Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Front Immunol ; 12: 748423, 2021.
Article in English | MEDLINE | ID: mdl-34691060

ABSTRACT

The Type I Interferon family of cytokines all act through the same cell surface receptor and induce phosphorylation of the same subset of response regulators of the STAT family. Despite their shared receptor, different Type I Interferons have different functions during immune response to infection. In particular, they differ in the potency of their induced anti-viral and anti-proliferative responses in target cells. It remains not fully understood how these functional differences can arise in a ligand-specific manner both at the level of STAT phosphorylation and the downstream function. We use a minimal computational model of Type I Interferon signaling, focusing on Interferon-α and Interferon-ß. We validate the model with quantitative experimental data to identify the key determinants of specificity and functional plasticity in Type I Interferon signaling. We investigate different mechanisms of signal discrimination, and how multiple system components such as binding affinity, receptor expression levels and their variability, receptor internalization, short-term negative feedback by SOCS1 protein, and differential receptor expression play together to ensure ligand specificity on the level of STAT phosphorylation. Based on these results, we propose phenomenological functional mappings from STAT activation to downstream anti-viral and anti-proliferative activity to investigate differential signal processing steps downstream of STAT phosphorylation. We find that the negative feedback by the protein USP18, which enhances differences in signaling between Interferons via ligand-dependent refractoriness, can give rise to functional plasticity in Interferon-α and Interferon-ß signaling, and explore other factors that control functional plasticity. Beyond Type I Interferon signaling, our results have a broad applicability to questions of signaling specificity and functional plasticity in signaling systems with multiple ligands acting through a bottleneck of a small number of shared receptors.


Subject(s)
Interferon-alpha/physiology , Interferon-beta/physiology , Models, Immunological , Receptor Cross-Talk/physiology , Receptor, Interferon alpha-beta/physiology , Signal Transduction/physiology , Animals , Computer Simulation , Dimerization , Feedback, Physiological , Female , Humans , Inhibitory Concentration 50 , Kinetics , Ligands , Mice , Mice, Inbred C57BL , Protein Binding , Protein Interaction Mapping , STAT Transcription Factors/metabolism , Spleen/cytology , Suppressor of Cytokine Signaling 1 Protein/physiology , T-Lymphocytes/immunology , Ubiquitin Thiolesterase
2.
Commun Biol ; 4(1): 508, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33927339

ABSTRACT

Immune evasion of pathogens can modify the course of infection and impact viral persistence and pathology. Here, using different strains of the lymphocytic choriomeningitis virus (LCMV) model system, we show that slower propagation results in limited type I interferon (IFN-I) production and viral persistence. Specifically, cells infected with LCMV-Docile exhibited reduced viral replication when compared to LCMV-WE and as a consequence, infection with LCMV-Docile resulted in reduced activation of bone marrow derived dendritic cells (BMDCs) and IFN-I production in vitro in comparison with LCMV-WE. In vivo, we observed a reduction of IFN-I, T cell exhaustion and viral persistence following infection of LCMV-Docile but not LCMV-WE. Mechanistically, block of intracellular protein transport uncovered reduced propagation of LCMV-Docile when compared to LCMV-WE. This reduced propagation was critical in blunting the activation of the innate and adaptive immune system. When mice were simultaneously infected with LCMV-Docile and LCMV-WE, immune function was restored and IFN-I production, T cell effector functions as well as viral loads were similar to that of mice infected with LCMV-WE alone. Taken together, this study suggests that reduced viral propagation can result in immune evasion and viral persistence.


Subject(s)
Arenaviridae Infections/virology , Dendritic Cells/virology , Lymphocytic choriomeningitis virus/immunology , Receptor, Interferon alpha-beta/physiology , T-Lymphocytes/virology , Virus Replication , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
Nat Med ; 27(4): 668-676, 2021 04.
Article in English | MEDLINE | ID: mdl-33837377

ABSTRACT

Drug repurposing provides a rapid approach to meet the urgent need for therapeutics to address COVID-19. To identify therapeutic targets relevant to COVID-19, we conducted Mendelian randomization analyses, deriving genetic instruments based on transcriptomic and proteomic data for 1,263 actionable proteins that are targeted by approved drugs or in clinical phase of drug development. Using summary statistics from the Host Genetics Initiative and the Million Veteran Program, we studied 7,554 patients hospitalized with COVID-19 and >1 million controls. We found significant Mendelian randomization results for three proteins (ACE2, P = 1.6 × 10-6; IFNAR2, P = 9.8 × 10-11 and IL-10RB, P = 2.3 × 10-14) using cis-expression quantitative trait loci genetic instruments that also had strong evidence for colocalization with COVID-19 hospitalization. To disentangle the shared expression quantitative trait loci signal for IL10RB and IFNAR2, we conducted phenome-wide association scans and pathway enrichment analysis, which suggested that IFNAR2 is more likely to play a role in COVID-19 hospitalization. Our findings prioritize trials of drugs targeting IFNAR2 and ACE2 for early management of COVID-19.


Subject(s)
COVID-19/genetics , Drug Repositioning , Mendelian Randomization Analysis/methods , SARS-CoV-2 , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/physiology , Genome-Wide Association Study , Humans , Interleukin-10 Receptor beta Subunit/genetics , Interleukin-10 Receptor beta Subunit/physiology , Quantitative Trait Loci , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/physiology , COVID-19 Drug Treatment
4.
PLoS Pathog ; 16(12): e1009163, 2020 12.
Article in English | MEDLINE | ID: mdl-33326500

ABSTRACT

The novel human coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a pandemic. Critical to the rapid evaluation of vaccines and antivirals against SARS-CoV-2 is the development of tractable animal models to understand the adaptive immune response to the virus. To this end, the use of common laboratory strains of mice is hindered by significant divergence of the angiotensin-converting enzyme 2 (ACE2), which is the receptor required for entry of SARS-CoV-2. In the current study, we designed and utilized an mRNA-based transfection system to induce expression of the hACE2 receptor in order to confer entry of SARS-CoV-2 in otherwise non-permissive cells. By employing this expression system in an in vivo setting, we were able to interrogate the adaptive immune response to SARS-CoV-2 in type 1 interferon receptor deficient mice. In doing so, we showed that the T cell response to SARS-CoV-2 is enhanced when hACE2 is expressed during infection. Moreover, we demonstrated that these responses are preserved in memory and are boosted upon secondary infection. Importantly, using this system, we functionally identified the CD4+ and CD8+ structural peptide epitopes targeted during SARS-CoV-2 infection in H2b restricted mice and confirmed their existence in an established model of SARS-CoV-2 pathogenesis. We demonstrated that, identical to what has been seen in humans, the antigen-specific CD8+ T cells in mice primarily target peptides of the spike and membrane proteins, while the antigen-specific CD4+ T cells target peptides of the nucleocapsid, membrane, and spike proteins. As the focus of the immune response in mice is highly similar to that of the humans, the identification of functional murine SARS-CoV-2-specific T cell epitopes provided in this study will be critical for evaluation of vaccine efficacy in murine models of SARS-CoV-2 infection.


Subject(s)
Adaptive Immunity/immunology , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/immunology , RNA, Messenger/metabolism , SARS-CoV-2/immunology , T-Lymphocytes/immunology , Virus Replication , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/metabolism , COVID-19/virology , Chlorocebus aethiops , Epitopes, T-Lymphocyte/immunology , Humans , Mice , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , Receptor, Interferon alpha-beta/physiology , T-Lymphocytes/virology , Vero Cells
5.
PLoS Pathog ; 16(10): e1008951, 2020 10.
Article in English | MEDLINE | ID: mdl-33052957

ABSTRACT

Both mosquito species-specific differences and virus strain -specific differences impact vector competence. Previous results in our laboratory with individual populations of N. American mosquitoes support studies suggesting Aedes aegypti are more competent than Ae. albopictus for American Zika virus (ZIKV) strains and demonstrate that U.S. Ae. albopictus have higher competence for an ancestral Asian ZIKV strain. A982V, an amino acid substitution in the NS1 gene acquired prior to the American outbreak, has been shown to increase competence in Ae. aegypti. We hypothesized that variability in the NS1 could therefore contribute to species-specific differences and developed a reverse genetics system based on a 2016 ZIKV isolate from Honduras (ZIKV-WTic) to evaluate the phenotypic correlates of individual amino acid substitutions. In addition to A982V, we evaluated G894A, which was acquired during circulation in the Americas. Reversion of 982 and 894 to ancestral residues increased infectivity, transmissibility and viral loads in Ae. albopictus but had no effect on competence or replication in Ae. aegypti. In addition, while host cell-specific differences in NS1 secretion were measured, with significantly higher secretion in mammalian cells relative to mosquito cells, strain-specific differences in secretion were not detected, despite previous reports. These results demonstrate that individual mutations in NS1 can influence competence in a species-specific manner independent of differences in NS1 secretion and further indicate that ancestral NS1 residues confer increased competence in Ae. albopictus. Lastly, experimental infections of Ifnar1-/- mice demonstrated that these NS1 substitutions can influence viral replication in the host and, specifically, that G894A could represent a compensatory change following a fitness loss from A982V with some viral genetic backgrounds. Together these data suggest a possible role for epistatic interactions in ZIKV fitness in invertebrate and vertebrate hosts and demonstrate that strains with increased transmission potential in U.S. Ae. albopictus could emerge.


Subject(s)
Aedes/virology , Host-Pathogen Interactions , Mosquito Vectors/virology , Viral Load , Viral Nonstructural Proteins/genetics , Zika Virus Infection/transmission , Zika Virus Infection/virology , Animals , Chlorocebus aethiops , Female , Mice , Mice, Knockout , Mutation , Receptor, Interferon alpha-beta/physiology , Vero Cells , Viral Nonstructural Proteins/metabolism , Virus Replication , Zika Virus/classification , Zika Virus/genetics
6.
PLoS Pathog ; 16(10): e1008973, 2020 10.
Article in English | MEDLINE | ID: mdl-33045014

ABSTRACT

The liver is a central regulator of metabolic homeostasis and serum metabolite levels. Hepatocytes are the functional units of the liver parenchyma and not only responsible for turnover of biomolecules but also act as central immune signaling platforms. Hepatotropic viruses infect liver tissue, resulting in inflammatory responses, tissue damage and hepatitis. Combining well-established in vitro and in vivo model systems with transcriptomic analyses, we show that type I interferon signaling initiates a robust antiviral immune response in hepatocytes. Strikingly, we also identify IFN-I as both, sufficient and necessary, to induce wide-spread metabolic reprogramming in hepatocytes. IFN-I specifically rewired tryptophan metabolism and induced hepatic tryptophan oxidation to kynurenine via Tdo2, correlating with altered concentrations of serum metabolites upon viral infection. Infected Tdo2-deficient animals displayed elevated serum levels of tryptophan and, unexpectedly, also vast increases in the downstream immune-suppressive metabolite kynurenine. Thus, Tdo2-deficiency did not result in altered serum homeostasis of the tryptophan to kynurenine ratio during infection, which seemed to be independent of hepatocyte-intrinsic compensation via the IDO-axis. These data highlight that inflammation-induced reprogramming of systemic tryptophan metabolism is tightly regulated in viral hepatitis.


Subject(s)
Antiviral Agents/metabolism , Hepatitis, Viral, Animal/immunology , Hepatocytes/immunology , Inflammation/immunology , Kynurenine/metabolism , Receptor, Interferon alpha-beta/physiology , Tryptophan/metabolism , Animals , Female , Hepatitis Viruses/isolation & purification , Hepatitis, Viral, Animal/metabolism , Hepatitis, Viral, Animal/virology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Immunity, Innate/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Interferon Regulatory Factor-7/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT1 Transcription Factor/physiology , Tryptophan Oxygenase/physiology
7.
Eur J Immunol ; 50(8): 1142-1153, 2020 08.
Article in English | MEDLINE | ID: mdl-32135578

ABSTRACT

Exposure to diesel exhaust particles (DEPs) is associated with acute inflammatory responses in the lung and exacerbation of respiratory diseases. However, the mechanism by which DEPs trigger the inflammatory responses remains unclear. Here, we demonstrated that the IFN response factors IRF3 and IRF7 played pivotal roles in DEP-induced pulmonary inflammation. DEPs could not directly induce inflammatory cytokine expression in mouse cells, whereas DEPs triggered autophagy both in vitro and in vivo. The DEP-induced autophagy was augmented in the absence of IRF3 and IRF7, but not in the absence of IFNAR. The expression of Raptor was induced by IRF3 and IRF7 in response to DEPs treatment. Furthermore, administration of the mechanistic target of rapamycin (mTOR) inhibitor alleviated the inflammatory responses in the lung during DEP exposure. Our findings define an IFNAR-independent role of increased autophagy in the absence of IRF3 and IRF7 during pulmonary DEP exposure, and provide the basis to develop new therapeutic approaches to counteract the adverse effects of DEPs and possibly other ambient particulate matters.


Subject(s)
Autophagy/physiology , Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Pneumonia/etiology , Vehicle Emissions/toxicity , Animals , Cytokines/biosynthesis , Mice , Mice, Inbred C57BL , Receptor, Interferon alpha-beta/physiology , Sirolimus/pharmacology
8.
Mol Cancer Ther ; 19(6): 1232-1242, 2020 06.
Article in English | MEDLINE | ID: mdl-32188623

ABSTRACT

Metastatic cancer involving spread to the peritoneal cavity is referred to as peritoneal carcinomatosis and has a very poor prognosis. Our previous study demonstrated a Toll-like receptor and C-type lectin receptor agonist pairing of monophosphoryl lipid A (MPL) and trehalose-6,6'-dicorynomycolate (TDCM) effectively inhibits tumor growth and ascites development following TA3-Ha and EL4 challenge through a mechanism dependent on B-1a cell-produced natural IgM and complement. In this study, we investigated additional players in the MPL/TDCM-elicited response. MPL/TDCM treatment rapidly increased type I IFN levels in the peritoneal cavity along with myeloid cell numbers, including macrophages and Ly6Chi monocytes. Type I IFN receptor (IFNAR1-/-) mice produced tumor-reactive IgM following MPL/TDCM treatment, but failed to recruit Ly6C+ monocytes and were not afforded protection during tumor challenges. Clodronate liposome depletion of phagocytic cells, as well as targeted depletion of Ly6C+ cells, also ablated MPL/TDCM-induced protection. Cytotoxic mediators known to be produced by these cells were required for effects. TNFα was required for effective TA3-Ha killing and nitric oxide was required for EL4 killing. Collectively, these data reveal a model whereby MPL/TDCM-elicited antitumor effects strongly depend on innate cell responses, with B-1a cell-produced tumor-reactive IgM and complement pairing with myeloid cell-produced cytotoxic mediators to effectively eradicate tumors in the peritoneal cavity.


Subject(s)
Antigens, Ly/metabolism , Cord Factors/pharmacology , Interferon Type I/metabolism , Lectins, C-Type/agonists , Lipid A/analogs & derivatives , Peritoneal Neoplasms/drug therapy , Toll-Like Receptors/agonists , Adjuvants, Immunologic/pharmacology , Animals , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Drug Therapy, Combination , Female , Gene Expression Regulation, Neoplastic , Humans , Lipid A/pharmacology , Lymphoma/drug therapy , Lymphoma/metabolism , Lymphoma/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Phagocytes , Receptor, Interferon alpha-beta/physiology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Am J Pathol ; 190(4): 844-861, 2020 04.
Article in English | MEDLINE | ID: mdl-32035058

ABSTRACT

Zika virus (ZIKV) is a reemerging human pathogen that causes congenital abnormalities, including microcephaly and eye disease. The cellular/molecular basis of ZIKV and host interactions inducing ocular and neuronal pathogenesis are unclear. Herein, we noted that the Hippo/Salvador-Warts-Hippo signaling pathway, which controls organ size through progenitor cell proliferation and differentiation, is dysregulated after ZIKV infection. In human fetal retinal pigment epithelial cells, there is an early induction of transcriptional coactivator, Yes-associated protein (YAP), which is later degraded with a corresponding activation of the TANK binding kinase 1/interferon regulatory factor 3 type I interferon pathway. YAP/transcriptional co-activator with a PDZ-binding domain (TAZ) silencing results in reduced ZIKV replication, indicating a direct role of Hippo pathway in regulating ZIKV infection. Using an in vivo Ifnar1-/- knockout mouse model, ZIKV infection was found to reduce YAP/TAZ protein levels while increasing phosphorylated YAP Ser127 in the retina and brain. Hippo pathway is activated in major cellular components of the blood-brain barrier, including endothelial cells and astrocytes. In addition, this result suggests AMP-activated protein kinase signaling pathway's role in regulating YAP/TAZ in ZIKV-infected cells. These data demonstrate that ZIKV infection might initiate a cross talk among AMP-activated protein kinase-Hippo-TBK1 pathways, which could regulate antiviral and energy stress responses during oculoneuronal inflammation.


Subject(s)
Inflammation/pathology , Neurodegenerative Diseases/pathology , Protein Serine-Threonine Kinases/metabolism , Receptor, Interferon alpha-beta/physiology , Virus Replication , Zika Virus Infection/complications , Zika Virus/isolation & purification , Animals , Hippo Signaling Pathway , Inflammation/virology , Male , Mice , Mice, Knockout , Neurodegenerative Diseases/virology , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Zika Virus Infection/virology
10.
Cell Immunol ; 349: 104043, 2020 03.
Article in English | MEDLINE | ID: mdl-32044112

ABSTRACT

Type I Interferon (IFN) signaling plays a critical role in dendritic cell (DC) development and functions. Inhibition of hyper type I IFN signaling promotes cDC2 subtype development. Relb is essential to development of cDC2 subtype and here we analyzed its effect on type I IFN signaling in DCs. We show that Relb suppresses the homeostatic type I IFN signaling in cDC2 cultures. TLR stimulation of FL-DCs led to RelB induction coinciding with fall in IFN signatures; conforming with the observation Relb expression reduced TLR stimulated IFN induction along with decrease in ISGs. Towards understanding mechanism, we show that effects of RelB are mediated by increased levels of IκBα. We demonstrate that RelB dampened antiviral responses by lowering ISG levels and the defect in cDC2 development in RelB null mice can be rescued in Ifnar1-/- background. Overall, we propose a novel role of RelB as a negative regulator of the type I IFN signaling pathway; fine tuning development of cDC2 subtype.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , NF-KappaB Inhibitor alpha/physiology , Transcription Factor RelB/physiology , Amino Acid Sequence , Animals , Cell Differentiation , Cells, Cultured , Crosses, Genetic , Dendritic Cells/classification , Dendritic Cells/cytology , Gene Expression Regulation/immunology , Mice , NIH 3T3 Cells , Newcastle disease virus/immunology , Peptides/pharmacology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/physiology , Signal Transduction/immunology , Spleen/cytology , Transcription Factor RelB/deficiency , Transcription Factor RelB/genetics , Viral Load
11.
J Leukoc Biol ; 108(3): 909-924, 2020 09.
Article in English | MEDLINE | ID: mdl-33448473

ABSTRACT

The type I IFNs activate an array of signaling pathways, which are initiated after IFNs bind their cognate receptors, IFNα/ß receptor (IFNAR)1 and IFNAR2. These signals contribute to many aspects of human health including defense against pathogens, cancer immunosurveillance, and regulation of inflammation. How these cytokines interact with their receptors influences the quality of these signals. As such, the integrity of receptor structure is pivotal to maintaining human health and the response to immune stimuli. This review brings together genome wide association studies and clinical reports describing the association of nonsynonymous IFNAR1 and IFNAR2 polymorphisms with clinical disease, including altered susceptibility to viral and bacterial pathogens, autoimmune diseases, cancer, and adverse reactions to live-attenuated vaccines. We describe the amino acid substitutions or truncations induced by these polymorphisms and, using the knowledge of IFNAR conformational changes, IFNAR-IFN interfaces and overall structure-function relationship of the signaling complexes, we hypothesize the effect of these polymorphisms on receptor structure. That these predicted changes to IFNAR structure are associated with clinical manifestations of human disease, highlights the importance of IFNAR structural integrity to maintaining functional quality of these receptor-mediated responses. Type I IFNs are pivotal to innate immune responses and ultimately, to human health. Understanding the consequences of altered structure on the actions of these clinically significant cell receptors provides important information on the roles of IFNARs in health and disease.


Subject(s)
Polymorphism, Single Nucleotide , Receptor, Interferon alpha-beta/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Codon, Nonsense/genetics , Crystallography, X-Ray , Disease Susceptibility , Humans , Immunity, Innate , Immunogenicity, Vaccine , Ligands , Macrophages/immunology , Mammals/genetics , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Receptor, Interferon alpha-beta/chemistry , Receptor, Interferon alpha-beta/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Structure-Activity Relationship , Tuberculosis/immunology
12.
Mol Ther ; 27(12): 2067-2079, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31653397

ABSTRACT

Zika virus (ZIKV) infection is associated with microcephaly in neonates and Guillain-Barré syndrome in adults. ZIKV produces a class of nonstructural (NS) regulatory proteins that play a critical role in viral transcription and replication, including NS5, which possesses RNA-dependent RNA polymerase (RdRp) activity. Here we demonstrate that rilpivirine (RPV), a non-nucleoside reverse transcriptase inhibitor (NNRTI) used in the treatment of HIV-1 infection, inhibits the enzymatic activity of NS5 and suppresses ZIKV infection and replication in primary human astrocytes. Similarly, other members of the NNRTI family, including etravirine and efavirenz, showed inhibitory effects on viral infection of brain cells. Site-directed mutagenesis identified 14 amino acid residues within the NS5 RdRp domain (AA265-903), which are important for the RPV interaction and the inhibition of NS5 polymerase activity. Administration of RPV to ZIKV-infected interferon-alpha/beta receptor (IFN-A/R) knockout mice improved the clinical outcome and prevented ZIKV-induced mortality. Histopathological examination of the brains from infected animals revealed that RPV reduced ZIKV RNA levels in the hippocampus, frontal cortex, thalamus, and cerebellum. Repurposing of NNRTIs, such as RPV, for the inhibition of ZIKV replication offers a possible therapeutic strategy for the prevention and treatment of ZIKV-associated disease.


Subject(s)
Anti-HIV Agents/pharmacology , Brain/drug effects , Receptor, Interferon alpha-beta/physiology , Rilpivirine/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus Infection/drug therapy , Zika Virus/drug effects , Animals , Brain/virology , Humans , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Conformation , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication , Zika Virus Infection/pathology , Zika Virus Infection/virology
13.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31413130

ABSTRACT

Dengue virus (DENV) infection causes serious clinical symptoms, including dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Vascular permeability change is the main feature of the diseases, and the abnormal expression of proinflammatory cytokines is the important cause of vascular permeability change. However, the mechanism underlying vascular permeability induced by DENV has not been fully elucidated. Here, we reveal a distinct mechanism by which DENV infection promotes NLRP3 inflammasome activation and interleukin-1 beta (IL-1ß) release to induce endothelial permeability and vascular leakage in mice. DENV M protein interacts with NLRP3 to facilitate NLRP3 inflammasome assembly and activation, which induce proinflammatory cytokine IL-1ß activation and release. Notably, M can induce vascular leakage in mouse tissues by activating the NLRP3 inflammasome and IL-1ß. More importantly, inflammatory cell infiltration and tissue injuries are induced by M in wild-type (WT) mouse tissues, but they are not affected by M in NLRP3 knockout (NLRP3-/-) mouse tissues. Evans blue intensities in WT mouse tissues are significantly higher than in NLRP3-/- mouse tissues, demonstrating an essential role of NLRP3 in M-induced vascular leakages in mice. Therefore, we propose that upon DENV infection, M interacts with NLRP3 to facilitate inflammasome activation and IL-1ß secretion, which lead to the induction of endothelial permeability and vascular leakage in mouse tissues. The important role of the DENV-M-NLRP3-IL-1ß axis in the induction of vascular leakage provides new insights into the mechanisms underlying DENV pathogenesis and DENV-associated DHF and DSS development.IMPORTANCE Dengue virus (DENV) is a mosquito-borne pathogen, and infections by this virus are prevalent in over 100 tropical and subtropical countries or regions, with approximately 2.5 billion people at risk. DENV infection induces a spectrum of clinical symptoms, ranging from classical dengue fever (DF) to severe dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Therefore, it is important to understand the mechanisms underlying DENV pathogenesis. In this study, we reveal that the DENV membrane protein (M) interacts with the host NLRP3 protein to promote NLRP3 inflammasome activation, which leads to the activation and release of a proinflammatory cytokine, interleukin-1 beta (IL-1ß). More importantly, we demonstrate that M protein can induce vascular permeability and vascular leakage and that NLRP3 is required for M-induced vascular leakage in mouse tissues. Collectively, this study reveals a distinct mechanism underlying DENV pathogeneses and provides new insights into the development of therapeutic agents for DENV-associated diseases.


Subject(s)
Dengue Virus/immunology , Dengue/immunology , Endothelium, Vascular/immunology , Inflammasomes/immunology , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Viral Matrix Proteins/metabolism , Animals , Capillary Permeability , Cells, Cultured , Dengue/pathology , Dengue/virology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Inflammasomes/metabolism , Interleukin-1beta/genetics , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/physiology , Viral Matrix Proteins/genetics
14.
Cell Rep ; 27(12): 3618-3628.e5, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31216479

ABSTRACT

Zika virus (ZIKV) infection is implicated in severe fetal developmental disorders, including microcephaly. MicroRNAs (miRNAs) post-transcriptionally regulate numerous processes associated with viral infection and neurodegeneration, but their contribution to ZIKV pathogenesis is unclear. We analyzed the mRNA and miRNA transcriptomes of human neuronal stem cells (hNSCs) during infection with ZIKV MR766 and Paraiba strains. Integration of the miRNA and mRNA expression data into regulatory interaction networks showed that ZIKV infection resulted in miRNA-mediated repression of genes regulating the cell cycle, stem cell maintenance, and neurogenesis. Bioinformatics analysis of Argonaute-bound RNAs in ZIKV-infected hNSCs identified a number of miRNAs with predicted involvement in microcephaly, including miR-124-3p, which dysregulates NSC maintenance through repression of the transferrin receptor (TFRC). Consistent with this, ZIKV infection upregulated miR-124-3p and downregulated TFRC mRNA in ZIKV-infected hNSCs and mouse brain tissue. These data provide insights into the roles of miRNAs in ZIKV pathogenesis, particularly the microcephaly phenotype.


Subject(s)
Antigens, CD/metabolism , Cell Cycle , MicroRNAs/genetics , Microcephaly/pathology , Neural Stem Cells/metabolism , Neurogenesis , Receptors, Transferrin/metabolism , Zika Virus Infection/pathology , Animals , Antigens, CD/genetics , Genome , Humans , Mice , Mice, Knockout , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/virology , Neural Stem Cells/pathology , Neural Stem Cells/virology , Receptor, Interferon alpha-beta/physiology , Receptors, Transferrin/genetics , Transcriptome , Zika Virus/isolation & purification , Zika Virus Infection/genetics , Zika Virus Infection/metabolism , Zika Virus Infection/virology
15.
Cell Rep ; 27(9): 2649-2664.e5, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31141689

ABSTRACT

Lung inflammation induced by silica impairs host control of tuberculosis, yet the underlying mechanism remains unclear. Here, we show that silica-driven exacerbation of M. tuberculosis infection associates with raised type 2 immunity. Silica increases pulmonary Th2 cell and M2 macrophage responses, while reducing type 1 immunity after M. tuberculosis infection. Silica induces lung damage that prompts extracellular self-DNA release and activates STING. This STING priming potentiates M. tuberculosis DNA sensing by and activation of cGAS/STING, which triggers enhanced type I interferon (IFNI) response and type 2 immunity. cGAS-, STING-, and IFNAR-deficient mice are resistant to silica-induced exacerbation of M. tuberculosis infection. Thus, silica-induced self-DNA primes the host response to M. tuberculosis-derived nucleic acids, which increases type 2 immunity while reducing type 1 immunity, crucial for controlling M. tuberculosis infection. These data show how cGAS/STING pathway activation, at the crossroads of sterile inflammation and infection, may affect the host response to pathogens such as M. tuberculosis.


Subject(s)
Host-Pathogen Interactions/immunology , Immunity, Innate/immunology , Membrane Proteins/physiology , Mycobacterium tuberculosis/immunology , Pneumonia/complications , Silicon Dioxide/toxicity , Tuberculosis/etiology , Animals , Dendritic Cells , Interferon Regulatory Factor-3/physiology , Interferon Type I/metabolism , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleotidyltransferases/physiology , Pneumonia/chemically induced , Receptor, Interferon alpha-beta/physiology , Signal Transduction , Tuberculosis/metabolism , Tuberculosis/pathology
16.
J Virol ; 93(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30429338

ABSTRACT

The impact of the Zika virus (ZIKV) epidemic highlights the need for vaccines that reduce or prevent infection and reliably prevent teratogenic complications. The live-attenuated measles virus (MV) vaccine strains are a promising vaccine platform, since they induce robust humoral and cellular immune responses against additional antigens and have an excellent safety record. To explore its potential to protect against ZIKV, we compared a recombinant Schwarz strain MV that encodes ZIKV prM and soluble E proteins (MV-Zika-sE) with a prototypic alum-adjuvanted whole inactivated ZIKV particle vaccine. Analysis of MV-Zika-sE-infected cells confirmed antigen expression, and the virus replicated with vaccine strain characteristics. Immunized IFNAR-/--CD46Ge mice developed E protein-specific and neutralizing antibodies, and ZIKV E-specific cellular immune responses were observed by gamma interferon (IFN-γ) enzyme-linked immunospot (ELISpot) and in vitro T cell proliferation assays. To analyze protective efficacy, vaccinated female mice were challenged with ZIKV after allogeneic mating. In MV-Zika-sE-vaccinated mice, weight gain was similar to that in uninfected mice, while no plasma viremia was detectable in the majority of the animals. In contrast, infected control animals gained less weight and experienced about 100-fold higher viremia over at least 3 days. Moreover, vaccination with MV-Zika-sE reduced the ZIKV load in different organs and the placentas and prevented infection of the fetus. Consequently, no fetal growth retardation, anemia, or death due to ZIKV infection was seen in MV-Zika-sE-vaccinated dams. In contrast, the inactivated ZIKV vaccine had little to no effect in our studies. Therefore, the MV-derived ZIKV vaccine is a promising candidate for further preclinical and clinical development.IMPORTANCE Zika virus (ZIKV) is a mosquito-borne flavivirus that causes a variety of neurological complications, including congenital birth defects. Despite the urgent need, no ZIKV vaccine has yet been licensed. Recombinant vaccine strain-derived measles viruses (MV) constitute a promising vector platform to induce immunity against foreign pathogens by expressing antigens from additional transcription units while at the same time possessing a remarkable safety profile. This concept has already been validated against different pathogens, including at least 3 other flaviviruses, and our data show that vaccination with MV expressing soluble ZIKV E protein significantly diminishes infection and prevents fetal loss or damage in an allogeneic mouse pregnancy model. It can thus be regarded as a promising emergency vaccine candidate with the potential for inclusion in routine vaccination settings in areas of endemicity to prevent teratogenic effects of circulating ZIKV during pregnancy, comparable to standard rubella virus vaccination.


Subject(s)
Disease Models, Animal , Measles Vaccine/administration & dosage , Measles virus/immunology , Viral Envelope Proteins/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Antibodies, Viral/blood , Female , Genome, Viral , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Measles Vaccine/immunology , Membrane Cofactor Protein/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pregnancy , Receptor, Interferon alpha-beta/physiology , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Zika Virus/genetics , Zika Virus Infection/immunology , Zika Virus Infection/virology
17.
PLoS Pathog ; 14(10): e1007360, 2018 10.
Article in English | MEDLINE | ID: mdl-30372491

ABSTRACT

Secondary bacterial infections contribute to the excess morbidity and mortality of influenza A virus (IAV) infection. Disruption of lung integrity and impaired antibacterial immunity during IAV infection participate in colonization and dissemination of the bacteria out of the lungs. One key feature of IAV infection is the profound alteration of lung myeloid cells, characterized by the recruitment of deleterious inflammatory monocytes. We herein report that IAV infection causes a transient decrease of lung conventional dendritic cells (cDCs) (both cDC1 and cDC2) peaking at day 7 post-infection. While triggering emergency monopoiesis, IAV transiently altered the differentiation of cDCs in the bone marrow, the cDC1-biaised pre-DCs being particularly affected. The impaired cDC differentiation during IAV infection was independent of type I interferons (IFNs), IFN-γ, TNFα and IL-6 and was not due to an intrinsic dysfunction of cDC precursors. The alteration of cDC differentiation was associated with a drop of local and systemic production of Fms-like tyrosine kinase 3 ligand (Flt3-L), a critical cDC differentiation factor. Overexpression of Flt3-L during IAV infection boosted the cDC progenitors' production in the BM, replenished cDCs in the lungs, decreased inflammatory monocytes' infiltration and lowered lung damages. This was associated with partial protection against secondary pneumococcal infection, as reflected by reduced bacterial dissemination and prolonged survival. These findings highlight the impact of distal viral infection on cDC genesis in the BM and suggest that Flt3-L may have potential applications in the control of secondary infections.


Subject(s)
Dendritic Cells/immunology , Influenza A virus/immunology , Lung/immunology , Membrane Proteins/immunology , Orthomyxoviridae Infections/virology , Pneumococcal Infections/immunology , Superinfection/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/microbiology , Dendritic Cells/virology , Lung/microbiology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/complications , Pneumococcal Infections/microbiology , Pneumococcal Infections/virology , Receptor, Interferon alpha-beta/physiology , Streptococcus pneumoniae/immunology
18.
J Virol ; 92(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29643242

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease caused by a tick-borne phlebovirus of the family Bunyaviridae, SFTS virus (SFTSV). Wild-type and type I interferon (IFN-I) receptor 1-deficient (IFNAR1-/-) mice have been established as nonlethal and lethal models of SFTSV infection, respectively. However, the mechanisms of IFN-I production in vivo and the factors causing the lethal disease are not well understood. Using bone marrow-chimeric mice, we found that IFN-I signaling in hematopoietic cells was essential for survival of lethal SFTSV infection. The disruption of IFN-I signaling in hematopoietic cells allowed an increase in viral loads in serum and produced an excess of multiple inflammatory cytokines and chemokines. The production of IFN-I and inflammatory cytokines was abolished by deletion of the signaling molecules IPS-1 and MyD88, essential for retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) and Toll-like receptor (TLR) signaling, respectively. However, IPS-1-/- MyD88-/- mice exhibited resistance to lethal SFTS with a moderate viral load in serum. Taken together, these results indicate that adequate activation of RLR and TLR signaling pathways under low to moderate levels of viremia contributed to survival through the IFN-I-dependent antiviral response during SFTSV infection, whereas overactivation of these signaling pathways under high levels of viremia resulted in abnormal induction of multiple inflammatory cytokines and chemokines, causing the lethal disease.IMPORTANCE SFTSV causes a severe infectious disease in humans, with a high fatality rate of 12 to 30%. To know the pathogenesis of the virus, we need to clarify the innate immune response as a front line of defense against viral infection. Here, we report that a lethal animal model showed abnormal induction of multiple inflammatory cytokines and chemokines by an uncontrolled innate immune response, which triggered the lethal SFTS. Our findings suggest a new strategy to target inflammatory humoral factors to treat patients with severe SFTS. Furthermore, this study may help the investigation of other tick-borne viruses.


Subject(s)
Bunyaviridae Infections/immunology , DEAD Box Protein 58/metabolism , Inflammation Mediators/metabolism , Phlebotomus Fever/immunology , Receptor, Interferon alpha-beta/physiology , Thrombocytopenia/immunology , Toll-Like Receptors/metabolism , Animals , Bunyaviridae Infections/metabolism , Bunyaviridae Infections/virology , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , DEAD Box Protein 58/genetics , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phlebotomus Fever/metabolism , Phlebotomus Fever/virology , Phlebovirus/pathogenicity , Severity of Illness Index , Thrombocytopenia/metabolism , Thrombocytopenia/virology , Toll-Like Receptors/genetics , Viral Load
19.
PLoS Pathog ; 14(4): e1006985, 2018 04.
Article in English | MEDLINE | ID: mdl-29672594

ABSTRACT

Regulatory T cells (Tregs) play a cardinal role in the immune system by suppressing detrimental autoimmune responses, but their role in acute, chronic infectious diseases and tumor microenvironment remains unclear. We recently demonstrated that IFN-α/ß receptor (IFNAR) signaling promotes Treg function in autoimmunity. Here we dissected the functional role of IFNAR-signaling in Tregs using Treg-specific IFNAR deficient (IFNARfl/flxFoxp3YFP-Cre) mice in acute LCMV Armstrong, chronic Clone-13 viral infection, and in tumor models. In both viral infection and tumor models, IFNARfl/flxFoxp3YFP-Cre mice Tregs expressed enhanced Treg associated activation antigens. LCMV-specific CD8+ T cells and tumor infiltrating lymphocytes from IFNARfl/flxFoxp3YFP-Cre mice produced less antiviral and antitumor IFN-γ and TNF-α. In chronic viral model, the numbers of antiviral effector and memory CD8+ T cells were decreased in IFNARfl/flxFoxp3YFP-Cre mice and the effector CD4+ and CD8+ T cells exhibited a phenotype compatible with enhanced exhaustion. IFNARfl/flxFoxp3YFP-Cre mice cleared Armstrong infection normally, but had higher viral titers in sera, kidneys and lungs during chronic infection, and higher tumor burden than the WT controls. The enhanced activated phenotype was evident through transcriptome analysis of IFNARfl/flxFoxp3YFP-Cre mice Tregs during infection demonstrated differential expression of a unique gene signature characterized by elevated levels of genes involved in suppression and decreased levels of genes mediating apoptosis. Thus, IFN signaling in Tregs is beneficial to host resulting in a more effective antiviral response and augmented antitumor immunity.


Subject(s)
Arenaviridae Infections/immunology , Colonic Neoplasms/immunology , Interferon Type I/pharmacology , Lymphocytic Choriomeningitis/immunology , Melanoma, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Animals , Antiviral Agents/pharmacology , Arenaviridae Infections/drug therapy , Arenaviridae Infections/metabolism , Arenaviridae Infections/virology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/virology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Interferon-gamma/metabolism , Lymphocytic Choriomeningitis/drug therapy , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/drug effects , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/physiology , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Tumor Microenvironment/drug effects
20.
Zool Res ; 39(1): 3-14, 2018 Jan 18.
Article in English | MEDLINE | ID: mdl-29511140

ABSTRACT

Due to their inability to generate a complete immune response, mice knockout for type I interferon (IFN) receptors (Ifnar-/-) are more susceptible to viral infections, and are thus commonly used for pathogenesis studies. This mouse model has been used to study many diseases caused by highly pathogenic viruses from many families, including the Flaviviridae, Filoviridae, Arenaviridae, Bunyaviridae, Henipaviridae, and Togaviridae. In this review, we summarize the findings from these animal studies, and discuss the pros and cons of using this model versus other known methods for studying pathogenesis in animals.


Subject(s)
Disease Models, Animal , Disease Outbreaks , Mice, Knockout/genetics , Receptor, Interferon alpha-beta/genetics , Virus Diseases/epidemiology , Animals , Mice , Mice, Knockout/immunology , Receptor, Interferon alpha-beta/physiology , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...