Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Allergy Clin Immunol ; 140(1): 190-203.e5, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27865862

ABSTRACT

BACKGROUND: Type 2 airway inflammation plays a central role in the pathogenesis of allergen-induced asthma, but the underlying mechanisms remain poorly understood. Recently, we demonstrated that reduced mucociliary clearance, a characteristic feature of asthma, produces spontaneous type 2 airway inflammation in juvenile ß-epithelial Na+ channel (Scnn1b)-transgenic (Tg) mice. OBJECTIVE: We sought to determine the role of impaired mucus clearance in the pathogenesis of allergen-induced type 2 airway inflammation and identify cellular sources of the signature cytokine IL-13. METHODS: We challenged juvenile Scnn1b-Tg and wild-type mice with Aspergillus fumigatus and house dust mite allergen and compared the effects on airway eosinophilia, type 2 cytokine levels, goblet cell metaplasia, and airway hyperresponsiveness. Furthermore, we determined cellular sources of IL-13 and effects of genetic deletion of the key type 2 signal-transducing molecule signal transducer and activator of transcription 6 (STAT6) and evaluated the effects of therapeutic improvement of mucus clearance. RESULTS: Reduced mucociliary allergen clearance exacerbated Stat6-dependent secretion of type 2 cytokines, airway eosinophilia, and airway hyperresponsiveness in juvenile Scnn1b-Tg mice. IL-13 levels were increased in airway epithelial cells, macrophages, type 2 innate lymphoid cells, and TH2 cells along with increased Il33 expression in the airway epithelium of Scnn1b-Tg mice. Treatment with the epithelial Na+ channel blocker amiloride, improving airway surface hydration and mucus clearance, reduced allergen-induced inflammation in Scnn1b-Tg mice. CONCLUSION: Our data support that impaired clearance of inhaled allergens triggering IL-13 production by multiple cell types in the airways plays an important role in the pathogenesis of type 2 airway inflammation and suggests therapeutic improvement of mucociliary clearance as a novel treatment strategy for children with allergen-induced asthma.


Subject(s)
Asthma/immunology , Asthma/physiopathology , Interleukin-13/immunology , Mucociliary Clearance , Allergens/immunology , Amiloride/pharmacology , Amiloride/therapeutic use , Animals , Aspergillus fumigatus/immunology , Asthma/drug therapy , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Sodium Channels/genetics , Lung/cytology , Lung/immunology , Mice, Transgenic , Pyroglyphidae/immunology , STAT6 Transcription Factor/genetics , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use
2.
J Innate Immun ; 8(5): 452-63, 2016.
Article in English | MEDLINE | ID: mdl-27423203

ABSTRACT

Respiratory syncytial virus (RSV) is a common cause of lower respiratory tract infections. Immunity to RSV is initiated upon detection of the virus by pattern recognition receptors, such as RIG-I-like receptors. RIG-I-like receptors signal via MAVS to induce the synthesis of proinflammatory mediators, including type I interferons (IFNs), which trigger and shape antiviral responses and protect cells from infection. Alveolar macrophages (AMs) are amongst the first cells to encounter invading viruses and the ones producing type I IFNs. However, it is unclear whether IFNs act to prevent AMs from serving as vehicles for viral replication. In this study, primary AMs from MAVS (Mavs-/-)- or type I IFN receptor (Ifnar1-/-)-deficient mice were exposed to RSV ex vivo. Wild-type (wt) AMs but not Mavs-/- and Ifnar1-/- AMs produced inflammatory mediators in response to RSV. Furthermore, Mavs-/- and Ifnar1-/- AMs accumulated more RSV proteins than wt AMs, but the infection was abortive. Thus, RIG-I-like receptor-MAVS and IFNAR signalling are important for the induction of proinflammatory mediators from AMs upon RSV infection, but this signalling is not central for controlling viral replication. The ability to restrict viral replication makes AMs ideal sensors of RSV infection and important initiators of immune responses in the lung.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Macrophages, Alveolar/immunology , Receptor, Interferon alpha-beta/metabolism , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Cytokines/metabolism , Inflammation Mediators/metabolism , Interferon Type I/metabolism , Macrophages, Alveolar/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Virus Replication
3.
Sci Rep ; 5: 18533, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26688048

ABSTRACT

Pattern recognition receptors (PRRs) and cytokine receptors are key players in the initiation of immune responses to infection. PRRs detecting viral RNA, such as toll like receptor (TLR)-3, -7/8, and RIG-I like receptors (RLRs; RIG-I and MDA-5), as well as cytokine receptors such as interleukin 1 receptor (IL-1R), have been implicated in responses to RNA viruses that infect the airways. The latter includes respiratory syncytial virus (RSV), a human pathogen that can cause severe lower respiratory tract infections, especially in infants. To evaluate the collective contribution of PRRs and IL-1R signalling to RSV immunity, we generated Myd88/Trif/Mavs(-/-) mice that are deficient in signalling by all TLRs, RLRs and IL-1R, as well as other cytokine receptors such as IL-18 receptor. Early production of pro-inflammatory mediators and lung infiltration by immune cells were completely abrogated in infected Myd88/Trif/Mavs(-/-) mice. However, RSV-specific CD8(+) T cells were elicited and recruited into the lungs and airways. Consistent with these findings, Myd88/Trif/Mavs(-/-) mice survived RSV infection but displayed higher viral load and weight loss. These data highlight an unappreciated level of redundancy in pathways that couple innate virus sensing to adaptive immunity, providing the host with remarkable resilience to infection.


Subject(s)
Infections/genetics , Receptors, Interleukin-1 Type II/genetics , Receptors, Interleukin-18/genetics , Respiratory Syncytial Virus Infections/genetics , Respiratory Tract Infections/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Vesicular Transport/genetics , Animals , Humans , Infections/immunology , Infections/virology , Mice , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , Receptors, Interleukin-18/immunology , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Viral Load
4.
J Exp Med ; 212(5): 699-714, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25897172

ABSTRACT

Type I interferons (IFNs) are important for host defense from viral infections, acting to restrict viral production in infected cells and to promote antiviral immune responses. However, the type I IFN system has also been associated with severe lung inflammatory disease in response to respiratory syncytial virus (RSV). Which cells produce type I IFNs upon RSV infection and how this directs immune responses to the virus, and potentially results in pathological inflammation, is unclear. Here, we show that alveolar macrophages (AMs) are the major source of type I IFNs upon RSV infection in mice. AMs detect RSV via mitochondrial antiviral signaling protein (MAVS)-coupled retinoic acid-inducible gene 1 (RIG-I)-like receptors (RLRs), and loss of MAVS greatly compromises innate immune restriction of RSV. This is largely attributable to loss of type I IFN-dependent induction of monocyte chemoattractants and subsequent reduced recruitment of inflammatory monocytes (infMo) to the lungs. Notably, the latter have potent antiviral activity and are essential to control infection and lessen disease severity. Thus, infMo recruitment constitutes an important and hitherto underappreciated, cell-extrinsic mechanism of type I IFN-mediated antiviral activity. Dysregulation of this system of host antiviral defense may underlie the development of RSV-induced severe lung inflammation.


Subject(s)
Immunity, Innate , Macrophages, Alveolar/immunology , Pneumonia, Viral/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Interferon Type I/genetics , Interferon Type I/immunology , Macrophages, Alveolar/pathology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Pneumonia, Viral/genetics , Pneumonia, Viral/pathology , Receptors, Cell Surface , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/pathology
5.
Eur J Immunol ; 44(8): 2340-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24777856

ABSTRACT

During respiratory syncytial virus (RSV) infection CD8(+) T cells both assist in viral clearance and contribute to immunopathology. CD8(+) T cells recognize viral peptides presented by dendritic cells (DCs), which can directly present viral antigens when infected or, alternatively, "cross-present" antigens after endocytosis of dead or dying infected cells. Mouse CD8α(+) and CD103(+) DCs excel at cross-presentation, in part because they express the receptor DNGR-1 that detects dead cells by binding to exposed F-actin and routes internalized cell debris into the cross-presentation pathway. As RSV causes death in infected epithelial cells, we tested whether cross-presentation via DNGR-1 is necessary for CD8(+) T-cell responses to the virus. DNGR-1-deficient or wild-type mice were intranasally inoculated with RSV and the magnitude of RSV-specific CD8(+) T-cell induction was measured. We found that during live RSV infection, cross-presentation via DNGR-1 did not have a major role in the generation of RSV-specific CD8(+) T-cell responses. However, after intranasal immunization with dead cells infected with RSV, a dependence on DNGR-1 for RSV-specific CD8(+) T-cell responses was observed, confirming the ascribed role of the receptor. Thus, direct presentation by DCs may be the major pathway initiating CD8(+) T-cell responses to RSV, while DNGR-1-dependent cross-presentation has no detectable role.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lectins, C-Type/immunology , Receptors, Immunologic/immunology , Respiratory Syncytial Virus Infections/immunology , Actins/immunology , Animals , Antigen Presentation/immunology , Antigens, Viral/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Epithelial Cells/immunology , Epithelial Cells/virology , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Respiratory Syncytial Viruses/immunology , Viral Load/immunology
6.
J Virol ; 88(11): 6128-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24648449

ABSTRACT

UNLABELLED: Type I interferons (IFNs) are produced early upon virus infection and signal through the alpha/beta interferon (IFN-α/ß) receptor (IFNAR) to induce genes that encode proteins important for limiting viral replication and directing immune responses. To investigate the extent to which type I IFNs play a role in the local regulation of inflammation in the airways, we examined their importance in early lung responses to infection with respiratory syncytial virus (RSV). IFNAR1-deficient (IFNAR1(-/-)) mice displayed increased lung viral load and weight loss during RSV infection. As expected, expression of IFN-inducible genes was markedly reduced in the lungs of IFNAR1(-/-) mice. Surprisingly, we found that the levels of proinflammatory cytokines and chemokines in the lungs of RSV-infected mice were also greatly reduced in the absence of IFNAR signaling. Furthermore, low levels of proinflammatory cytokines were also detected in the lungs of IFNAR1(-/-) mice challenged with noninfectious innate immune stimuli such as selected Toll-like receptor (TLR) agonists. Finally, recombinant IFN-α was sufficient to potentiate the production of inflammatory mediators in the lungs of wild-type mice challenged with innate immune stimuli. Thus, in addition to its well-known role in antiviral resistance, type I IFN receptor signaling acts as a central driver of early proinflammatory responses in the lung. Inhibiting the effects of type I IFNs may therefore be useful in dampening inflammation in lung diseases characterized by enhanced inflammatory cytokine production. IMPORTANCE: The initial response to viral infection is characterized by the production of interferons (IFNs). One group of IFNs, the type I IFNs, are produced early upon virus infection and signal through the IFN-α/ß receptor (IFNAR) to induce proteins important for limiting viral replication and directing immune responses. Here we examined the importance of type I IFNs in early responses to respiratory syncytial virus (RSV). Our data suggest that type I IFN production and IFNAR receptor signaling not only induce an antiviral state but also serve to amplify proinflammatory responses in the respiratory tract. We also confirm this conclusion in another model of acute inflammation induced by noninfectious stimuli. Our findings are of relevance to human disease, as RSV is a major cause of infant bronchiolitis and polymorphisms in the IFN system are known to impact disease severity.


Subject(s)
Cytokines/biosynthesis , Gene Expression Regulation/immunology , Lung/metabolism , Respiratory Syncytial Virus Infections/immunology , Signal Transduction/physiology , Animals , DNA Primers/genetics , Lung/virology , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Statistics, Nonparametric , Viral Load
7.
Proc Natl Acad Sci U S A ; 110(14): 5576-81, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23509276

ABSTRACT

Respiratory syncytial virus (RSV) infects most children in the first year of life and is a major single cause of hospitalization in infants and young children. There is no effective vaccine, and antibody generated by primary neonatal infection is poorly protective against reinfection even with antigenically homologous viral strains. Studying the immunological basis of these observations in neonatal mice, we found that antibody responses to infection were low and unaffected by CD4 depletion, in contrast with adult mice, which had stronger CD4-dependent antibody responses. Natural killer cell depletion or codepletion of CD4(+) and CD8(+) cells during neonatal RSV infection caused a striking increase in anti-RSV antibody titer. These cells are major sources of the cytokine IFN-γ, and blocking IFN-γ also enhanced RSV-specific antibody responses in neonates. In addition, infection with a recombinant RSV engineered to produce IFN-γ reduced antibody titer, confirming that IFN-γ plays a pivotal role in inhibition of antibody responses after neonatal infection. These unexpected findings show that the induction of a strong cellular immune response may limit antibody responses in early life and that vaccines that induce IFN-γ-secreting cells might, in some situations, be less protective than those that do not.


Subject(s)
Animals, Newborn , Antibodies, Viral/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Respiratory Syncytial Virus Infections/immunology , T-Lymphocytes/immunology , Analysis of Variance , Animals , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pregnancy
8.
J Proteome Res ; 10(5): 2161-71, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21417265

ABSTRACT

Dilated cardiomyopathy (DCM) is characterized by contractile dysfunction leading to heart failure. The molecular changes in the human heart associated with this disease have so far mostly been addressed at the gene expression level and only a few studies have analyzed global changes in the myocardial proteome. Therefore, our objective was to investigate the changes in the proteome in patients suffering from inflammatory DCM (iDCM) and chronic viral infection by a comprehensive quantitative approach. Comparative proteomic profiling of endomyocardial biopsies (EMB) from 10 patients with iDCM (left ventricular ejection fraction <40%, symptoms of heart failure) as well as 7 controls with normal left ventricular function and histology was performed by label-free proteome analysis (LC-MS/MS). Mass spectrometric data were analyzed with the Rosetta Elucidator software package. The analysis covered a total of 485 proteins. Among the 174 proteins displaying at least a 1.3-fold change in intensity (p < 0.05), major changes were observed for mitochondrial and cytoskeletal proteins, but also metabolic pathways were affected in iDCM compared to controls. In iDCM patients, we observed decreased levels of mitochondrial proteins involved in oxidative phosphorylation and tricarboxylic acid cycle. Furthermore, deregulation of proteins of carbohydrate metabolism, the actin cytoskeleton, and extracellular matrix remodeling was observed. Proteomic observations were confirmed by gene expression data and immunohistochemistry (e.g. collagen I and VI). This study demonstrates that label-free, mass spectrometry-centered approaches can identify disease dependent alterations in the proteome from small tissue samples such as endomyocardial biopsies. Thus, this technique might allow better disease characterization and may be a valuable tool in potential clinical proteomic studies.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Gene Expression Regulation/genetics , Myocarditis/metabolism , Myocarditis/virology , Myocardium/metabolism , Proteomics/methods , Biopsy , Cardiomyopathy, Dilated/pathology , Chromatography, Liquid , Computational Biology/methods , Gene Expression Profiling , Heart Ventricles/physiopathology , Humans , Immunohistochemistry , Microarray Analysis , Myocarditis/pathology , Myocardium/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...