: Most safety and efficacy trials of the SARS-CoV-2 vaccines excluded patients with cancer, yet these patients are more likely than healthy individuals to contract SARS-CoV-2 and more likely to become seriously ill after infection. Our objective was to record short-term adverse reactions to the COVID-19 vaccine in patients with cancer, to compare the magnitude and duration of these reactions with those of patients without cancer, and to determine whether adverse reactions are related to active cancer therapy. PATIENTS AND METHODS: A prospective, single-institution observational study was performed at an NCI-designated Comprehensive Cancer Center. All study participants received 2 doses of the Pfizer BNT162b2 vaccine separated by approximately 3 weeks. A report of adverse reactions to dose 1 of the vaccine was completed upon return to the clinic for dose 2. Participants completed an identical survey either online or by telephone 2 weeks after the second vaccine dose. RESULTS: The cohort of 1,753 patients included 67.5% who had a history of cancer and 12.0% who were receiving active cancer treatment. Local pain at the injection site was the most frequently reported symptom for all respondents and did not distinguish patients with cancer from those without cancer after either dose 1 (39.3% vs 43.9%; P=.07) or dose 2 (42.5% vs 40.3%; P=.45). Among patients with cancer, those receiving active treatment were less likely to report pain at the injection site after dose 1 compared with those not receiving active treatment (30.0% vs 41.4%; P=.002). The onset and duration of adverse events was otherwise unrelated to active cancer treatment. CONCLUSIONS: When patients with cancer were compared with those without cancer, few differences in reported adverse events were noted. Active cancer treatment had little impact on adverse event profiles.


Subject(s)
COVID-19 , Neoplasms , BNT162 Vaccine , COVID-19 Vaccines , Humans , Neoplasms/drug therapy , Prospective Studies , RNA, Messenger , SARS-CoV-2
3.
J Virol ; 94(9)2020 04 16.
Article in English | MEDLINE | ID: mdl-32051270

ABSTRACT

Influenza A viruses (IAV) are lytic viruses that have recently been found to activate necroptosis in many of the cell types they infect. Necroptotic cell death is potently immunogenic and limits IAV spread by directly eliminating infected cells and by mobilizing both innate and adaptive immune responses. The benefits of necroptosis to the host, however, may sometimes be outweighed by the potentially deleterious hyperinflammatory consequences of activating this death modality in pulmonary and other tissues.


Subject(s)
Influenza, Human/metabolism , Necroptosis/physiology , Orthomyxoviridae/metabolism , Animals , Apoptosis , Caspase 8/metabolism , Cell Death , Humans , Influenza A virus/metabolism , Influenza, Human/virology , Necrosis , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/metabolism , Phosphorylation , Protein Kinases/metabolism
4.
Nat Rev Neurosci ; 17(12): 766-776, 2016 12.
Article in English | MEDLINE | ID: mdl-27811921

ABSTRACT

It is becoming clear that the manner by which the immune response resolves or contains infection by a pathogen varies according to the tissue that is affected. Unlike many peripheral cell types, CNS neurons are generally non-renewable. Thus, the cytolytic and inflammatory strategies that are effective in controlling infections in the periphery could be damaging if deployed in the CNS. Perhaps for this reason, the immune response to some CNS viral infections favours maintenance of neuronal integrity and non-neurolytic viral control. This modified immune response - when combined with the unique anatomy and physiology of the CNS - provides an ideal environment for the maintenance of viral genomes, including those of RNA viruses. Therefore, it is possible that such viruses can reactivate long after initial viral exposure, contributing to CNS disease.


Subject(s)
Brain/immunology , Immunity, Innate/physiology , Neurons/immunology , RNA Virus Infections/immunology , RNA Viruses/immunology , Animals , Brain/metabolism , Brain/pathology , Central Nervous System Diseases/immunology , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Humans , Neurons/metabolism , Neurons/pathology , RNA Virus Infections/metabolism , RNA Virus Infections/pathology , RNA Viruses/metabolism
5.
J Immunol ; 203(5): 1348-1355, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31358656

ABSTRACT

Receptor-interacting protein kinase 1 (RIPK1) regulates cell fate and proinflammatory signaling downstream of multiple innate immune pathways, including those initiated by TNF-α, TLR ligands, and IFNs. Genetic ablation of Ripk1 results in perinatal lethality arising from both RIPK3-mediated necroptosis and FADD/caspase-8-driven apoptosis. IFNs are thought to contribute to the lethality of Ripk1-deficient mice by activating inopportune cell death during parturition, but how IFNs activate cell death in the absence of RIPK1 is not understood. In this study, we show that Z-form nucleic acid binding protein 1 (ZBP1; also known as DAI) drives IFN-stimulated cell death in settings of RIPK1 deficiency. IFN-activated Jak/STAT signaling induces robust expression of ZBP1, which complexes with RIPK3 in the absence of RIPK1 to trigger RIPK3-driven pathways of caspase-8-mediated apoptosis and MLKL-driven necroptosis. In vivo, deletion of either Zbp1 or core IFN signaling components prolong viability of Ripk1-/- mice for up to 3 mo beyond parturition. Together, these studies implicate ZBP1 as the dominant activator of IFN-driven RIPK3 activation and perinatal lethality in the absence of RIPK1.


Subject(s)
Cell Death/physiology , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/physiology , Caspase 8/metabolism , Cell Line , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
6.
J Cell Sci ; 131(17)2018 09 05.
Article in English | MEDLINE | ID: mdl-30154209

ABSTRACT

T cell-mediated adaptive immunity requires naïve, unstimulated T cells to transition from a quiescent metabolic state into a highly proliferative state upon T cell receptor engagement. This complex process depends on transcriptional changes mediated by Ca2+-dependent NFAT signaling, mTOR-mediated signaling and increased activity of the guanine nucleotide biosynthetic inosine-5'-monophosphate (IMP) dehydrogenase 1 and 2 enzymes (IMPDH1 and IMPDH2, hereafter IMPDH). Inhibitors of these pathways serve as potent immunosuppressants. Unexpectedly, we discovered that all three pathways converge to promote the assembly of IMPDH protein into micron-scale macromolecular filamentous structures in response to T cell activation. Assembly is post-transcriptionally controlled by mTOR and the Ca2+ influx regulator STIM1. Furthermore, IMPDH assembly and catalytic activity were negatively regulated by guanine nucleotide levels, suggesting a negative feedback loop that limits biosynthesis of guanine nucleotides. Filamentous IMPDH may be more resistant to this inhibition, facilitating accumulation of the higher GTP levels required for T cell proliferation.


Subject(s)
IMP Dehydrogenase/metabolism , T-Lymphocytes/enzymology , Animals , Cells, Cultured , Guanine Nucleotides/metabolism , IMP Dehydrogenase/genetics , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Spleen/enzymology , Spleen/immunology , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
7.
J Virol ; 93(18)2019 09 15.
Article in English | MEDLINE | ID: mdl-31270232

ABSTRACT

Genomic material from many neurotropic RNA viruses (e.g., measles virus [MV], West Nile virus [WNV], Sindbis virus [SV], rabies virus [RV], and influenza A virus [IAV]) remains detectable in the mouse brain parenchyma long after resolution of the acute infection. The presence of these RNAs in the absence of overt central nervous system (CNS) disease has led to the suggestion that they are viral remnants, with little or no potential to reactivate. Here we show that MV RNA remains detectable in permissive mouse neurons long after challenge with MV and, moreover, that immunosuppression can cause RNA and protein synthesis to rebound, triggering neuropathogenesis months after acute viral control. Robust recrudescence of viral transcription and protein synthesis occurs after experimental depletion of cells of the adaptive immune response and is associated with a loss of T resident memory (Trm) lymphocytes within the brain. The disease associated with loss of immune control is distinct from that seen during the acute infection: immune cell-depleted, long-term-infected mice display severe gait and motor problems, in contrast to the wasting and lethal disease that occur during acute infection of immunodeficient hosts. These results illuminate the potential consequences of noncytolytic, immune-mediated viral control in the CNS and demonstrate that what were once considered "resolved" RNA viral infections may, in fact, induce diseases later in life that are distinct from those caused by acute infection.IMPORTANCE Viral infections of neurons are often not cytopathic; thus, once-infected neurons survive, and viral RNAs can be detected long after apparent viral control. These RNAs are generally considered viral fossils, unlikely to contribute to central nervous system (CNS) disease. Using a mouse model of measles virus (MV) neuronal infection, we show that MV RNA is maintained in the CNS of infected mice long after acute control and in the absence of overt disease. Viral replication is suppressed by the adaptive immune response; when these immune cells are depleted, viral protein synthesis recurs, inducing a CNS disease that is distinct from that observed during acute infection. The studies presented here provide the basis for understanding how persistent RNA infections in the CNS are controlled by the host immune response, as well as the pathogenic consequences of noncytolytic viral control.


Subject(s)
Measles virus/genetics , Neurons/virology , RNA Virus Infections/virology , Animals , Brain/virology , Central Nervous System/virology , Disease Models, Animal , Female , Male , Measles/virology , Measles virus/metabolism , Mice , Mice, Transgenic , Neurons/metabolism , RNA/genetics , RNA/metabolism , RNA Virus Infections/genetics , RNA Virus Infections/metabolism , RNA Viruses/genetics , RNA Viruses/metabolism
8.
J Virol ; 89(21): 11011-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26311886

ABSTRACT

UNLABELLED: In permissive mouse central nervous system (CNS) neurons, measles virus (MV) spreads in the absence of hallmark viral budding or neuronal death, with transmission occurring efficiently and exclusively via the synapse. MV infection also initiates a robust type I interferon (IFN) response, resulting in the synthesis of a large number of genes, including bone marrow stromal antigen 2 (Bst2)/tetherin/CD317. Bst2 restricts the release of some enveloped viruses, but to date, its role in viral infection of neurons has not been assessed. Consequently, we investigated how Bst2 was induced and what role it played in MV neuronal infection. The magnitude of induction of neuronal Bst2 RNA and protein following IFN exposure and viral infection was notably higher than in similarly treated mouse embryo fibroblasts (MEFs). Bst2 synthesis was both IFN and Stat1 dependent. Although Bst2 prevented MV release from nonneuronal cells, its deletion had no effect on viral pathogenesis in MV-challenged mice. Our findings underscore how cell-type-specific differences impact viral infection and pathogenesis. IMPORTANCE: Viral infections of the central nervous system can lead to debilitating disease and death. Moreover, it is becoming increasingly clear that nonrenewable cells, including most central nervous system neurons, combat neurotropic viral infections in fundamentally different ways than other rapidly dividing and renewable cell populations. Here we identify type I interferon signaling as a key inducer of a known antiviral protein (Bst2) in neurons. Unexpectedly, the gene is dispensable for clearance of neurotropic viral infection despite its well-defined contribution to limiting the spread of enveloped viruses in proliferating cells. A deeper appreciation of the importance of cell type heterogeneity in antiviral immunity will aid in the identification of unique therapeutic targets for life-threatening viral infections.


Subject(s)
Antigens, CD/metabolism , Gene Expression Regulation, Viral/physiology , Interferon Type I/metabolism , Measles virus/physiology , Measles/immunology , Membrane Glycoproteins/metabolism , Neurons/metabolism , Analysis of Variance , Animals , Blotting, Western , Cells, Cultured , DNA Primers/genetics , Fluorescent Antibody Technique , Hippocampus/cytology , Mice , Neurons/virology , Reverse Transcriptase Polymerase Chain Reaction
9.
Proc Natl Acad Sci U S A ; 110(33): E3109-18, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23898178

ABSTRACT

Interferons (IFNs) are cytokines with powerful immunomodulatory and antiviral properties, but less is known about how they induce cell death. Here, we show that both type I (α/ß) and type II (γ) IFNs induce precipitous receptor-interacting protein (RIP)1/RIP3 kinase-mediated necrosis when the adaptor protein Fas-associated death domain (FADD) is lost or disabled by phosphorylation, or when caspases (e.g., caspase 8) are inactivated. IFN-induced necrosis proceeds via progressive assembly of a RIP1-RIP3 "necrosome" complex that requires Jak1/STAT1-dependent transcription, but does not need the kinase activity of RIP1. Instead, IFNs transcriptionally activate the RNA-responsive protein kinase PKR, which then interacts with RIP1 to initiate necrosome formation and trigger necrosis. Although IFNs are powerful activators of necrosis when FADD is absent, these cytokines are likely not the dominant inducers of RIP kinase-driven embryonic lethality in FADD-deficient mice. We also identify phosphorylation on serine 191 as a mechanism that disables FADD and collaborates with caspase inactivation to allow IFN-activated necrosis. Collectively, these findings outline a mechanism of IFN-induced RIP kinase-dependent necrotic cell death and identify FADD and caspases as negative regulators of this process.


Subject(s)
Cell Cycle Checkpoints/physiology , Fas-Associated Death Domain Protein/metabolism , Interferon-gamma/metabolism , Models, Molecular , Necrosis/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Fas-Associated Death Domain Protein/chemistry , Fas-Associated Death Domain Protein/genetics , GTPase-Activating Proteins/metabolism , Immunoprecipitation , Mice , Mice, Knockout , Phosphorylation , RNA Interference , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , STAT1 Transcription Factor/metabolism , eIF-2 Kinase/metabolism
10.
J Neurochem ; 135(2): 309-22, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26190522

ABSTRACT

The signal transduction molecule, Stat1, is critical for the expression of type I and II interferon (IFN)-responsive genes in most cells; however, we previously showed that primary hippocampal mouse neurons express low basal Stat1, with delayed and attenuated expression of IFN-responsive genes. Moreover, IFNγ-dependent resolution of a neurotropic viral challenge in permissive mice is Stat1-independent. Here, we show that exogenous IFNγ has no deleterious impact on neuronal viability, and staurosporine-induced apoptosis in neurons is significantly blunted by the addition of IFNγ, suggesting that IFNγ confers a pro-survival signal in neurons. To identify the pathways induced by IFNγ in neurons, the activation of alternative signal transducers associated with IFNγ signaling was assessed. Rapid and pronounced activation of extracellular signal regulated kinase (Erk1/2) was observed in neurons, compared to a modest response in fibroblasts. Moreover, the absence of Stat1 in primary fibroblasts led to enhanced Erk activation following IFNγ addition, implying that the cell-specific availability of signal transducers can diversify the cellular response following IFN engagement.


Subject(s)
Interferon-gamma/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Animals , Antimetabolites/pharmacology , Bromodeoxyuridine/pharmacology , Cell Survival/drug effects , Cytosol/metabolism , Female , Hippocampus/cytology , Hippocampus/embryology , MAP Kinase Signaling System/drug effects , Mice , Mice, Transgenic , Neuroglia/drug effects , Pregnancy , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Staurosporine/pharmacology
11.
PLoS Pathog ; 8(10): e1002971, 2012.
Article in English | MEDLINE | ID: mdl-23071441

ABSTRACT

Rabies virus (RABV) is a highly neurotropic pathogen that typically leads to mortality of infected animals and humans. The precise etiology of rabies neuropathogenesis is unknown, though it is hypothesized to be due either to neuronal death or dysfunction. Analysis of human brains post-mortem reveals surprisingly little tissue damage and neuropathology considering the dramatic clinical symptomology, supporting the neuronal dysfunction model. However, whether or not neurons survive infection and clearance and, provided they do, whether they are functionally restored to their pre-infection phenotype has not been determined in vivo for RABV, or any neurotropic virus. This is due, in part, to the absence of a permanent "mark" on once-infected cells that allow their identification long after viral clearance. Our approach to study the survival and integrity of RABV-infected neurons was to infect Cre reporter mice with recombinant RABV expressing Cre-recombinase (RABV-Cre) to switch neurons constitutively expressing tdTomato (red) to expression of a Cre-inducible EGFP (green), permanently marking neurons that had been infected in vivo. We used fluorescence microscopy and quantitative real-time PCR to measure the survival of neurons after viral clearance; we found that the vast majority of RABV-infected neurons survive both infection and immunological clearance. We were able to isolate these previously infected neurons by flow cytometry and assay their gene expression profiles compared to uninfected cells. We observed transcriptional changes in these "cured" neurons, predictive of decreased neurite growth and dysregulated microtubule dynamics. This suggests that viral clearance, though allowing for survival of neurons, may not restore them to their pre-infection functionality. Our data provide a proof-of-principle foundation to re-evaluate the etiology of human central nervous system diseases of unknown etiology: viruses may trigger permanent neuronal damage that can persist or progress in the absence of sustained viral antigen.


Subject(s)
Brain/virology , Neurons/physiology , Rabies virus/immunology , Rabies/immunology , Animals , Brain/immunology , Brain/pathology , Cell Survival , Gene Expression , Green Fluorescent Proteins/genetics , Integrases/genetics , Mice , Neurons/immunology , Neurons/virology , Rabies/genetics , Rabies/pathology , Rabies/virology , Rabies virus/pathogenicity , Real-Time Polymerase Chain Reaction , Transcriptome
12.
J Immunol ; 188(4): 1915-23, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22246627

ABSTRACT

Neurons are chiefly nonrenewable; thus, cytolytic immune strategies to clear or control neurotropic viral infections could have lasting neurologic consequences. IFN-γ is a potent antiviral cytokine that is critical for noncytolytic clearance of multiple neurotropic viral infections, including measles virus (MV); however, the downstream pathways through which IFN-γ functions in neurons have not been defined. Unlike most cell types studied to date in which IFN-γ affects gene expression via rapid and robust activation of STAT1, basal STAT1 levels in primary hippocampal neurons are constitutively low, resulting in attenuated STAT1 activation and consequently slower kinetics of IFN-γ-driven STAT1-dependent gene expression. Given this altered expression and activation of STAT1 in neurons, we sought to determine whether STAT1 was required for IFN-γ-mediated protection from infection in neurons. To do so, we evaluated the consequences of MV challenge of STAT1-deficient mice and primary hippocampal neurons explanted from these mice. Surprisingly, the absence of STAT1 did not restrict the ability of IFN-γ to control viral infection either in vivo or ex vivo. Moreover, the canonical IFN-γ-triggered STAT1 gene expression profile was not induced in STAT1-deficient neurons, suggesting that IFN-γ regulates neuronal STAT1-independent pathways to control viral replication.


Subject(s)
Interferon-gamma/immunology , Measles virus/immunology , Measles/immunology , Neurons/immunology , Neurons/virology , STAT1 Transcription Factor/metabolism , Animals , Cells, Cultured , Hippocampus/cytology , Hippocampus/immunology , Hippocampus/metabolism , Interferon-gamma/metabolism , Measles/metabolism , Measles/virology , Measles virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , Virus Replication
13.
PLoS Pathog ; 7(12): e1002462, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22216008

ABSTRACT

Although viruses have been implicated in central nervous system (CNS) diseases of unknown etiology, including multiple sclerosis and amyotrophic lateral sclerosis, the reproducible identification of viral triggers in such diseases has been largely unsuccessful. Here, we explore the hypothesis that viruses need not replicate in the tissue in which they cause disease; specifically, that a peripheral infection might trigger CNS pathology. To test this idea, we utilized a transgenic mouse model in which we found that immune cells responding to a peripheral infection are recruited to the CNS, where they trigger neurological damage. In this model, mice are infected with both CNS-restricted measles virus (MV) and peripherally restricted lymphocytic choriomeningitis virus (LCMV). While infection with either virus alone resulted in no illness, infection with both viruses caused disease in all mice, with ∼50% dying following seizures. Co-infection resulted in a 12-fold increase in the number of CD8+ T cells in the brain as compared to MV infection alone. Tetramer analysis revealed that a substantial proportion (>35%) of these infiltrating CD8+ lymphocytes were LCMV-specific, despite no detectable LCMV in CNS tissues. Mechanistically, CNS disease was due to edema, induced in a CD8-dependent but perforin-independent manner, and brain herniation, similar to that observed in mice challenged intracerebrally with LCMV. These results indicate that T cell trafficking can be influenced by other ongoing immune challenges, and that CD8+ T cell recruitment to the brain can trigger CNS disease in the apparent absence of cognate antigen. By extrapolation, human CNS diseases of unknown etiology need not be associated with infection with any particular agent; rather, a condition that compromises and activates the blood-brain barrier and adjacent brain parenchyma can render the CNS susceptible to pathogen-independent immune attack.


Subject(s)
Brain/immunology , CD8-Positive T-Lymphocytes/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Measles virus/immunology , Measles/immunology , Animals , Brain/virology , Brain Edema/genetics , Brain Edema/immunology , Brain Edema/metabolism , Brain Edema/pathology , Brain Edema/virology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/genetics , Humans , Lymphocytic Choriomeningitis/complications , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/metabolism , Measles/complications , Measles/genetics , Measles/pathology , Measles/virology , Measles virus/metabolism , Mice , Mice, Knockout
14.
J Virol ; 84(1): 312-20, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19828618

ABSTRACT

Although much is known about lymphocytic choriomeningitis virus (LCMV) infection and the subsequent immune response in its natural murine host, some crucial aspects of LCMV-mediated pathogenesis remain undefined, including the underlying basis of the characteristic central nervous system disease that occurs following intracerebral (i.c.) challenge. We show that the classic seizures and paresis that occur following i.c. infection of adult, immunocompetent mice with LCMV are accompanied by anatomical and histological changes that are consistent with brain herniation, likely of the uncal subtype, as a causative basis for disease and precipitous death. Both by water weight determinations and by magnetic resonance imaging of infected brain tissues, edema was detected only at the terminal stages of disease, likely caused by the leakage of cerebrospinal fluid from the ventricles into the parenchyma. Furthermore, death was accompanied by unilateral pupillary dilation, which is indicative of uncal herniation. While immunohistochemical analysis revealed periventricular inflammation and a loss of integrity of the blood-brain barrier (BBB), these events preceded seizures by 2 to 3 days. Moreover, surviving perforin knockout mice showed barrier permeability equivalent to that of moribund, immunocompetent mice; thus, BBB damage does not appear to be the basis of LCMV-induced neuropathogenesis. Importantly, brain herniation can occur in humans as a consequence of injuries that would be predicted to increase intracranial pressure, including inflammation, head trauma, and brain tumors. Thus, a mechanistic dissection of the basis of LCMV neuropathogenesis may be informative for the development of interventive therapies to prevent this typically fatal human condition.


Subject(s)
Edema/etiology , Encephalocele/etiology , Lymphocytic Choriomeningitis/mortality , Lymphocytic choriomeningitis virus , Animals , Blood-Brain Barrier/pathology , Edema/pathology , Encephalocele/pathology , Inflammation , Magnetic Resonance Imaging , Mice , Mice, Knockout , Mortality , Paresis , Perforin/deficiency , Seizures
15.
mBio ; 12(2)2021 03 23.
Article in English | MEDLINE | ID: mdl-33758092

ABSTRACT

Viruses, including members of the herpes-, entero-, and morbillivirus families, are the most common cause of infectious encephalitis in mammals worldwide. During most instances of acute viral encephalitis, neurons are typically the initial cell type that is infected. However, as replication and spread ensue, other parenchymal cells can become viral targets, especially in chronic infections. Consequently, to ascertain how neurotropic viruses trigger neuropathology, it is crucial to identify which central nervous system (CNS) cell populations are susceptible and permissive throughout the course of infection, and to define how viruses spread between distinct cell types. Using a measles virus (MV) transgenic mouse model that expresses human CD46 (hCD46), the MV vaccine strain receptor, under the control of a neuron-specific enolase promoter (NSE-hCD46+ mice), a novel mode of viral spread between neurons and astrocytes was identified. Although hCD46 is required for initial neuronal infection, it is dispensable for heterotypic spread to astrocytes, which instead depends on glutamate transporters and direct neuron-astrocyte contact. Moreover, in the presence of RNase A, astrocyte infection is reduced, suggesting that nonenveloped ribonucleoproteins (RNP) may cross the neuron-astrocyte synaptic cleft. The characterization of this novel mode of intercellular transport offers insights into the unique interaction of neurons and glia and may reveal therapeutic targets to mitigate the life-threatening consequences of measles encephalitis.IMPORTANCE Viruses are the most important cause of infectious encephalitis in mammals worldwide; several thousand people, primarily the very young and the elderly, are impacted annually, and few therapies are reliably successful once neuroinvasion has occurred. To understand how viruses contribute to neuropathology, and to develop tools to prevent or ameliorate such infections, it is crucial to define if and how viruses disseminate among the different cell populations within the highly complex central nervous system. This study defines a noncanonical mode of viral transmission between neurons and astrocytes within the brain.


Subject(s)
Astrocytes/virology , Measles Vaccine/analysis , Measles virus/physiology , Neurons/virology , Animals , Cells, Cultured , Disease Models, Animal , Encephalitis, Viral/virology , Female , Humans , Male , Membrane Cofactor Protein/genetics , Mice , Mice, Transgenic
16.
Virology ; 563: 38-43, 2021 11.
Article in English | MEDLINE | ID: mdl-34416448

ABSTRACT

BST2/tetherin is a transmembrane protein with antiviral activity; it is synthesized following exposure to interferons, and restricts the release of budding virus particles by tethering them to the host cell membrane. We previously showed that BST2 is induced in primary neurons following measles virus (MV) infection or type I interferon; however, BST2 was dispensable for protection against challenge with neuron-restricted MV. Here, we define the contribution of BST-2 in neuronal MV infection. Surprisingly, and in contrast to its antiviral role in non-neuronal cells, murine BST2 promotes MV infection in brains of permissive mice and in primary neuron cultures. Moreover, BST2 expression was predominantly observed in the non-synaptic fraction of purified neurons. These studies highlight a cell-type dependent role of a well-characterized antiviral protein in enhancing neuronal infection.


Subject(s)
Antigens, CD/metabolism , Measles virus/physiology , Membrane Glycoproteins/metabolism , Neurons/virology , Animals , Antigens, CD/genetics , Brain/metabolism , Brain/virology , Gene Expression Regulation , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neurons/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Synapses
17.
PLoS One ; 16(2): e0244334, 2021.
Article in English | MEDLINE | ID: mdl-33544724

ABSTRACT

Wild elephant populations are declining rapidly due to rampant killing for ivory and body parts, range fragmentation, and human-elephant conflict. Wild and captive elephants are further impacted by viruses, including highly pathogenic elephant endotheliotropic herpesviruses. Moreover, while the rich genetic diversity of the ancient elephant lineage is disappearing, elephants, with their low incidence of cancer, have emerged as a surprising resource in human cancer research for understanding the intrinsic cellular response to DNA damage. However, studies on cellular resistance to transformation and herpesvirus reproduction have been severely limited, in part due to the lack of established elephant cell lines to enable in vitro experiments. This report describes creation of a recombinant plasmid, pAelPyV-1-Tag, derived from a wild isolate of African Elephant Polyomavirus (AelPyV-1), that can be used to create immortalized lines of elephant cells. This isolate was extracted from a trunk nodule biopsy isolated from a wild African elephant, Loxodonta africana, in Botswana. The AelPyV-1 genome contains open-reading frames encoding the canonical large (LTag) and small (STag) tumor antigens. We cloned the entire early region spanning the LTag and overlapping STag genes from this isolate into a high-copy vector to construct a recombinant plasmid, pAelPyV-1-Tag, which effectively transformed primary elephant endothelial cells. We expect that the potential of this reagent to transform elephant primary cells will, at a minimum, facilitate study of elephant-specific herpesviruses.


Subject(s)
Antigens, Viral, Tumor/genetics , Genome, Viral , Polyomavirus Infections/veterinary , Polyomavirus/isolation & purification , Tumor Virus Infections/veterinary , Animals , Animals, Wild , Elephants , Endothelial Cells/virology , Polyomavirus Infections/diagnosis , Tumor Virus Infections/diagnosis
18.
J Neuroimmunol ; 192(1-2): 145-56, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18006082

ABSTRACT

As immune responses in the CNS are highly regulated, cell-specific differences in IFNgamma signaling may be integral in dictating the outcome of host cell responses. In comparing the response of IFNgamma-treated primary neurons to control MEF, we observed that neurons demonstrated lower basal expression of both STAT1 and STAT3, the primary signal transducers responsible for IFNgamma signaling. Following IFNgamma treatment of these cell populations, we noted muted and delayed STAT1 phosphorylation, no detectable STAT3 phosphorylation, and a 3-10-fold lower level of representative IFNgamma-responsive gene transcripts. Moreover, in response to a brief pulse of IFNgamma, a steady increase in STAT1 phosphorylation and IFNgamma gene expression over 48 h was observed in neurons, as compared to rapid attenuation in MEF. These distinct response kinetics in IFNgamma-stimulated neurons may reflect modifications in the IFNgamma negative feedback loop, which may provide a mechanism for the cell-specific heterogeneity of responses to IFNgamma.


Subject(s)
Gene Expression Regulation/drug effects , Interferon-gamma/pharmacology , Neurons/drug effects , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Drug Administration Schedule , Embryo, Mammalian , Fibroblasts/drug effects , Hippocampus/cytology , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interleukin-6/pharmacology , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Time Factors
19.
J Neuroimmunol ; 308: 25-29, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28187911

ABSTRACT

Central nervous system consequences of viral infections are rare, but when they do occur, they are often serious and clinically challenging to manage. Our awareness of the perils of neuroinvasion by viruses is growing: the recently appreciated impact of Ebola and Zika virus infections on CNS integrity, decreases in vaccination coverage for potentially neurotropic viruses such as measles, and increased neurovirulence of some influenza strains collectively highlight the need for a better understanding of the viral-neural interaction. Defining these interactions and how they result in neuropathogenesis is paramount for the development of better clinical strategies, especially given the limited treatment options that are available due to the unique physiology of the brain that limits migration of blood-borne molecules into the CNS parenchyma. In this perspective, we discuss some unique aspects of neuronal viral infections and immune-mediated control that impact the pathogenic outcomes of these infections. Further, we draw attention to an often overlooked aspect of neuropathogenesis research: that lack of overt disease, which is often equated with survival post-infection, likely only scratches the surface of the myriad ways by which neurotropic infections can impair CNS function.


Subject(s)
Central Nervous System Viral Diseases/mortality , Central Nervous System/pathology , Kaplan-Meier Estimate , Animals , Central Nervous System/virology , Central Nervous System Viral Diseases/genetics , Disease Models, Animal , Humans , Interferon-gamma/deficiency , Interferon-gamma/genetics , Membrane Cofactor Protein/deficiency , Membrane Cofactor Protein/genetics , Mice , Mice, Transgenic , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics
20.
ACS Chem Neurosci ; 7(4): 435-41, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26854733

ABSTRACT

The central nervous system (CNS) coordinates all aspects of life, autonomic and sentient, though how it has evolved to contend with pathogenic infections remains, to a great degree, a mystery. The skull and cerebrospinal fluid (CSF) provide protection from blunt force contacts, and it was once thought that the blood-brain barrier (BBB) was a fortress that restricted pathogen entry and limited inflammation. Recent studies, however, have caused a revision of this viewpoint: the CNS is monitored by blood-borne lymphocytes, but can use alternative strategies to prevent or resolve many pathogenic challenges. In this Review, we discuss emerging principles that indicate how the CNS is immunologically unique from peripheral tissues. We focus on developments that include glymphatics, recently characterized brain lymphatic vessels, distinctions in innate and adaptive immune strategies, novel points of entry for neurotropic viruses, and, finally, how the periphery can influence CNS homeostasis and immune responses within the brain. Collectively, these attributes demand a re-evaluation of immunity in the brain: not privileged, but distinct.


Subject(s)
Brain , Central Nervous System Viral Diseases , Neuroimmunomodulation/physiology , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Brain/immunology , Brain/pathology , Brain/virology , Central Nervous System Viral Diseases/immunology , Central Nervous System Viral Diseases/pathology , Humans , Inflammation , Lymphatic System/immunology , Lymphatic System/metabolism , Lymphatic System/virology , Neurons/classification , Neurons/immunology , Neurons/virology
SELECTION OF CITATIONS
SEARCH DETAIL