Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 23(18)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36142628

ABSTRACT

The Asian seabass is of importance both as a farmed and wild animal. With the emergence of infectious diseases, there is a need to understand and characterize the immune system. In humans, the highly polymorphic MHC class I (MHC-I) molecules play an important role in antigen presentation for the adaptive immune system. In the present study, we characterized a single MHC-I gene in Asian seabass (Lates calcarifer) by amplifying and sequencing the MHC-I alpha 1 and alpha 2 domains, followed by multi-sequence alignment analyses. The results indicated that the Asian seabass MHC-I α1 and α2 domain sequences showed an overall similarity within Asian seabass and retained the majority of the conserved binding residues of human leukocyte antigen-A2 (HLA-A2). Phylogenetic tree analysis revealed that the sequences belonged to the U lineage. Mapping the conserved binding residue positions on human HLA-A2 and grass carp crystal structure showed a high degree of similarity. In conclusion, the availability of MHC-I α1 and α2 sequences enhances the quality of MHC class I genetic information in Asian seabass, providing new tools to analyze fish immune responses to pathogen infections, and will be applicable in the study of the phylogeny and the evolution of antigen-specific receptors.


Subject(s)
Bass , Perciformes , Animals , Bass/genetics , Fishes , HLA-A2 Antigen/genetics , Humans , Perciformes/genetics , Phylogeny
2.
J Med Chem ; 65(17): 11759-11775, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35984914

ABSTRACT

Glucagon-like peptide-1 (GLP-1) lowers blood glucose by inducing insulin but also has other poorly understood properties. Here, we show that hydroxy amino acids (Thr11, Ser14, Ser17, Ser18) in GLP-1(7-36) act in concert to direct cell signaling. Mutating any single residue to alanine removes one hydroxyl group, thereby reducing receptor affinity and cAMP 10-fold, with Ala11 or Ala14 also reducing ß-arrestin-2 10-fold, while Ala17 or Ala18 also increases ERK1/2 phosphorylation 5-fold. Multiple alanine mutations more profoundly bias signaling, differentially silencing or restoring one or more signaling properties. Mutating three serines silences only ERK1/2, the first example of such bias. Mutating all four residues silences ß-arrestin-2, ERK1/2, and Ca2+ maintains the ligand and receptor at the membrane but still potently stimulates cAMP and insulin secretion in cells and mice. These novel findings indicate that hydrogen bonding cooperatively controls cell signaling and highlight an important regulatory hydroxyl patch in hormones that activate class B G protein-coupled receptors.


Subject(s)
Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Alanine , Animals , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Insulin/metabolism , Mice , Signal Transduction , beta-Arrestin 2/metabolism
3.
J Med Chem ; 64(4): 2186-2204, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33570940

ABSTRACT

The zinc-containing histone deacetylase enzyme HDAC7 is emerging as an important regulator of immunometabolism and cancer. Here, we exploit a cavity in HDAC7, filled by Tyr303 in HDAC1, to derive new inhibitors. Phenacetyl hydroxamates and 2-phenylbenzoyl hydroxamates bind to Zn2+ and are 50-2700-fold more selective inhibitors of HDAC7 than HDAC1. Phenylbenzoyl hydroxamates are 30-70-fold more potent HDAC7 inhibitors than phenacetyl hydroxamates, which is attributed to the benzoyl aromatic group interacting with Phe679 and Phe738. Phthalimide capping groups, including a saccharin analogue, decrease rotational freedom and provide hydrogen bond acceptor carbonyl/sulfonamide oxygens that increase inhibitor potency, liver microsome stability, solubility, and cell activity. Despite being the most potent HDAC7 inhibitors to date, they are not selective among class IIa enzymes. These strategies may help to produce tools for interrogating HDAC7 biology related to its catalytic site.


Subject(s)
Benzamides/pharmacology , Benzeneacetamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Benzamides/chemical synthesis , Benzamides/metabolism , Benzeneacetamides/chemical synthesis , Benzeneacetamides/metabolism , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/metabolism , Biphenyl Compounds/pharmacology , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/metabolism , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/metabolism , Molecular Docking Simulation , Molecular Structure , Protein Binding , Structure-Activity Relationship , THP-1 Cells
4.
Elife ; 92020 07 22.
Article in English | MEDLINE | ID: mdl-32697191

ABSTRACT

Diabetes mellitus is a known susceptibility factor for severe influenza virus infections. However, the mechanisms that underlie this susceptibility remain incompletely understood. Here, the effects of high glucose levels on influenza severity were investigated using an in vitro model of the pulmonary epithelial-endothelial barrier as well as an in vivo murine model of type II diabetes. In vitro we show that high glucose conditions prior to IAV infection increased virus-induced barrier damage. This was associated with an increased pro-inflammatory response in endothelial cells and the subsequent damage of the epithelial junctional complex. These results were subsequently validated in vivo. This study provides the first evidence that hyperglycaemia may increase influenza severity by damaging the pulmonary epithelial-endothelial barrier and increasing pulmonary oedema. These data suggest that maintaining long-term glucose control in individuals with diabetes is paramount in reducing the morbidity and mortality associated with influenza virus infections.


Subject(s)
Endothelial Cells/physiology , Epithelial Cells/physiology , Glucose/metabolism , Influenza A virus/physiology , Influenza, Human/virology , Lung/physiopathology , Animals , Disease Models, Animal , Endothelial Cells/virology , Epithelial Cells/virology , Female , Humans , Lung/virology , Male , Mice , Mice, Inbred C57BL
5.
PLoS Pathog ; 16(7): e1008651, 2020 07.
Article in English | MEDLINE | ID: mdl-32658914

ABSTRACT

Type-2 immunity elicits tissue repair and homeostasis, however dysregulated type-2 responses cause aberrant tissue remodelling, as observed in asthma. Severe respiratory viral infections in infancy predispose to later asthma, however, the processes that mediate tissue damage-induced type-2 inflammation and the origins of airway remodelling remain ill-defined. Here, using a preclinical mouse model of viral bronchiolitis, we find that increased epithelial and mesenchymal high-mobility group box 1 (HMGB1) expression is associated with increased numbers of IL-13-producing type-2 innate lymphoid cell (ILC2s) and the expansion of the airway smooth muscle (ASM) layer. Anti-HMGB1 ablated lung ILC2 numbers and ASM growth in vivo, and inhibited ILC2-mediated ASM cell proliferation in a co-culture model. Furthermore, we identified that HMGB1/RAGE (receptor for advanced glycation endproducts) signalling mediates an ILC2-intrinsic IL-13 auto-amplification loop. In summary, therapeutic targeting of the HMGB1/RAGE signalling axis may act as a novel asthma preventative by dampening ILC2-mediated type-2 inflammation and associated ASM remodelling.


Subject(s)
Airway Remodeling/immunology , HMGB1 Protein/immunology , Inflammation/immunology , Lymphocytes/immunology , Muscle, Smooth/immunology , Animals , Mice , Muscle, Smooth/pathology , Receptor for Advanced Glycation End Products/immunology
6.
Am J Respir Crit Care Med ; 201(11): 1358-1371, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32105156

ABSTRACT

Rationale: Respiratory syncytial virus (RSV) bronchiolitis causes significant infant mortality. Bronchiolitis is characterized by airway epithelial cell (AEC) death; however, the mode of death remains unknown.Objectives: To determine whether necroptosis contributes to RSV bronchiolitis pathogenesis via HMGB1 (high mobility group box 1) release.Methods: Nasopharyngeal samples were collected from children presenting to the hospital with acute respiratory infection. Primary human AECs and neonatal mice were inoculated with RSV and murine Pneumovirus, respectively. Necroptosis was determined via viability assays and immunohistochemistry for RIPK1 (receptor-interacting protein kinase-1), MLKL (mixed lineage kinase domain-like pseudokinase) protein, and caspase-3. Necroptosis was blocked using pharmacological inhibitors and RIPK1 kinase-dead knockin mice.Measurements and Main Results: HMGB1 levels were elevated in nasopharyngeal samples of children with acute RSV infection. RSV-induced epithelial cell death was associated with increased phosphorylated RIPK1 and phosphorylated MLKL but not active caspase-3 expression. Inhibition of RIPK1 or MLKL attenuated RSV-induced HMGB1 translocation and release, and lowered viral load. MLKL inhibition increased active caspase-3 expression in a caspase-8/9-dependent manner. In susceptible mice, Pneumovirus infection upregulated RIPK1 and MLKL expression in the airway epithelium at 8 to 10 days after infection, coinciding with AEC sloughing, HMGB1 release, and neutrophilic inflammation. Genetic or pharmacological inhibition of RIPK1 or MLKL attenuated these pathologies, lowered viral load, and prevented type 2 inflammation and airway remodeling. Necroptosis inhibition in early life ameliorated asthma progression induced by viral or allergen challenge in later life.Conclusions: Pneumovirus infection induces AEC necroptosis. Inhibition of necroptosis may be a viable strategy to limit the severity of viral bronchiolitis and break its nexus with asthma.


Subject(s)
Bronchiolitis/virology , Epithelial Cells/metabolism , Epithelial Cells/pathology , HMGB1 Protein/metabolism , Necroptosis , Respiratory Mucosa/cytology , Respiratory Syncytial Virus Infections/metabolism , Animals , Child, Preschool , Humans , Infant , Mice , Prospective Studies
7.
Br J Pharmacol ; 176(19): 3775-3790, 2019 10.
Article in English | MEDLINE | ID: mdl-31236923

ABSTRACT

BACKGROUND AND PURPOSE: Chronic liver diseases feature excessive collagen and matrix protein deposition or crosslinking that characterises fibrosis, leads to scar tissue, and disrupts liver functions. There is no effective treatment. This study investigated whether treatment with selective histone deacetylase (HDAC) inhibitors might specifically reduce type 2 inflammation in the injured liver, thereby attenuating fibrogenesis in mice. EXPERIMENTAL APPROACH: Thioacetamide (TAA) was used to induce hepatic inflammation, fibrosis, and liver damage in female C57BL/6 mice, similar to the clinical features of chronic human liver disease. We used eight inhibitors of different human HDAC enzymes to probe histological (IHC and TUNEL), biochemical and immunological changes (flow cytometry, qPCR, Legendplex, and ELISA) in pathology, fibrosis, hepatic immune cell flux, and inflammatory cytokine expression. KEY RESULTS: Inhibitors of class I, but not class II, HDAC enzymes potently suppressed chronic hepatic inflammation and fibrosis in mice, attenuating accumulation and activation of IL-33-dependent, but not IL-25-dependent, group 2 innate lymphoid cells (ILC2) and inhibiting type 2 inflammation that drives hepatic stellate cells to secrete excessive collagen and matrix proteins. CONCLUSIONS AND IMPLICATIONS: The results show that potent and selective inhibitors of class I only HDAC enzymes profoundly inhibit hepatocyte death and type 2 inflammation to prevent TAA-induced liver fibrosis in mice. The specific HDAC enzymes identified here may be key promoters of inflammation in chronic liver fibrosis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Liver Cirrhosis/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Female , Histone Deacetylase Inhibitors/chemistry , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Ligands , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Mice , Mice, Inbred C57BL , Thioacetamide
8.
JCI Insight ; 3(14)2018 07 26.
Article in English | MEDLINE | ID: mdl-30046009

ABSTRACT

Chronic liver disease (CLD) is associated with tissue-destructive fibrosis. Considering that common mechanisms drive fibrosis across etiologies, and that steatosis is an important cofactor for pathology, we performed RNA sequencing on liver biopsies of patients with different fibrosis stages, resulting from infection with hepatitis C virus (HCV) (with or without steatosis) or fatty liver disease. In combination with enhanced liver fibrosis score correlation analysis, we reveal a common set of genes associated with advanced fibrosis, as exemplified by those encoding the transcription factor ETS-homologous factor (EHF) and the extracellular matrix protein versican (VCAN). We identified 17 fibrosis-associated genes as candidate EHF targets and demonstrated that EHF regulates multiple fibrosis-associated genes, including VCAN, in hepatic stellate cells. Serum VCAN levels were also elevated in advanced fibrosis patients. Comparing biopsies from patients with HCV with or without steatosis, we identified a steatosis-enriched gene set associated with advanced fibrosis, validating follistatin-like protein 1 (FSTL1) as an exemplar of this profile. In patients with advanced fibrosis, serum FSTL1 levels were elevated in those with steatosis (versus those without). Liver Fstl1 mRNA levels were also elevated in murine CLD models. We thus reveal a common gene signature for CLD-associated liver fibrosis and potential biomarkers and/or targets for steatosis-associated liver fibrosis.


Subject(s)
Fatty Liver/complications , Fatty Liver/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/genetics , Adult , Biomarkers/blood , Biopsy , Cohort Studies , Female , Follistatin-Related Proteins/blood , Follistatin-Related Proteins/genetics , Gene Expression , Gene Expression Profiling , Hepacivirus , Hepatitis C/complications , Humans , Liver/metabolism , Male , Meta-Analysis as Topic , Middle Aged , Versicans/blood , Versicans/metabolism , Young Adult
9.
J Exp Med ; 215(2): 537-557, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29273643

ABSTRACT

Respiratory syncytial virus-bronchiolitis is a major independent risk factor for subsequent asthma, but the causal mechanisms remain obscure. We identified that transient plasmacytoid dendritic cell (pDC) depletion during primary Pneumovirus infection alone predisposed to severe bronchiolitis in early life and subsequent asthma in later life after reinfection. pDC depletion ablated interferon production and increased viral load; however, the heightened immunopathology and susceptibility to subsequent asthma stemmed from a failure to expand functional neuropilin-1+ regulatory T (T reg) cells in the absence of pDC-derived semaphorin 4a (Sema4a). In adult mice, pDC depletion predisposed to severe bronchiolitis only after antibiotic treatment. Consistent with a protective role for the microbiome, treatment of pDC-depleted neonates with the microbial-derived metabolite propionate promoted Sema4a-dependent T reg cell expansion, ameliorating both diseases. In children with viral bronchiolitis, nasal propionate levels were decreased and correlated with an IL-6high/IL-10low microenvironment. We highlight a common but age-related Sema4a-mediated pathway by which pDCs and microbial colonization induce T reg cell expansion to protect against severe bronchiolitis and subsequent asthma.


Subject(s)
Asthma/prevention & control , Bronchiolitis, Viral/prevention & control , Dendritic Cells/immunology , Semaphorins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Animals, Newborn , Asthma/immunology , Bronchiolitis, Viral/etiology , Bronchiolitis, Viral/immunology , Child , Child, Preschool , Disease Models, Animal , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Female , Humans , Interleukin-10/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microbiota/immunology , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/immunology , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/immunology , Semaphorins/antagonists & inhibitors , T-Lymphocytes, Regulatory/cytology
10.
Nat Commun ; 8(1): 351, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28839129

ABSTRACT

Complement C3a is an important protein in innate and adaptive immunity, but its specific roles in vivo remain uncertain because C3a degrades rapidly to form the C3a-desArg protein, which does not bind to the C3a receptor and is indistinguishable from C3a using antibodies. Here we develop the most potent, stable and highly selective small molecule modulators of C3a receptor, using a heterocyclic hinge to switch between agonist and antagonist ligand conformations. This enables characterization of C3 areceptor-selective pro- vs. anti-inflammatory actions in human mast cells and macrophages, and in rats. A C3a receptor-selective agonist induces acute rat paw inflammation by first degranulating mast cells before activating macrophages and neutrophils. An orally administered C3a receptor-selective antagonist inhibits mast cell degranulation, thereby blocking recruitment and activation of macrophages and neutrophils, expression of inflammatory mediators and inflammation in a rat paw edema model. These novel tools reveal the mechanism of C3a-induced inflammation and provide new insights to complement-based medicines.Complement C3a is an important protein in innate and adaptive immunity, but its roles in vivo are unclear. Here the authors develop novel chemical agonists and antagonists for the C3a receptor, and show that they modulate mast cell degranulation and inflammation in a rat paw edema model.


Subject(s)
Complement C3a/physiology , Immunity, Innate/genetics , Receptors, Complement/chemistry , Animals , Anti-Asthmatic Agents/pharmacology , Cell Degranulation/drug effects , Cells, Cultured , Complement C3a/genetics , Complement C3a/metabolism , Cromolyn Sodium/pharmacology , Humans , Ligands , Macrophages/immunology , Male , Mast Cells/immunology , Neutrophils/immunology , Protein Conformation , Rats , Rats, Wistar , Receptors, Complement/agonists , Receptors, Complement/antagonists & inhibitors
11.
Sci Rep ; 7(1): 2353, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28539639

ABSTRACT

Respiratory syncytial virus (RSV)-bronchiolitis is a major cause of infant morbidity and mortality and a risk factor for subsequent asthma. We showed previously that toll-like receptor (TLR)7 in plasmacytoid dendritic cells (pDCs) is critical for protection against bronchiolitis and asthma in mice infected with pneumonia virus of mice (PVM), the mouse homolog of RSV. This lack of redundancy was unexpected as interferon-ß promotor stimulator-1 (IPS-1) signalling, downstream of RIG-I-like receptor (RLR) and not TLR7 activation, contributes to host defence in hRSV-inoculated adult mice. To further clarify the role of IPS-1 signalling, we inoculated IPS-1-/- and WT mice with PVM in early-life, and again in later-life, to model the association between bronchiolitis and asthma. IPS-1 deficiency predisposed to severe PVM bronchiolitis, characterised by neutrophilic inflammation and necroptotic airway epithelial cell death, high mobility group box 1 (HMGB1) and IL-33 release, and downstream type-2 inflammation. Secondary infection induced an eosinophilic asthma-like pathophysiology in IPS-1-/- but not WT mice. Mechanistically, we identified that IPS-1 is necessary for pDC recruitment, IFN-α production and viral control. Our findings suggest that TLR7 and RLR signalling work collaboratively to optimally control the host response to pneumovirus infection thereby protecting against viral bronchiolitis and subsequent asthma.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Asthma/metabolism , Bronchiolitis/metabolism , Murine pneumonia virus/physiology , Pneumovirus Infections/virology , Adaptor Proteins, Signal Transducing/genetics , Animals , Asthma/genetics , Bronchiolitis/genetics , DEAD Box Protein 58/metabolism , Dendritic Cells/metabolism , Host-Pathogen Interactions , Interferon-alpha/metabolism , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Pneumovirus Infections/genetics , Pneumovirus Infections/metabolism , Signal Transduction , Toll-Like Receptor 7/metabolism
12.
Elife ; 62017 01 18.
Article in English | MEDLINE | ID: mdl-28099113

ABSTRACT

Asthma is a chronic inflammatory disease. Although many patients with asthma develop type-2 dominated eosinophilic inflammation, a number of individuals develop paucigranulocytic asthma, which occurs in the absence of eosinophilia or neutrophilia. The aetiology of paucigranulocytic asthma is unknown. However, both respiratory syncytial virus (RSV) infection and mutations in the receptor for advanced glycation endproducts (RAGE) are risk factors for asthma development. Here, we show that RAGE deficiency impairs anti-viral immunity during an early-life infection with pneumonia virus of mice (PVM; a murine analogue of RSV). The elevated viral load was associated with the release of high mobility group box-1 (HMGB1) which triggered airway smooth muscle remodelling in early-life. Re-infection with PVM in later-life induced many of the cardinal features of asthma in the absence of eosinophilic or neutrophilic inflammation. Anti-HMGB1 mitigated both early-life viral disease and asthma-like features, highlighting HMGB1 as a possible novel therapeutic target.


Subject(s)
Agranulocytosis/complications , Agranulocytosis/genetics , Asthma/genetics , Asthma/pathology , Genetic Predisposition to Disease , HMGB1 Protein/metabolism , Receptor for Advanced Glycation End Products/deficiency , Animals , Mice , Murine pneumonia virus/immunology , Viral Load
13.
J Allergy Clin Immunol ; 138(5): 1326-1337, 2016 11.
Article in English | MEDLINE | ID: mdl-27236500

ABSTRACT

BACKGROUND: Frequent viral lower respiratory infections in early life are an independent risk factor for asthma onset. This risk and the development of persistent asthma are significantly greater in children who later become sensitized. OBJECTIVE: We sought to elucidate the pathogenic processes that underlie the synergistic interplay between allergen exposures and viral infections. METHODS: Mice were inoculated with a murine-specific Pneumovirus species (pneumonia virus of mice [PVM]) and exposed to low-dose cockroach extract (CRE) in early and later life, and airway inflammation, remodeling, and hyperreactivity assessed. Mice were treated with anti-IL-33 or apyrase to neutralize or block IL-33 release. RESULTS: PVM infection or CRE exposure alone did not induce disease, whereas PVM/CRE coexposure acted synergistically to induce the hallmark features of asthma. CRE exposure during viral infection in early life induced a biphasic IL-33 response and impaired IFN-α and IFN-λ production, which in turn increased epithelial viral burden, airway smooth muscle growth, and type 2 inflammation. These features were ameliorated when CRE-induced IL-33 release was blocked or neutralized, whereas substitution of CRE with exogenous IL-33 recapitulated the phenotype observed in PVM/CRE-coexposed mice. Mechanistically, IL-33 downregulated viperin and interferon regulatory factor 7 gene expression and rapidly degraded IL-1 receptor-associated kinase 1 expression in plasmacytoid dendritic cells both in vivo and in vitro, leading to Toll-like receptor 7 hyporesponsiveness and impaired IFN-α production. CONCLUSION: We identified a hitherto unrecognized function of IL-33 as a potent suppressor of innate antiviral immunity and demonstrate that IL-33 contributes significantly to the synergistic interplay between respiratory virus and allergen exposures in the onset and progression of asthma.


Subject(s)
Allergens/immunology , Asthma/immunology , Cockroaches , Cytokines/immunology , Insect Proteins/immunology , Murine pneumonia virus , Pneumovirus Infections/immunology , Air Pollutants/immunology , Animals , Asthma/virology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/virology , Dendritic Cells/immunology , Lung/virology , Mice, Inbred BALB C , Pneumovirus Infections/virology , Viral Load
14.
J Allergy Clin Immunol ; 136(4): 1065-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25930193

ABSTRACT

BACKGROUND: A variant in the IL-6 receptor (IL-6R) gene increases asthma risk and is predicted to decrease IL-6 classic signaling and increase IL-6 trans-signaling. This suggests that inhibition of IL-6 trans-signaling, but not classic signaling, might suppress allergic airway inflammation. OBJECTIVES: We sought to determine whether IL-6 signaling contributes to (1) acute experimental asthma induced by clinically relevant allergens and (2) variation in asthma clinical phenotypes in asthmatic patients. METHODS: Mice were sensitized to house dust mite (HDM) or cockroach at day 0, treated with IL-6R inhibitors at day 13, and challenged with the same allergen at days 14 to 17. End points were measured 3 hours after the final challenge. IL-6 and soluble IL-6 receptor (sIL-6R) expression in induced sputum of asthmatic patients was correlated with asthma clinical phenotypes. RESULTS: Both HDM and cockroach induced a type 2/type 17 cytokine profile and mixed granulocytic inflammation in the airways. Both allergens increased IL-6 expression in the airways, but only cockroach induced sIL-6R expression. Therefore HDM challenge promoted IL-6 classic signaling but not trans-signaling; in this model treatment with anti-IL-6R did not suppress airway inflammation. In contrast, cockroach-induced inflammation involved activation of IL-6 trans-signaling and production of IL-17A by γδ T cells. Anti-IL-6R, selective blockade of sIL-6R, or γδ T-cell deficiency significantly attenuated cockroach-induced inflammation. Asthmatic patients with high airway IL-6 and sIL-6R levels were enriched for the neutrophilic and mixed granulocytic subtypes. CONCLUSION: Experimental asthma associated with both high IL-6 and high sIL-6R levels in the airways is attenuated by treatment with IL-6R inhibitors.


Subject(s)
Asthma/immunology , Interleukin-6/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Interleukin-6/immunology , Signal Transduction/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Allergens/immunology , Allergens/toxicity , Animals , Asthma/chemically induced , Asthma/pathology , Cockroaches/immunology , Mice , Pyroglyphidae/immunology , Signal Transduction/drug effects , Th17 Cells/pathology , Th2 Cells/pathology
15.
Am J Pathol ; 184(6): 1795-806, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24726644

ABSTRACT

Human metapneumovirus (hMPV) is a leading cause of respiratory tract disease in children and is associated with acute bronchiolitis, pneumonia, and asthma exacerbations, yet the mechanisms by which the host immune response to hMPV is regulated are poorly understood. By using gene-deleted neonatal mice, we examined the contributions of the innate receptor signaling molecules interferon (IFN)-ß promoter stimulator 1 (IPS-1), IFN regulatory factor (IRF) 3, and IRF7. Viral load in the lungs was markedly greater in IPS-1(-/-) > IRF3/7(-/-) > IRF3(-/-), but not IRF7(-/-), mice compared with wild-type mice. IFN-ß and IFN-λ2/3 (IL-28A/B) production was attenuated in the bronchoalveolar lavage fluid in all factor-deficient mice compared with wild-type mice at 1 day after infection, although IFN-λ2/3 was greater in IRF3/7(-/-) mice at 5 days after infection. IRF7(-/-) and IRF3/7(-/-) mice presented with airway eosinophilia, whereas only IRF3/7(-/-) mice developed an exaggerated type 1 and 17 helper T-cell response, characterized by natural killer T-cell and neutrophilic inflammation. Despite having the highest viral load, IPS-1(-/-) mice did not develop a proinflammatory cytokine or granulocytic response to hMPV infection. Our findings demonstrate that IFN-ß, but not IFN-λ2/3, produced via an IPS-1-IRF3 signaling pathway, is important for hMPV clearance. In the absence of a robust type I IFN-α/ß response, targeting the IPS-1 signaling pathway may limit the overexuberant inflammatory response that occurs as a consequence of viral persistence.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-7/immunology , Metapneumovirus/immunology , Paramyxoviridae Infections/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Acute Disease , Adaptor Proteins, Signal Transducing/genetics , Animals , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/genetics , Interferons/genetics , Interferons/immunology , Metapneumovirus/genetics , Mice , Mice, Knockout , Paramyxoviridae Infections/genetics , Paramyxoviridae Infections/pathology , Signal Transduction/genetics , Signal Transduction/immunology , Th1 Cells/pathology , Th17 Cells/pathology
16.
J Allergy Clin Immunol ; 134(2): 440-50, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24506934

ABSTRACT

BACKGROUND: The receptor for advanced glycation end products (RAGE) shares common ligands and signaling pathways with TLR4, a key mediator of house dust mite (Dermatophagoides pteronyssinus) (HDM) sensitization. We hypothesized that RAGE and its ligand high-mobility group box-1 (HMGB1) cooperate with TLR4 to mediate HDM sensitization. OBJECTIVES: To determine the requirement for HMGB1 and RAGE, and their relationship with TLR4, in airway sensitization. METHODS: TLR4(-/-), RAGE(-/-), and RAGE-TLR4(-/-) mice were intranasally exposed to HDM or cockroach (Blatella germanica) extracts, and features of allergic inflammation were measured during the sensitization or challenge phase. Anti-HMGB1 antibody and the IL-1 receptor antagonist Anakinra were used to inhibit HMGB1 and the IL-1 receptor, respectively. RESULTS: The magnitude of allergic airway inflammation in response to either HDM or cockroach sensitization and/or challenge was significantly reduced in the absence of RAGE but not further diminished in the absence of both RAGE and TLR4. HDM sensitization induced the release of HMGB1 from the airway epithelium in a biphasic manner, which corresponded to the sequential activation of TLR4 then RAGE. Release of HMGB1 in response to cockroach sensitization also was RAGE dependent. Significantly, HMGB1 release occurred downstream of TLR4-induced IL-1α, and upstream of IL-25 and IL-33 production. Adoptive transfer of HDM-pulsed RAGE(+/+)dendritic cells to RAGE(-/-) mice recapitulated the allergic responses after HDM challenge. Immunoneutralization of HMGB1 attenuated HDM-induced allergic airway inflammation. CONCLUSION: The HMGB1-RAGE axis mediates allergic airway sensitization and airway inflammation. Activation of this axis in response to different allergens acts to amplify the allergic inflammatory response, which exposes it as an attractive target for therapeutic intervention.


Subject(s)
Allergens/immunology , HMGB1 Protein/immunology , Receptors, Immunologic/immunology , Respiratory Hypersensitivity/immunology , Administration, Intranasal , Adoptive Transfer , Allergens/administration & dosage , Animals , Antibodies, Neutralizing/pharmacology , Blattellidae/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/transplantation , Dermatophagoides pteronyssinus/immunology , Gene Expression Regulation , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-33 , Interleukins/genetics , Interleukins/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Mice , Mice, Knockout , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/pathology , Signal Transduction , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
17.
Eur Respir J ; 43(1): 264-75, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23429916

ABSTRACT

The onset, progression and exacerbations of asthma are frequently associated with viral infections of the lower respiratory tract. An emerging paradigm suggests that this relationship may be underpinned by a defect in the host's antiviral response, typified by the impaired production of type I and type III interferons (IFNs). The failure to control viral burden probably causes damage to the lung architecture and contributes to an aberrant immune response, which together compromise lung function. Although a relatively rare cell type, the plasmacytoid dendritic cell dedicates much of its transcriptome to the synthesis of IFNs and is pre-armed with virus-sensing pattern recognition receptors. Thus, plasmacytoid dendritic cells are specialised to ensure early viral detection and the rapid induction of the antiviral state to block viral replication and spread. In addition, plasmacytoid dendritic cells can limit immunopathology, and promote peripheral tolerance to prevent allergic sensitisation to harmless antigens, possibly through the induction of regulatory T-cells. Thus, this enigmatic cell may lie at an important intersection, orchestrating the immediate phase of antiviral immunity to effect viral clearance while regulating tolerance. Here, we review the evidence to support the hypothesis that a primary defect in plasmacytoid dendritic function may underlie the development of asthma.


Subject(s)
Asthma/immunology , Dendritic Cells/immunology , Interferons/immunology , Lung/immunology , Pneumonia, Viral/immunology , Asthma/physiopathology , Humans , Lung/physiopathology , Respiratory Syncytial Virus Infections/immunology , T-Lymphocytes, Regulatory/immunology
18.
Am J Respir Cell Mol Biol ; 49(3): 481-90, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23614768

ABSTRACT

Allergic asthma is a chronic inflammatory disease predominately associated with the activation of CD4(+) T helper Type 2 (Th2) cells. Innate pattern recognition receptors are widely acknowledged to shape the adaptive immune response. For example, the activation of airway epithelial Toll-like receptor-4 (TLR4) is necessary for the generation of house dust mite (HDM)-specific Th2 responses and the development of asthma in mice. Here we sought to determine whether the absence of Toll-interleukin-1 receptor (TIR)-8, a negative regulator of TLR4 signaling that is highly expressed in airway epithelial cells, would exacerbate HDM-induced asthma in a murine model. We found that Th2 but not Th1 or Th17 cytokine expression was significantly reduced in the lung and draining lymph nodes in HDM-sensitized/challenged TIR8 gene-deleted mice. Mucus-producing goblet cells, HDM-specific IgG1, and airway hyperreactivity were also significantly reduced in HDM-exposed, TIR8-deficient mice. Consistent with the attenuated Th2 response, eotaxin-2/CCL24 expression and airway and peribronchial eosinophils were significantly reduced in the absence of TIR8. In contrast, IL-17A-responsive chemokines and neutrophil numbers were unaffected. Similar findings were obtained for cockroach allergen. HDM sensitization alone up-regulated the expression of IL-1F5, a putative TIR8 ligand and inducer of IL-4. Of note, innate IL-4, IL-5, IL-13, and IL-33 cytokine expression was reduced during HDM sensitization in the absence of TIR8, as was the recruitment of conventional dendritic cells and basophils to the draining lymph nodes. Our findings suggest that TIR8 enhances the development of HDM-induced innate and adaptive Th2, but not Th1 or Th17 type immunity.


Subject(s)
Allergens/immunology , Antigens, Dermatophagoides/immunology , Insect Proteins/immunology , Receptors, Interleukin-1/immunology , Respiratory Hypersensitivity/immunology , Adaptive Immunity , Allergens/administration & dosage , Animals , Antigens, Dermatophagoides/administration & dosage , Chemokine CCL24/genetics , Chemokine CCL24/immunology , Eosinophils/immunology , Gene Expression Regulation/immunology , Immunity, Innate , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Insect Proteins/administration & dosage , Interleukin-1/genetics , Interleukin-1/immunology , Lung/immunology , Lymph Nodes/immunology , Male , Mice , Mice, Knockout , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/pathology , Severity of Illness Index , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology
19.
J Allergy Clin Immunol ; 131(5): 1331-9.e10, 2013 May.
Article in English | MEDLINE | ID: mdl-23561801

ABSTRACT

BACKGROUND: Respiratory tract viruses are a major environmental risk factor for both the inception and exacerbations of asthma. Genetic defects in Toll-like receptor (TLR) 7-mediated signaling, impaired type I interferon responses, or both have been reported in asthmatic patients, although their contribution to the onset and exacerbation of asthma remains poorly understood. OBJECTIVE: We sought to determine whether Pneumovirus infection in the absence of TLR7 predisposes to bronchiolitis and the inception of asthma. METHODS: Wild-type and TLR7-deficient (TLR7(-/-)) mice were inoculated with the rodent-specific pathogen pneumonia virus of mice at 1 (primary), 7 (secondary), and 13 (tertiary) weeks of age, and pathologic features of bronchiolitis or asthma were assessed. In some experiments infected mice were exposed to low-dose cockroach antigen. RESULTS: TLR7 deficiency increased viral load in the airway epithelium, which became sloughed and necrotic, and promoted an IFN-α/ß(low), IL-12p70(low), IL-1ß(high), IL-25(high), and IL-33(high) cytokine microenvironment that was associated with the recruitment of type 2 innate lymphoid cells/nuocytes and increased TH2-type cytokine production. Viral challenge of TLR7(-/-) mice induced all of the cardinal pathophysiologic features of asthma, including tissue eosinophilia, mast cell hyperplasia, IgE production, airway smooth muscle alterations, and airways hyperreactivity in a memory CD4(+) T cell-dependent manner. Importantly, infections with pneumonia virus of mice promoted allergic sensitization to inhaled cockroach antigen in the absence but not the presence of TLR7. CONCLUSION: TLR7 gene defects and Pneumovirus infection interact to establish an aberrant adaptive response that might underlie virus-induced asthma exacerbations in later life.


Subject(s)
Asthma/immunology , Asthma/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Murine pneumonia virus , Pneumovirus Infections/complications , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/genetics , Animals , Animals, Newborn , Asthma/etiology , Disease Models, Animal , Lung/pathology , Lung/physiopathology , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Murine pneumonia virus/pathogenicity , Pneumovirus Infections/immunology , Pneumovirus Infections/pathology , Viral Load
20.
J Immunol ; 186(10): 5938-48, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21482736

ABSTRACT

Human respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection in infants. In human infants, plasmacytoid dendritic cells (pDC) are recruited to the nasal compartment during infection and initiate host defense through the secretion of type I IFN, IL-12, and IL-6. However, RSV-infected pDC are refractory to TLR7-mediated activation. In this study, we used the rodent-specific pathogen, pneumonia virus of mice (PVM), to determine the contribution of pDC and TLR7 signaling to the development of the innate inflammatory and early adaptive immune response. In wild-type, but not TLR7- or MyD88-deficient mice, PVM inoculation led to a marked infiltration of pDC and increased expression of type I, II, and III IFNs. The delayed induction of IFNs in the absence of TLR7 or MyD88 was associated with a diminished innate inflammatory response and augmented virus recovery from lung tissue. In the absence of TLR7, PVM-specific CD8(+) T cell cytokine production was abrogated. The adoptive transfer of TLR7-sufficient, but not TLR7-deficient pDC to TLR7 gene-deleted mice recapitulated the antiviral responses observed in wild-type mice and promoted virus clearance. In summary, TLR7-mediated signaling by pDC is required for appropriate innate responses to acute pneumovirus infection. It is conceivable that as-yet-unidentified defects in the TLR7 signaling pathway may be associated with elevated levels of RSV-associated morbidity and mortality among otherwise healthy human infants.


Subject(s)
Dendritic Cells/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Murine pneumonia virus/immunology , Myeloid Differentiation Factor 88/metabolism , Pneumovirus Infections/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Adaptive Immunity , Adoptive Transfer , Animals , Interferons/genetics , Interferons/immunology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Signal Transduction , Toll-Like Receptor 7/genetics
SELECTION OF CITATIONS
SEARCH DETAIL