Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Immunity ; 46(5): 875-890.e6, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514692

ABSTRACT

Lambda interferons (IFNλs) or type III IFNs share homology, expression patterns, signaling cascades, and antiviral functions with type I IFNs. This has complicated the unwinding of their unique non-redundant roles. Through the systematic study of influenza virus infection in mice, we herein show that IFNλs are the first IFNs produced that act at the epithelial barrier to suppress initial viral spread without activating inflammation. If infection progresses, type I IFNs come into play to enhance viral resistance and induce pro-inflammatory responses essential for confronting infection but causing immunopathology. Central to this are neutrophils which respond to both cytokines to upregulate antimicrobial functions but exhibit pro-inflammatory activation only to type I IFNs. Accordingly, Ifnlr1-/- mice display enhanced type I IFN production, neutrophilia, lung injury, and lethality, while therapeutic administration of PEG-IFNλ potently suppresses these effects. IFNλs therefore constitute the front line of antiviral defense in the lung without compromising host fitness.


Subject(s)
Genetic Fitness , Host-Pathogen Interactions , Influenza A virus/immunology , Influenza, Human/immunology , Influenza, Human/metabolism , Interferon-gamma/metabolism , Animals , Cluster Analysis , Cytokines/biosynthesis , Disease Models, Animal , Disease Resistance/genetics , Disease Resistance/immunology , Female , Gene Expression , Gene Expression Profiling , Genes, Reporter , Humans , Inflammation Mediators/metabolism , Influenza A virus/genetics , Influenza, Human/drug therapy , Influenza, Human/virology , Interferon-gamma/genetics , Interferon-gamma/pharmacology , Lung/immunology , Lung/metabolism , Lung/pathology , Lung/virology , Male , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Viral Load , Virus Replication
2.
Proc Natl Acad Sci U S A ; 120(9): e2220120120, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36802427

ABSTRACT

The activation of thymic B cells is critical for their licensing as antigen presenting cells and resulting ability to mediate T cell central tolerance. The processes leading to licensing are still not fully understood. By comparing thymic B cells to activated Peyer's patch B cells at steady state, we found that thymic B cell activation starts during the neonatal period and is characterized by TCR/CD40-dependent activation, followed by immunoglobulin class switch recombination (CSR) without forming germinal centers. Transcriptional analysis also demonstrated a strong interferon signature, which was not apparent in the periphery. Thymic B cell activation and CSR were primarily dependent on type III IFN signaling, and loss of type III IFN receptor in thymic B cells resulted in reduced thymocyte regulatory T cell (Treg) development. Finally, from TCR deep sequencing, we estimate that licensed B cells induce development of a substantial fraction of the Treg cell repertoire. Together, these findings reveal the importance of steady-state type III IFN in generating licensed thymic B cells that induce T cell tolerance to activated B cells.


Subject(s)
Interferon Lambda , T-Lymphocytes, Regulatory , Humans , Infant, Newborn , Thymus Gland , Thymocytes , Receptors, Antigen, T-Cell
3.
Proc Natl Acad Sci U S A ; 119(37): e2201645119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36070344

ABSTRACT

Neuroimmune interactions are crucial for regulating immunity and inflammation. Recent studies have revealed that the central nervous system (CNS) senses peripheral inflammation and responds by releasing molecules that limit immune cell activation, thereby promoting tolerance and tissue integrity. However, the extent to which this is a bidirectional process, and whether peripheral immune cells also promote tolerance mechanisms in the CNS remains poorly defined. Here we report that helminth-induced type 2 inflammation promotes monocyte responses in the brain that are required to inhibit excessive microglial activation and host death. Mechanistically, infection-induced monocytes express YM1 that is sufficient to inhibit tumor necrosis factor production from activated microglia. Importantly, neuroprotective monocytes persist in the brain, and infected mice are protected from subsequent lipopolysaccharide-induced neuroinflammation months after infection-induced inflammation has resolved. These studies demonstrate that infiltrating monocytes promote CNS homeostasis in response to inflammation in the periphery and demonstrate that a peripheral infection can alter the immunologic landscape of the host brain.


Subject(s)
Brain , Encephalitis , Homeostasis , Monocytes , Neuroimmunomodulation , Trichinella spiralis , Trichinellosis , Animals , Brain/immunology , Brain/parasitology , Encephalitis/immunology , Encephalitis/parasitology , Homeostasis/immunology , Lectins/metabolism , Mice , Microglia/immunology , Monocytes/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Trichinellosis/pathology , beta-N-Acetylhexosaminidases/metabolism
4.
J Immunol ; 208(12): 2806-2816, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35675958

ABSTRACT

Infants with attenuated type III IFN (IFN-λ) responses are at increased risk of severe lower respiratory tract infection (sLRI). The IL-28Rα-chain and IL-10Rß-chain form a heterodimeric receptor complex, necessary for IFN-λ signaling. Therefore, to better understand the immunopathogenic mechanisms through which an IFN-λlo microenvironment predisposes to a sLRI, we inoculated neonatal wild-type and IL-28R-deficient (IL-28R -/-) mice with pneumonia virus of mice, a rodent-specific pneumovirus. Infected IL-28R -/- neonates displayed an early, pronounced, and persistent neutrophilia that was associated with enhanced reactive oxygen species (ROS) production, NETosis, and mucus hypersecretion. Targeted deletion of the IL-28R in neutrophils was sufficient to increase neutrophil activation, ROS production, NET formation, and mucus production in the airways. Inhibition of protein-arginine deiminase type 4 (PAD4), a regulator of NETosis, had no effect on myeloperoxidase expression, citrullinated histones, and the magnitude of the inflammatory response in the lungs of infected IL-28R -/- mice. In contrast, inhibition of ROS production decreased NET formation, cellular inflammation, and mucus hypersecretion. These data suggest that IFN-λ signaling in neutrophils dampens ROS-induced NETosis, limiting the magnitude of the inflammatory response and mucus production. Therapeutics that promote IFN-λ signaling may confer protection against sLRI.


Subject(s)
Bronchiolitis, Viral , Extracellular Traps , Interferons/metabolism , Animals , Animals, Newborn , Bronchiolitis, Viral/metabolism , Bronchiolitis, Viral/pathology , Extracellular Traps/metabolism , Humans , Mice , NADPH Oxidases/metabolism , Neutrophils/metabolism , Protein-Arginine Deiminase Type 4 , Reactive Oxygen Species/metabolism
5.
Int J Mol Sci ; 25(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38673989

ABSTRACT

Mertk, a type I receptor tyrosine kinase and member of the TAM family of receptors, has important functions in promoting efferocytosis and resolving inflammation under physiological conditions. In recent years, Mertk has also been linked to pathophysiological roles in cancer, whereby, in several cancer types, including solid cancers and leukemia/lymphomas. Mertk contributes to oncogenic features of proliferation and cell survival as an oncogenic tyrosine kinase. In addition, Mertk expressed on macrophages, including tumor-associated macrophages, promotes immune evasion in cancer and is suggested to act akin to a myeloid checkpoint inhibitor that skews macrophages towards inhibitory phenotypes that suppress host T-cell anti-tumor immunity. In the present study, to better understand the post-translational regulation mechanisms controlling Mertk expression in monocytes/macrophages, we used a PMA-differentiated THP-1 cell model to interrogate the regulation of Mertk expression and developed a novel Mertk reporter cell line to study the intracellular trafficking of Mertk. We show that PMA treatment potently up-regulates Mertk as well as components of the ectodomain proteolytic processing platform ADAM17, whereas PMA differentially regulates the canonical Mertk ligands Gas6 and Pros1 (Gas6 is down-regulated and Pros1 is up-regulated). Under non-stimulated homeostatic conditions, Mertk in PMA-differentiated THP1 cells shows active constitutive proteolytic cleavage by the sequential activities of ADAM17 and the Presenilin/γ-secretase complex, indicating that Mertk is cleaved homeostatically by the combined sequential action of ADAM17 and γ-secretase, after which the cleaved intracellular fragment of Mertk is degraded in a proteasome-dependent mechanism. Using chimeric Flag-Mertk-EGFP-Myc reporter receptors, we confirm that inhibitors of γ-secretase and MG132, which inhibits the 26S proteasome, stabilize the intracellular fragment of Mertk without evidence of nuclear translocation. Finally, the treatment of cells with active γ-carboxylated Gas6, but not inactive Warfarin-treated non-γ-carboxylated Gas6, regulates a distinct proteolytic itinerary-involved receptor clearance and lysosomal proteolysis. Together, these results indicate that pleotropic and complex proteolytic activities regulate Mertk ectodomain cleavage as a homeostatic negative regulatory event to safeguard against the overactivation of Mertk.


Subject(s)
ADAM17 Protein , Amyloid Precursor Protein Secretases , Proteolysis , c-Mer Tyrosine Kinase , Humans , c-Mer Tyrosine Kinase/metabolism , c-Mer Tyrosine Kinase/genetics , ADAM17 Protein/metabolism , ADAM17 Protein/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/genetics , Intercellular Signaling Peptides and Proteins/metabolism , THP-1 Cells , Macrophages/metabolism , Protein S/metabolism , Monocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology
6.
J Virol ; 96(2): e0124121, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34705554

ABSTRACT

Coronaviruses are a major health care threat to humankind. Currently, the host factors that contribute to limit disease severity in healthy young patients are not well defined. Interferons are key antiviral molecules, especially type I and type III interferons. The role of these interferons during coronavirus disease is a subject of debate. Here, using mice that are deficient in type I (IFNAR1-/-), type III (IFNLR1-/-), or both (IFNAR1/LR1-/-) interferon signaling pathways and murine-adapted coronavirus (MHV-A59) administered through the intranasal route, we define the role of interferons in coronavirus infection. We show that type I interferons play a major role in host survival in this model, while a minimal role of type III interferons was manifested only in the absence of type I interferons or during a lethal dose of coronavirus. IFNAR1-/- and IFNAR1/LR1-/- mice had an uncontrolled viral burden in the airways and lung and increased viral dissemination to other organs. The absence of only type III interferon signaling had no measurable difference in the viral load. The increased viral load in IFNAR1-/- and IFNAR1/LR1-/- mice was associated with increased tissue injury, especially evident in the lung and liver. Type I but not type III interferon treatment was able to promote survival if treated during early disease. Further, we show that type I interferon signaling in macrophages contributes to the beneficial effects during coronavirus infection in mice. IMPORTANCE The antiviral and pathological potential of type I and type III interferons during coronavirus infection remains poorly defined, and opposite findings have been reported. We report that both type I and type III interferons have anticoronaviral activities, but their potency and organ specificity differ. Type I interferon deficiency rendered the mice susceptible to even a sublethal murine coronavirus infection, while the type III interferon deficiency impaired survival only during a lethal infection or during a sublethal infection in the absence of type I interferon signaling. While treatment with both type I and III interferons promoted viral clearance in the airways and lung, only type I interferons promoted the viral clearance in the liver and improved host survival upon early treatment (12 h postinfection). This study demonstrates distinct roles and potency of type I and type III interferons and their therapeutic potential during coronavirus lung infection.


Subject(s)
Coronavirus Infections/immunology , Interferon Type I/immunology , Interferons/immunology , Lung , Animals , Female , Lung/immunology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Interferon Lambda
7.
Blood ; 138(8): 722-737, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34436524

ABSTRACT

Immunopathology and intestinal stem cell (ISC) loss in the gastrointestinal (GI) tract is the prima facie manifestation of graft-versus-host disease (GVHD) and is responsible for significant mortality after allogeneic bone marrow transplantation (BMT). Approaches to prevent GVHD to date focus on immune suppression. Here, we identify interferon-λ (IFN-λ; interleukin-28 [IL-28]/IL-29) as a key protector of GI GVHD immunopathology, notably within the ISC compartment. Ifnlr1-/- mice displayed exaggerated GI GVHD and mortality independent of Paneth cells and alterations to the microbiome. Ifnlr1-/- intestinal organoid growth was significantly impaired, and targeted Ifnlr1 deficiency exhibited effects intrinsic to recipient Lgr5+ ISCs and natural killer cells. PEGylated recombinant IL-29 (PEG-rIL-29) treatment of naive mice enhanced Lgr5+ ISC numbers and organoid growth independent of both IL-22 and type I IFN and modulated proliferative and apoptosis gene sets in Lgr5+ ISCs. PEG-rIL-29 treatment improved survival, reduced GVHD severity, and enhanced epithelial proliferation and ISC-derived organoid growth after BMT. The preservation of ISC numbers in response to PEG-rIL-29 after BMT occurred both in the presence and absence of IFN-λ-signaling in recipient natural killer cells. IFN-λ is therefore an attractive and rapidly testable approach to prevent ISC loss and immunopathology during GVHD.


Subject(s)
Bone Marrow Transplantation , Cytokines/pharmacology , Gastrointestinal Diseases , Graft vs Host Disease , Interleukins/pharmacokinetics , Signal Transduction , Animals , Cytokines/immunology , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/immunology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Interleukins/immunology , Mice , Mice, Knockout , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Severity of Illness Index , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Transplantation, Homologous
8.
J Immunol ; 207(2): 436-448, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34215655

ABSTRACT

Phosphatidylserine (PS)-targeting monoclonal Abs (mAbs) that directly target PS and target PS via ß2-gp1 (ß2GP1) have been in preclinical and clinical development for over 10 y for the treatment of infectious diseases and cancer. Although the intended targets of PS-binding mAbs have traditionally included pathogens as well as stressed tumor cells and its associated vasculature in oncology, the effects of PS-targeting mAbs on activated immune cells, notably T cells, which externalize PS upon Ag stimulation, is not well understood. Using human T cells from healthy donor PBMCs activated with an anti-CD3 + anti-CD28 Ab mixture (anti-CD3/CD28) as a model for TCR-mediated PS externalization and T cell stimulation, we investigated effects of two different PS-targeting mAbs, 11.31 and bavituximab (Bavi), on TCR activation and TCR-mediated cytokine production in an ex vivo paradigm. Although 11.31 and Bavi bind selectivity to anti-CD3/28 activated T cells in a PS-dependent manner, surprisingly, they display distinct functional activities in their effect on IFN-γ and TNF-ɑ production, whereby 11.31, but not Bavi, suppressed cytokine production. This inhibitory effect on anti-CD3/28 activated T cells was observed on both CD4+ and CD8+ cells and independently of monocytes, suggesting the effects of 11.31 were directly mediated by binding to externalized PS on activated T cells. Imaging showed 11.31 and Bavi bind at distinct focal depots on the cell membrane. Collectively, our findings indicate that PS-targeting mAb 11.31 suppresses cytokine production by anti-CD3/28 activated T cells.


Subject(s)
Antibodies, Monoclonal/immunology , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Muromonab-CD3/immunology , Phosphatidylserines/immunology , Tumor Necrosis Factor-alpha/immunology , CD3 Complex/immunology , Cell Line , HEK293 Cells , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology
9.
Semin Immunol ; 43: 101303, 2019 06.
Article in English | MEDLINE | ID: mdl-31771761

ABSTRACT

The unexpected discovery of a novel family of antiviral mediators, type III IFNs or IFN-λs, challenged the widely accepted primacy of type I IFNs in antiviral immunity, and it is now well recognized that the IFN-λ-based antiviral system plays a major role in antiviral protection of epithelial barriers. The recent characterization of previously unknown IFN-λ-mediated activities has prompted further reassessment of the role of type I IFNs in innate and adaptive immune and inflammatory responses. Since type I and type III IFNs are co-produced in response to a variety of stimuli, it is likely that many physiological processes are simultaneously and coordinately regulated by these cytokines in pathological conditions, and likely at steady state, as baseline expression of both IFN types is maintained by microbiota. In this review, we discuss emerging differences in the production and signaling of type I and type III IFNs, and summarize results of recent studies describing the involvement of type III IFNs in anti-bacterial and anti-fungal, as well as antiviral, defenses.


Subject(s)
Bacterial Infections/immunology , Interferon Type I/metabolism , Interferons/metabolism , Microbiota/immunology , Mycoses/immunology , Virus Diseases/immunology , Animals , Humans , Immunity , Inflammation , Signal Transduction , Interferon Lambda
10.
Proc Natl Acad Sci U S A ; 117(10): 5409-5419, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32094169

ABSTRACT

Type III IFN lambdas (IFN-λ) have recently been described as important mediators of immune responses at barrier surfaces. However, their role in autoimmune diseases such as systemic lupus erythematosus (SLE), a condition characterized by aberrant type I IFN signaling, has not been determined. Here, we identify a nonredundant role for IFN-λ in immune dysregulation and tissue inflammation in a model of TLR7-induced lupus. IFN-λ protein is increased in murine lupus and IFN-λ receptor (Ifnlr1) deficiency significantly reduces immune cell activation and associated organ damage in the skin and kidneys without effects on autoantibody production. Single-cell RNA sequencing in mouse spleen and human peripheral blood revealed that only mouse neutrophils and human B cells are directly responsive to this cytokine. Rather, IFN-λ activates keratinocytes and mesangial cells to produce chemokines that induce immune cell recruitment and promote tissue inflammation. These data provide insights into the immunobiology of SLE and identify type III IFNs as important factors for tissue-specific pathology in this disease.


Subject(s)
Interferons/physiology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Animals , B-Lymphocytes/immunology , Cell Line , Gene Deletion , Humans , Imiquimod/pharmacology , Inflammation/immunology , Inflammation/pathology , Interferon Inducers/pharmacology , Interferon Type I/physiology , Interferons/pharmacology , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/pathology , Mesangial Cells/drug effects , Mesangial Cells/immunology , Mesangial Cells/pathology , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, Interferon/genetics , Signal Transduction , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/physiology , Interferon Lambda
11.
Proc Natl Acad Sci U S A ; 116(6): 2103-2111, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30655338

ABSTRACT

Cells of all mammals recognize double-stranded RNA (dsRNA) as a foreign material. In response, they release interferons (IFNs) and activate a ubiquitously expressed pseudokinase/endoribonuclease RNase L. RNase L executes regulated RNA decay and halts global translation. Here, we developed a biosensor for 2',5'-oligoadenylate (2-5A), the natural activator of RNase L. Using this biosensor, we found that 2-5A was acutely synthesized by cells in response to dsRNA sensing, which immediately triggered cellular RNA cleavage by RNase L and arrested host protein synthesis. However, translation-arrested cells still transcribed IFN-stimulated genes and secreted IFNs of types I and III (IFN-ß and IFN-λ). Our data suggest that IFNs escape from the action of RNase L on translation. We propose that the 2-5A/RNase L pathway serves to rapidly and accurately suppress basal protein synthesis, preserving privileged production of defense proteins of the innate immune system.


Subject(s)
Biosensing Techniques , Endoribonucleases/chemistry , Interferon-beta/chemistry , Interferons/chemistry , Protein Biosynthesis , Cell Line , Endoribonucleases/metabolism , Humans , Interferon-beta/metabolism , Interferons/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Structure-Activity Relationship
12.
J Immunol ; 201(7): 2082-2093, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30111632

ABSTRACT

Crystal structure of the ternary complex of human IL-24 with two receptors, IL-22R1 and IL-20R2, has been determined at 2.15 Å resolution. A crystallizable complex was created by a novel approach involving fusing the ligand with a flexible linker to the presumed low-affinity receptor, and coexpression of this construct in Drosophila S2 cells together with the presumed high-affinity receptor. This approach, which may be generally applicable to other multiprotein complexes with low-affinity components, was necessitated by the instability of IL-24 expressed by itself in either bacteria or insect cells. Although IL-24 expressed in Escherichia coli was unstable and precipitated almost immediately upon its refolding and purification, a small fraction of IL-24 remaining in the folded state was shown to be active in a cell-based assay. In the crystal structure presented here, we found that two cysteine residues in IL-24 do not form a predicted disulfide bond. Lack of structural restraint by disulfides, present in other related cytokines, is most likely reason for the low stability of IL-24. Although the contact area between IL-24 and IL-22R1 is larger than between the cytokine and IL-20R2, calculations show the latter interaction to be slightly more stable, suggesting that the shared receptor (IL-20R2) might be the higher-affinity receptor.


Subject(s)
Interleukins/metabolism , Multiprotein Complexes/metabolism , Receptors, Interleukin/metabolism , Animals , Cell Line , Crystallography, X-Ray , Cytokines , Drosophila , Humans , Protein Binding , Protein Conformation , Protein Domains/genetics , Receptors, Interleukin/genetics , Signal Transduction
14.
J Immunol ; 199(7): 2460-2474, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28848065

ABSTRACT

Nucleic acids carrying pathogen-associated molecular patterns trigger innate immune responses and are used to activate host immunity. Although synthetic nucleic acids have been used for that purpose, they have shown limitations for in vivo and clinical applications. To address this issue, we tested a naturally occurring dsRNA extracted from rice bran (rb-dsRNA) and characterized it as a potent ligand of TLR3 and MDA5. In this study, intranasal administration of rb-dsRNA induced production of type I IFNs by alveolar macrophages and protected mice from morbidity and mortality resulting from respiratory virus infection, such as influenza A virus. This protection was completely absent in mice lacking both TRIF and MDA5, indicating the essential role of TLR3- and MDA5-dependent pathways. Interestingly, IFNAR1-deficient mice retained residual antiviral protection, which was abolished by pharmacological inhibition of caspase 1, but not IL-1ß signaling. In fact, rb-dsRNA activated caspase 1 via TRIF, resulting in the release of IL-1ß and LDH. In addition to the direct antiviral activity, rb-dsRNA modulated the immune cell population in the lungs by repopulating virus-depleted alveolar macrophages. Our data demonstrate that rb-dsRNA orchestrates IFN-dependent and -independent direct antiviral protection and that it is a potent immune stimulator modulating antiviral immunity in the lungs. These findings open doors to a range of precise immune-modulating studies and therapeutic options.


Subject(s)
Antiviral Agents/isolation & purification , Influenza A virus/immunology , Interferon Type I/immunology , Orthomyxoviridae Infections/immunology , Oryza/genetics , RNA, Double-Stranded/immunology , RNA, Double-Stranded/isolation & purification , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Animals , Antiviral Agents/immunology , Caspase Inhibitors/administration & dosage , Immunity, Innate , Interferon Type I/biosynthesis , Interferon-Induced Helicase, IFIH1/chemistry , Interferon-Induced Helicase, IFIH1/deficiency , Interferon-Induced Helicase, IFIH1/genetics , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Ligands , Lung/immunology , Lung/virology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Mice , Orthomyxoviridae Infections/prevention & control , Oryza/chemistry , Plants/chemistry , Plants/genetics , RNA, Double-Stranded/administration & dosage , RNA, Double-Stranded/pharmacology , Receptor, Interferon alpha-beta/deficiency , Signal Transduction/drug effects , Toll-Like Receptor 3/chemistry
15.
J Biol Chem ; 292(18): 7295-7303, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28289095

ABSTRACT

Type I interferons (IFN-α/ß) and the more recently identified type III IFNs (IFN-λ) function as the first line of defense against virus infection and regulate the development of both innate and adaptive immune responses. Type III IFNs were originally identified as a novel ligand-receptor system acting in parallel with type I IFNs, but subsequent studies have provided increasing evidence for distinct roles for each IFN family. In addition to their compartmentalized antiviral actions, these two systems appear to have multiple levels of cross-regulation and act coordinately to achieve effective antimicrobial protection with minimal collateral damage to the host.


Subject(s)
Adaptive Immunity , Immunity, Innate , Interleukins/immunology , Virus Diseases/immunology , Animals , Humans , Interferon Type I/immunology , Interferons
17.
PLoS Pathog ; 12(4): e1005600, 2016 04.
Article in English | MEDLINE | ID: mdl-27128797

ABSTRACT

Type I (IFN-α/ß) and type III (IFN-λ) interferons (IFNs) exert shared antiviral activities through distinct receptors. However, their relative importance for antiviral protection of different organ systems against specific viruses remains to be fully explored. We used mouse strains deficient in type-specific IFN signaling, STAT1 and Rag2 to dissect distinct and overlapping contributions of type I and type III IFNs to protection against homologous murine (EW-RV strain) and heterologous (non-murine) simian (RRV strain) rotavirus infections in suckling mice. Experiments demonstrated that murine EW-RV is insensitive to the action of both types of IFNs, and that timely viral clearance depends upon adaptive immune responses. In contrast, both type I and type III IFNs can control replication of the heterologous simian RRV in the gastrointestinal (GI) tract, and they cooperate to limit extra-intestinal simian RRV replication. Surprisingly, intestinal epithelial cells were sensitive to both IFN types in neonatal mice, although their responsiveness to type I, but not type III IFNs, diminished in adult mice, revealing an unexpected age-dependent change in specific contribution of type I versus type III IFNs to antiviral defenses in the GI tract. Transcriptional analysis revealed that intestinal antiviral responses to RV are triggered through either type of IFN receptor, and are greatly diminished when receptors for both IFN types are lacking. These results also demonstrate a murine host-specific resistance to IFN-mediated antiviral effects by murine EW-RV, but the retention of host efficacy through the cooperative action by type I and type III IFNs in restricting heterologous simian RRV growth and systemic replication in suckling mice. Collectively, our findings revealed a well-orchestrated spatial and temporal tuning of innate antiviral responses in the intestinal tract where two types of IFNs through distinct patterns of their expression and distinct but overlapping sets of target cells coordinately regulate antiviral defenses against heterologous or homologous rotaviruses with substantially different effectiveness.


Subject(s)
Interferon Type I/immunology , Interferon-gamma/immunology , Intestines/immunology , Rotavirus Infections/immunology , Animals , Animals, Newborn , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Rotavirus
18.
Immunol Rev ; 255(1): 25-39, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23947345

ABSTRACT

Interferons (IFNs) are produced in response to virus infection and induce an antiviral state in virtually all cell types. In addition to upregulating the transcription of genes that inhibit virus replication, type I (or -α/ß) IFNs also act to orchestrate the adaptive immune response to virus infection. Recently a new family of antiviral cytokines, the type III (or -λ) IFNs, has been identified that activate the same antiviral pathways via a distinct receptor. Although the identical transcription factor, IFN-stimulated gene factor 3 is activated by either IFN-α/ß or IFN-λ signaling, differences in the induction and action of these two cytokine families are beginning to be appreciated. In this article, we review this emerging body of literature on the differing roles these cytokines play in host defense of the mucosal surface. Although many viruses enter the body through the respiratory and gastrointestinal tracts, we have focused the discussion on influenza A virus, respiratory syncytial virus, and rotavirus, three ubiquitous human pathogens that target the epithelial lining and are associated with a major disease burden.


Subject(s)
Interferons/immunology , Interferons/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Animals , Gene Expression Regulation , Humans , Janus Kinases/metabolism , Ligands , Mucous Membrane/virology , Phosphorylation , Protein Biosynthesis , STAT Transcription Factors/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism , Viruses/immunology
19.
Cell Commun Signal ; 14(1): 19, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27595981

ABSTRACT

BACKGROUND: Tyro3, Axl, and Mertk (TAMs) are a family of three conserved receptor tyrosine kinases that have pleiotropic roles in innate immunity and homeostasis and when overexpressed in cancer cells can drive tumorigenesis. METHODS: In the present study, we engineered EGFR/TAM chimeric receptors (EGFR/Tyro3, EGFR/Axl, and EGF/Mertk) with the goals to interrogate post-receptor functions of TAMs, and query whether TAMs have unique or overlapping post-receptor activation profiles. Stable expression of EGFR/TAMs in EGFR-deficient CHO cells afforded robust EGF inducible TAM receptor phosphorylation and activation of downstream signaling. RESULTS: Using a series of unbiased screening approaches, that include kinome-view analysis, phosphor-arrays, RNAseq/GSEA analysis, as well as cell biological and in vivo readouts, we provide evidence that each TAM has unique post-receptor signaling platforms and identify an intrinsic role for Axl that impinges on cell motility and invasion compared to Tyro3 and Mertk. CONCLUSION: These studies demonstrate that TAM show unique post-receptor signatures that impinge on distinct gene expression profiles and tumorigenic outcomes.


Subject(s)
ErbB Receptors/metabolism , Mammary Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , CHO Cells , Cell Movement , Cricetinae , Cricetulus , ErbB Receptors/genetics , Female , Humans , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
20.
J Biol Chem ; 289(37): 25750-63, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25074926

ABSTRACT

TYRO3, AXL, and MER receptors (TAMs) are three homologous type I receptor-tyrosine kinases that are activated by endogenous ligands, protein S (PROS1) and growth arrest-specific gene 6 (GAS6). These ligands can either activate TAMs as soluble factors, or, in turn, opsonize phosphatidylserine (PS) on apoptotic cells (ACs) and serve as bridging molecules between ACs and TAMs. Abnormal expression and activation of TAMs have been implicated in promoting proliferation and survival of cancer cells, as well as in suppressing anti-tumor immunity. Despite the fact that TAM receptors share significant similarity, little is known about the specificity of interaction between TAM receptors and their ligands, particularly in the context of ACs, and about the functional diversity of TAM receptors. To study ligand-mediated activation of TAMs, we generated a series of reporter cell lines expressing chimeric TAM receptors. Using this system, we found that each TAM receptor has a unique pattern of interaction with and activation by GAS6 and PROS1, which is also differentially affected by the presence of ACs, PS-containing lipid vesicles and enveloped virus. We also demonstrated that γ-carboxylation of ligands is essential for the full activation of TAMs and that soluble immunoglobulin-like TAM domains act as specific ligand antagonists. These studies demonstrate that, despite their similarity, TYRO3, AXL, and MER are likely to perform distinct functions in both immunoregulation and the recognition and removal of ACs.


Subject(s)
Apoptosis/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Blood Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Protein S , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Vesicular Stomatitis/genetics , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL