Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Immunity ; 55(3): 542-556.e5, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35151371

ABSTRACT

Some patients hospitalized with acute COVID-19 suffer respiratory symptoms that persist for many months. We delineated the immune-proteomic landscape in the airways and peripheral blood of healthy controls and post-COVID-19 patients 3 to 6 months after hospital discharge. Post-COVID-19 patients showed abnormal airway (but not plasma) proteomes, with an elevated concentration of proteins associated with apoptosis, tissue repair, and epithelial injury versus healthy individuals. Increased numbers of cytotoxic lymphocytes were observed in individuals with greater airway dysfunction, while increased B cell numbers and altered monocyte subsets were associated with more widespread lung abnormalities. A one-year follow-up of some post-COVID-19 patients indicated that these abnormalities resolved over time. In summary, COVID-19 causes a prolonged change to the airway immune landscape in those with persistent lung disease, with evidence of cell death and tissue repair linked to the ongoing activation of cytotoxic T cells.


Subject(s)
B-Lymphocytes/immunology , COVID-19/immunology , Monocytes/immunology , Respiration Disorders/immunology , Respiratory System/immunology , SARS-CoV-2/physiology , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , COVID-19/complications , Female , Follow-Up Studies , Humans , Immunity, Cellular , Immunoproteins , Male , Middle Aged , Proteome , Respiration Disorders/etiology , Respiratory System/pathology
2.
Am J Respir Crit Care Med ; 208(10): 1115-1125, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37713301

ABSTRACT

Rationale: Mounting evidence demonstrates a role for extracellular vesicles (EVs) in driving lung disorders, such as chronic obstructive pulmonary disease (COPD). Although cigarette smoke (CS) is the primary risk factor for COPD, a link between CS and the EVs that could lead to COPD is unknown. Objective: To ascertain whether exposure to CS elicits a proteolytic EV signature capable of driving disease pathogenesis. Methods: Protease expression and enzymatic activity were measured in EVs harvested from the BAL fluid of smoke-exposed mice and otherwise healthy human smokers. Pathogenicity of EVs was examined using pathological tissue scoring after EV transfer into naive recipient mice. Measurements and Main Results: The analyses revealed a unique EV profile defined by neutrophil- and macrophage-derived EVs. These EVs are characterized by abundant surface expression of neutrophil elastase (NE) and matrix metalloproteinase 12 (MMP12), respectively. CS-induced mouse or human-derived airway EVs had a robust capacity to elicit rapid lung damage in naive recipient mice, with an additive effect of NE- and MMP12-expressing EVs. Conclusions: These studies demonstrate the capacity of CS to drive the generation of unique EV populations containing NE and MMP12. The coordinated action of these EVs is completely sufficient to drive emphysematous disease, and their presence could operate as a prognostic indicator for COPD development. Furthermore, given the robust capacity of these EVs to elicit emphysema in naive mice, they provide a novel model to facilitate preclinical COPD research. Indeed, the development of this model has led to the discovery of a previously unrecognized CS-induced protective mechanism against EV-mediated damage.


Subject(s)
Emphysema , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Animals , Mice , Peptide Hydrolases/metabolism , Matrix Metalloproteinase 12/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Lung , Pulmonary Emphysema/etiology , Pancreatic Elastase/metabolism , Smoking/adverse effects , Disease Models, Animal
3.
Cytometry A ; 103(2): 110-116, 2023 02.
Article in English | MEDLINE | ID: mdl-36331092

ABSTRACT

This 25-parameter, 22-color full spectrum flow cytometry panel was designed and optimized for the comprehensive enumeration and functional characterization of innate lymphoid cell (ILC) subsets in mouse tissues. The panel presented here allows the discrimination of ILC progenitors (ILCP), ILC1, ILC2, NCR+ ILC3, NCR- ILC3, CCR6+ lymphoid tissue-inducer (LTi)-like ILC3 and mature natural killer (NK) cell populations. Further characterization of ILC and NK cell functional profiles in response to stimulation is provided by the inclusion of subset-specific cytokine markers, and proliferation markers. Development and optimization of this panel was performed on freshly isolated cells from adult BALB/c lungs and small intestine lamina propria, and ex vivo stimulation with phorbol 12-myrisate 13-acetate, ionomycin, and pro-ILC activating cytokines.


Subject(s)
Immunity, Innate , Lymphocytes , Mice , Animals , Immunophenotyping , Flow Cytometry , Killer Cells, Natural , Cytokines
4.
Immunology ; 164(4): 701-721, 2021 12.
Article in English | MEDLINE | ID: mdl-34547115

ABSTRACT

Neutrophils are critical components of the body's immune response to infection, being loaded with a potent arsenal of toxic mediators and displaying immense destructive capacity. Given the potential of neutrophils to impart extensive tissue damage, it is perhaps not surprising that when augmented these cells are also implicated in the pathology of inflammatory diseases. Prominent neutrophilic inflammation is a hallmark feature of patients with chronic lung diseases such as chronic obstructive pulmonary disease, severe asthma, bronchiectasis and cystic fibrosis, with their numbers frequently associating with worse prognosis. Accordingly, it is anticipated that neutrophils are central to the pathology of these diseases and represent an attractive therapeutic target. However, in many instances, evidence directly linking neutrophils to the pathology of disease has remained somewhat circumstantial and strategies that have looked to reduce neutrophilic inflammation in the clinic have proved largely disappointing. We have classically viewed neutrophils as somewhat crude, terminally differentiated, insular and homogeneous protagonists of pathology. However, it is now clear that this does not do the neutrophil justice, and we now recognize that these cells exhibit heterogeneity, a pronounced awareness of the localized environment and a remarkable capacity to interact with and modulate the behaviour of a multitude of cells, even exhibiting anti-inflammatory, pro-resolving and pro-repair functions. In this review, we discuss evidence for the role of neutrophils in chronic lung disease and how our evolving view of these cells may impact upon our perceived assessment of their contribution to disease pathology and efforts to target them therapeutically.


Subject(s)
Disease Susceptibility , Lung Diseases/etiology , Lung Diseases/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Animals , Biomarkers , Cell Plasticity/immunology , Chronic Disease , Diagnosis, Differential , Gene Expression Regulation , Humans , Lung Diseases/diagnosis , Neutrophils/pathology , Organ Specificity
5.
Eur J Immunol ; 49(9): 1380-1390, 2019 09.
Article in English | MEDLINE | ID: mdl-31365119

ABSTRACT

CD200 receptor 1(CD200R1) signalling limits myeloid cell responses and reduces autoimmunity, alloimmunity and viral-mediated immunopathology, but has never been examined in the context of eosinophilic inflammation. Susceptibility to lung fungal infection is associated with T-helper 2 (Th2) cytokine dominated responses and strong eosinophilic pathology. Blockade of CD200R1 enhances type I cytokine responses in many infectious and non-infectious settings and so may promote a more protective response to fungal infection. By contrast, we demonstrate that, rather than promoting type I cytokine responses, CD200R1 blockade enhanced eosinophilia in a mouse model of Cryptococcus neoformans infection, whereas CD200R1 agonism reduced lung eosinophilia - with neither strategy completely altering fungal burden. Thus, we reveal a surprising disconnect between pulmonary eosinophilia and cryptococcal burden and dissemination. This research has 2 important implications. Firstly, a lack of CD200R1 signalling enhances immune responses regardless of cytokine polarisation, and secondly reducing eosinophils does not allow protective immunity to develop in susceptible fungal system. Therefore, agonists of CD200R1 may be beneficial for eosinophilic pathologies.


Subject(s)
Lung Diseases, Fungal/immunology , Orexin Receptors/immunology , Pulmonary Eosinophilia/immunology , Animals , Cryptococcosis/immunology , Cryptococcosis/microbiology , Cryptococcus neoformans/immunology , Cytokines/immunology , Disease Models, Animal , Inflammation/immunology , Inflammation/microbiology , Lung , Lung Diseases, Fungal/microbiology , Mice , Myeloid Cells/immunology , Myeloid Cells/microbiology , Pulmonary Eosinophilia/microbiology , Th2 Cells/immunology , Th2 Cells/microbiology
6.
Nat Immunol ; 9(9): 1074-83, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18660812

ABSTRACT

The lung must maintain a high threshold of immune 'ignorance' to innocuous antigens to avoid inflammatory disease that depends on the balance of positive inflammatory signals and repressor pathways. We demonstrate here that airway macrophages had higher expression of the negative regulator CD200 receptor (CD200R) than did their systemic counterparts. Lung macrophages were restrained by CD200 expressed on airway epithelium. Mice lacking CD200 had more macrophage activity and enhanced sensitivity to influenza infection, which led to delayed resolution of inflammation and, ultimately, death. The administration of agonists that bind CD200R, however, prevented inflammatory lung disease. Thus, CD200R is critical for lung macrophage immune homeostasis in the resting state and limits inflammatory amplitude and duration during pulmonary influenza infection.


Subject(s)
Antigens, CD/immunology , Homeostasis/physiology , Influenza, Human/immunology , Lung/immunology , Myeloid Cells/immunology , Animals , Cytokines/biosynthesis , Homeostasis/immunology , Humans , Influenza, Human/pathology , Lung/metabolism , Mice
8.
Thorax ; 73(6): 546-556, 2018 06.
Article in English | MEDLINE | ID: mdl-29079611

ABSTRACT

BACKGROUND: Alveolar macrophages are sentinels of the airways that must exhibit immune restraint to innocuous antigens but elicit a robust inflammatory response to pathogenic threats. How distinction between these dichotomous functions is controlled is poorly defined.Neutrophils are the first responders to infection, and we hypothesised that they may free alveolar macrophages from their hyporesponsive state, promoting their activation. Activation of the inflammasome and interleukin (IL)-1ß release is a key early inflammatory event that must be tightly regulated. Thus, the role of neutrophils in defining inflammasome activation in the alveolar macrophage was assessed. METHODS: Mice were infected with the X31 strain of influenza virus and the role of neutrophils in alveolar macrophage activation established through administration of a neutrophil-depleting (1A8) antibody. RESULTS: Influenza elicited a robust IL-1ß release that correlated (r=0.6849; p<0.001) with neutrophil infiltrate and was ablated by neutrophil depletion. Alveolar macrophages were shown to be the prominent source of IL-1ß during influenza infection, and virus triggered the expression of Nod-like receptor protein 3 (NLRP3) inflammasome and pro-IL-1ß in these cells. However, subsequent activation of the inflammasome complex and release of mature IL-1ß from alveolar macrophages were critically dependent on the provision of a secondary signal, in the form of antimicrobial peptide mCRAMP, from infiltrating neutrophils. CONCLUSIONS: Neutrophils are critical for the activation of the NLRP3 inflammasome in alveolar macrophages during respiratory viral infection. Accordingly, we rationalise that neutrophils are recruited to the lung to confront a viable pathogenic threat and subsequently commit alveolar macrophages to a pro-inflammatory phenotype to combat infection.


Subject(s)
Interleukin-1beta/immunology , Macrophages, Alveolar/immunology , Neutrophils/immunology , Respiratory Tract Infections/immunology , Virus Diseases/immunology , Animals , Female , Inflammasomes/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Respiratory Tract Infections/virology
11.
PLoS Pathog ; 11(2): e1004641, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25654642

ABSTRACT

CD200 receptor (CD200R) negatively regulates peripheral and mucosal innate immune responses. Viruses, including herpesviruses, have acquired functional CD200 orthologs, implying that viral exploitation of this pathway is evolutionary advantageous. However, the role that CD200R signaling plays during herpesvirus infection in vivo requires clarification. Utilizing the murine cytomegalovirus (MCMV) model, we demonstrate that CD200R facilitates virus persistence within mucosal tissue. Specifically, MCMV infection of CD200R-deficient mice (CD200R(-/-)) elicited heightened mucosal virus-specific CD4 T cell responses that restricted virus persistence in the salivary glands. CD200R did not directly inhibit lymphocyte effector function. Instead, CD200R(-/-) mice exhibited enhanced APC accumulation that in the mucosa was a consequence of elevated cellular proliferation. Although MCMV does not encode an obvious CD200 homolog, productive replication in macrophages induced expression of cellular CD200. CD200 from hematopoietic and non-hematopoietic cells contributed independently to suppression of antiviral control in vivo. These results highlight the CD200-CD200R pathway as an important regulator of antiviral immunity during cytomegalovirus infection that is exploited by MCMV to establish chronicity within mucosal tissue.


Subject(s)
Antigens, CD/immunology , Cytomegalovirus Infections/immunology , Macrophages/immunology , Mucous Membrane/immunology , Mucous Membrane/virology , Animals , Cytomegalovirus/immunology , Cytomegalovirus Infections/metabolism , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Macrophages/metabolism , Macrophages/virology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis
12.
Respir Res ; 18(1): 55, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28399855

ABSTRACT

BACKGROUND: The incidence of asthma is increasing at an alarming rate. While the current available therapies are effective, there are associated side effects and they fail to adequately control symptoms in all patient subsets. In the search to understand disease pathogenesis and find effective therapies hypotheses are often tested in animal models before progressing into clinical studies. However, current dogma is that animal model data is often not predictive of clinical outcome. One possible reason for this is the end points measured such as antigen-challenge induced late asthmatic response (LAR) is often used in early clinical development, but seldom in animal model systems. As the mouse is typically selected as preferred species for pre-clinical models, we wanted to characterise and probe the validity of a murine model exhibiting an allergen induced LAR. METHODS: C57BL/6 mice were sensitised with antigen and subsequently topically challenged with the same antigen. The role of AlumTM adjuvant, glucocorticoid, long acting muscarinic receptor antagonist (LAMA), TRPA1, CD4+ and CD8+ T cells, B cells, Mast cells and IgE were determined in the LAR using genetically modified mice and a range of pharmacological tools. RESULTS: Our data showed that unlike other features of asthma (e.g. cellular inflammation, elevated IgE levels and airway hyper-reactivity (AHR) the LAR required AlumTMadjuvant. Furthermore, the LAR appeared to be sensitive to glucocorticoid and required CD4+ T cells. Unlike in other species studied, the LAR was not sensitive to LAMA treatment nor required the TRPA1 ion channel, suggesting that airway sensory nerves are not involved in the LAR in this species. Furthermore, the data suggested that CD8+ T cells and the mast cell-B-cell - IgE axis appear to be protective in this murine model. CONCLUSION: Together we can conclude that this model does feature steroid sensitive, CD4+ T cell dependent, allergen induced LAR. However, collectively our data questions the validity of using the murine pre-clinical model of LAR in the assessment of future asthma therapies.


Subject(s)
Antigens/immunology , Asthma/immunology , Cytokines/immunology , Disease Models, Animal , Immunity, Innate/immunology , Animals , Male , Mice , Mice, Inbred C57BL , Time Factors
13.
Proc Natl Acad Sci U S A ; 111(6): 2289-94, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24469824

ABSTRACT

An immune response is essential for protection against infection, but, in many individuals, aberrant responses against self tissues cause autoimmune diseases such as rheumatoid arthritis (RA). How to diminish the autoimmune response while not augmenting infectious risk is a challenge. Modern targeted therapies such as anti-TNF or anti-CD20 antibodies ameliorate disease, but at the cost of some increase in infectious risk. Approaches that might specifically reduce autoimmunity and tissue damage without infectious risk would be important. Here we describe that TNF superfamily member OX40 ligand (OX40L; CD252), which is expressed predominantly on antigen-presenting cells, and its receptor OX40 (on activated T cells), are restricted to the inflamed joint in arthritis in mice with collagen-induced arthritis and humans with RA. Blockade of this pathway in arthritic mice reduced inflammation and restored tissue integrity predominantly by inhibiting inflammatory cytokine production by OX40L-expressing macrophages. Furthermore, we identify a previously unknown role for OX40L in steady-state bone homeostasis. This work shows that more targeted approaches may augment the "therapeutic window" and increase the benefit/risk in RA, and possibly other autoimmune diseases, and are thus worth testing in humans.


Subject(s)
Arthritis, Rheumatoid/therapy , Membrane Glycoproteins/immunology , Osteoclasts/cytology , Tumor Necrosis Factors/immunology , Animals , Antibodies, Monoclonal/immunology , Arthritis, Rheumatoid/pathology , Cytokines/biosynthesis , Homeostasis , Inflammation Mediators/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Mice , OX40 Ligand , Signal Transduction , Tumor Necrosis Factor Inhibitors
14.
Respir Res ; 17(1): 67, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27255083

ABSTRACT

BACKGROUND: Asthma prevalence has increased world-wide especially in children; thus there is a need to develop new therapies that are safe and effective especially for patients with severe/refractory asthma. CD4(+) T cells are thought to play a central role in disease pathogenesis and associated symptoms. Recently, TRPV1 has been demonstrated to regulate the activation and inflammatory properties of CD4(+) cells. The aim of these experiments was to demonstrate the importance of CD4(+) T cells and the role of TRPV1 in an asthma model using a clinically ready TRPV1 inhibitor (XEN-D0501) and genetically modified (GM) animals. METHODS: Mice (wild type, CD4 (-/-) or TRPV1 (-/-)) and rats were sensitised with antigen (HDM or OVA) and subsequently topically challenged with the same antigen. Key features associated with an allergic asthma type phenotype were measured: lung function (airway hyperreactivity [AHR] and late asthmatic response [LAR]), allergic status (IgE levels) and airway inflammation. RESULTS: CD4(+) T cells play a central role in both disease model systems with all the asthma-like features attenuated. Targeting TRPV1 using either GM mice or a pharmacological inhibitor tended to decrease IgE levels, airway inflammation and lung function changes. CONCLUSION: Our data suggests the involvement of TRPV1 in allergic asthma and thus we feel this target merits further investigation.


Subject(s)
Asthma/metabolism , Lung/metabolism , Pneumonia/metabolism , TRPV Cation Channels/metabolism , Animals , Anti-Allergic Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Asthma/chemically induced , Asthma/immunology , Asthma/prevention & control , CD4 Antigens/genetics , CD4 Antigens/metabolism , Disease Models, Animal , Female , Genetic Predisposition to Disease , Immunoglobulin E/metabolism , Lung/drug effects , Lung/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin , Phenotype , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/prevention & control , Pyroglyphidae/immunology , Rats, Inbred BN , Signal Transduction , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
16.
J Allergy Clin Immunol ; 136(2): 312-22.e7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25746970

ABSTRACT

BACKGROUND: The mechanism underlying severe asthma with fungal sensitization (SAFS) is unknown. IL-33 is important in fungus-induced asthma exacerbations, but its role in fungal sensitization is unexplored. OBJECTIVE: We sought to determine whether fungal sensitization in children with severe therapy-resistant asthma is mediated by IL-33. METHODS: Eighty-two children (median age, 11.7 years; 63% male) with severe therapy-resistant asthma were included. SAFS (n = 38) was defined as specific IgE or skin prick test response positivity to Aspergillus fumigatus, Alternaria alternata, or Cladosporium herbarum. Clinical features and airway immunopathology were assessed. Chronic exposure to house dust mite and A alternata were compared in a neonatal mouse model. RESULTS: Children with SAFS had earlier symptom onset (0.5 vs 1.5 years, P = .006), higher total IgE levels (637 vs 177 IU/mL, P = .002), and nonfungal inhalant allergen-specific IgE. Significantly more children with SAFS were prescribed maintenance oral steroids (42% vs 14%, P = .02). SAFS was associated with higher airway IL-33 levels. In neonatal mice A alternata exposure induced higher serum IgE levels, pulmonary IL-33 levels, and IL-13(+) innate lymphoid cell (ILC) and TH2 cell numbers but similar airway hyperresponsiveness (AHR) compared with those after house dust mite exposure. Lung IL-33 levels, IL-13(+) ILC numbers, TH2 cell numbers, IL-13 levels, and AHR remained increased with inhaled budesonide during A alternata exposure, but all features were significantly reduced in ST2(-/-) mice lacking a functional receptor for IL-33. CONCLUSION: Pediatric SAFS was associated with more oral steroid therapy and higher IL-33 levels. A alternata exposure resulted in increased IL-33-mediated ILC2 numbers, TH2 cell numbers, and steroid-resistant AHR. IL-33 might be a novel therapeutic target for SAFS.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Asthma/immunology , Budesonide/therapeutic use , Interleukins/immunology , Mycoses/drug therapy , Mycoses/immunology , Adolescent , Alternaria/immunology , Animals , Animals, Newborn , Antibodies, Anti-Idiotypic/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Aspergillus fumigatus/immunology , Asthma/complications , Asthma/pathology , Child , Cladosporium/immunology , Disease Models, Animal , Female , Humans , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-33 , Interleukins/genetics , Male , Mice , Mycoses/complications , Mycoses/pathology , Omalizumab , Pyroglyphidae/chemistry , Pyroglyphidae/immunology , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Severity of Illness Index , Skin Tests , Th2 Cells/immunology , Th2 Cells/pathology
18.
J Allergy Clin Immunol ; 134(3): 583-592.e6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24636086

ABSTRACT

BACKGROUND: The fungal allergen Alternaria alternata is implicated in severe asthma and rapid onset life-threatening exacerbations of disease. However, the mechanisms that underlie this severe pathogenicity remain unclear. OBJECTIVE: We sought to investigate the mechanism whereby Alternaria was capable of initiating severe, rapid onset allergic inflammation. METHODS: IL-33 levels were quantified in wild-type and ST2(-/-) mice that lacked the IL-33 receptor given inhaled house dust mite, cat dander, or Alternaria, and the effect of inhibiting allergen-specific protease activities on IL-33 levels was assessed. An exacerbation model of allergic airway disease was established whereby mice were sensitized with house dust mite before subsequently being challenged with Alternaria (with or without serine protease activity), and inflammation, remodeling, and lung function assessed 24 hours later. RESULTS: Alternaria, but not other common aeroallergens, possessed intrinsic serine protease activity that elicited the rapid release of IL-33 into the airways of mice through a mechanism that was dependent upon the activation of protease activated receptor-2 and adenosine triphosphate signaling. The unique capacity of Alternaria to drive this early IL-33 release resulted in a greater pulmonary inflammation by 24 hours after challenge relative to the common aeroallergen house dust mite. Furthermore, this Alternaria serine protease-IL-33 axis triggered a rapid, augmented inflammation, mucus release, and loss of lung function in our exacerbation model. CONCLUSION: Alternaria-specific serine protease activity causes rapid IL-33 release, which underlies the development of a robust TH2 inflammation and exacerbation of allergic airway disease.


Subject(s)
Alternaria/immunology , Alternariosis/microbiology , Fungal Proteins/immunology , Hypersensitivity/microbiology , Serine Proteases/immunology , Adenosine Triphosphate/metabolism , Alternariosis/immunology , Animals , Antigens, Dermatophagoides/immunology , Disease Models, Animal , Disease Progression , Female , Humans , Hypersensitivity/immunology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pyroglyphidae , Receptor, PAR-2/metabolism , Receptors, Interleukin/genetics , Signal Transduction
19.
Trends Immunol ; 32(7): 328-34, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21612981

ABSTRACT

The lung is exposed to a myriad of innocuous antigens on a daily basis and must maintain a state of immune ignorance or tolerance to these harmless stimuli to retain pulmonary homeostasis and to prevent potentially fatal immunopathology. Here, we examine how, in the lower airways, resident cell populations contribute to the immune regulatory strategies that restrain inflammation. During influenza infection, these suppressive signals must be overcome to elicit a protective immune response that eliminates the virus. We also discuss how, after resolution of infection, the lung does not return to the original homeostatic state, and how the induced altered state can persist for long periods, which leaves the lung more susceptible to other infectious insults.


Subject(s)
Homeostasis , Influenza, Human/immunology , Orthomyxoviridae/immunology , Pneumonia/immunology , Animals , Humans , Influenza Vaccines/immunology , Influenza, Human/complications , Influenza, Human/virology , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/immunology , Pneumonia/etiology
SELECTION OF CITATIONS
SEARCH DETAIL