Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 112(16): 5111-6, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25820174

ABSTRACT

Silencing of interleukin-32 (IL-32) in a differentiated human promonocytic cell line impairs killing of Mycobacterium tuberculosis (MTB) but the role of IL-32 in vivo against MTB remains unknown. To study the effects of IL-32 in vivo, a transgenic mouse was generated in which the human IL-32γ gene is expressed using the surfactant protein C promoter (SPC-IL-32γTg). Wild-type and SPC-IL-32γTg mice were infected with a low-dose aerosol of a hypervirulent strain of MTB (W-Beijing HN878). At 30 and 60 d after infection, the transgenic mice had 66% and 85% fewer MTB in the lungs and 49% and 68% fewer MTB in the spleens, respectively; the transgenic mice also exhibited greater survival. Increased numbers of host-protective innate and adaptive immune cells were present in SPC-IL-32γTg mice, including tumor necrosis factor-alpha (TNFα) positive lung macrophages and dendritic cells, and IFN-gamma (IFNγ) and TNFα positive CD4(+) and CD8(+) T cells in the lungs and mediastinal lymph nodes. Alveolar macrophages from transgenic mice infected with MTB ex vivo had reduced bacterial burden and increased colocalization of green fluorescent protein-labeled MTB with lysosomes. Furthermore, mouse macrophages made to express IL-32γ but not the splice variant IL-32ß were better able to limit MTB growth than macrophages capable of producing both. The lungs of patients with tuberculosis showed increased IL-32 expression, particularly in macrophages of granulomas and airway epithelial cells but also B cells and T cells. We conclude that IL-32γ enhances host immunity to MTB.


Subject(s)
Interleukins/metabolism , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/immunology , Tuberculosis/prevention & control , Adaptive Immunity/immunology , Animals , Antigens, Ly/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Humans , Immunity, Innate/immunology , Interferon-gamma , Lung/immunology , Lung/microbiology , Lung/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Macrophages, Alveolar/immunology , Mice, Transgenic , Mutation/genetics , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Pulmonary Surfactant-Associated Protein C/metabolism , RNA Splice Sites/genetics , T-Lymphocytes, Regulatory/immunology , Transfection , Transgenes , Tuberculosis/microbiology , Tumor Necrosis Factor-alpha/metabolism , Virulence/immunology
2.
BMC Immunol ; 17(1): 27, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27501951

ABSTRACT

BACKGROUND: The lung is exposed to airborne fungal spores, and fungi that colonize the oral cavity such as Candida albicans, but does not develop disease to opportunistic fungal pathogens unless the immune system is compromised. The Group IVA cytosolic phospholipase A2 (cPLA2α) is activated in response to Candida albicans infection resulting in the release of arachidonic acid for eicosanoid production. Although eicosanoids such as prostaglandins and leukotrienes modulate inflammation and immune responses, the role of cPLA2α and eicosanoids in regulating C. albicans lung infection is not understood. METHODS: The responses of cPLA2α(+/+) and cPLA2α(-/-) Balb/c mice to intratracheal instillation of C. albicans were compared. After challenge, we evaluated weight loss, organ fungal burden, and the recruitment of cells and the levels of cytokines and eicosanoids in bronchoalveolar lavage fluid. The ability of macrophages and neutrophils from cPLA2α(+/+) and cPLA2α(-/-) mice to recognize and kill C. albicans was also compared. RESULTS: After C. albicans instillation, cPLA2α(+/+) mice recovered a modest weight loss by 48 h and completely cleared fungi from the lung by 12 h with no dissemination to the kidneys. In cPLA2α(-/-) mice, weight loss continued for 72 h, C. albicans was not completely cleared from the lung and disseminated to the kidneys. cPLA2α(-/-) mice exhibited greater signs of inflammation including higher neutrophil influx, and elevated levels of albumin and pro-inflammatory cytokines/chemokines (IL1α, IL1ß, TNFα, IL6, CSF2, CXCL1, CCL20) in bronchoalveolar lavage fluid. The amounts of cysteinyl leukotrienes, thromboxane B2 and prostaglandin E2 were significantly lower in bronchoalveolar lavage fluid from C. albicans-infected cPLA2α(-/-) mice compared to cPLA2α(+/+) mice. Alveolar macrophages and neutrophils from uninfected cPLA2α(-/-) mice exhibited less killing of C. albicans in vitro than cells from cPLA2α(+/+) mice. In addition alveolar macrophages from cPLA2α(-/-) mice isolated 6 h after instillation of GFP-C. albicans contained fewer internalized fungi than cPLA2α(+/+) macrophages. CONCLUSIONS: The results demonstrate that cPLA2α contributes to immune surveillance and host defense in the lung to prevent infection by the commensal fungus C. albicans and to dampen inflammation.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Lung Diseases, Fungal/immunology , Lung/immunology , Macrophages, Alveolar/physiology , Neutrophils/physiology , Phospholipases A2/metabolism , Animals , Arachidonic Acid/metabolism , Cell Movement , Cells, Cultured , Cytokines/metabolism , Eicosanoids/metabolism , Immunity, Innate , Inflammation Mediators/metabolism , Lung/microbiology , Macrophages, Alveolar/microbiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/microbiology , Phospholipases A2/genetics , Phospholipases A2/immunology
3.
J Immunol ; 193(5): 2238-47, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25070841

ABSTRACT

Glucocorticoid-induced TNFR family-related protein (GITR)-mediated activation of JNK was shown to regulate the suppressive activity of CD4(+)CD25(+) naturally occurring T regulatory cells (nTregs) in wild-type (WT) hosts. In this study, CD4(+)CD25(+) T cells were shown to be capable of becoming pathogenic effector cells in sensitized and challenged CD8(-/-) recipient mice. Only GITR-expressing CD4(+)CD25(+) T cells, but neither GITR knocked-in CD4(+)CD25(-) T cells nor GITR-silenced CD4(+)CD25(+) T cells, enhanced development of lung allergic responses. Inhibition of JNK in WT nTregs or nTregs from GITR(-/-)and JNK2(-/-) mice failed to enhance lung allergic responses in sensitized and challenged CD8(-/-) recipient mice. The failure to enhance responses was associated with increased bronchoalveolar lavage fluid levels of IL-10 and TGF-ß and decreased levels of IL-5, IL-6, and IL-13. In contrast, nTregs from JNK1(-/-) mice, similar to WT nTregs, were fully effective in enhancing responses. Thus, GITR stimulation of nTregs and signaling through JNK2, but not JNK1, triggered the loss of regulatory function while concomitantly gaining pathogenic CD4(+) T effector cell function responsible for exacerbating asthma-like immunopathology.


Subject(s)
Asthma/immunology , Lung/immunology , MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinase 9/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Asthma/genetics , Asthma/pathology , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Lung/pathology , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/immunology , Mitogen-Activated Protein Kinase 9/genetics , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
4.
Respir Res ; 16: 117, 2015 Sep 26.
Article in English | MEDLINE | ID: mdl-26410792

ABSTRACT

BACKGROUND: Ozone pollution has adverse effects on respiratory health in children and adults. This study was carried out in the mouse model to investigate the influence of age and to define the role of toll-like receptor four (TLR4) in the lung response to ozone exposure during postnatal development. METHODS: Female mice (1 to 6 weeks of age) were exposed for 3 h to ozone (1 part per million) or filtered air. Analyses were carried out at six and 24 h after completion of exposure, to assess the effects on lung permeability, airway neutrophilia, expression of antioxidants and chemokines, and mucus production. The role of TLR4 was defined by examining TLR4 expression in the lung during development, and by investigating the response to ozone in tlr4-deficient mice. RESULTS: Metallothionein-1, calcitonin gene-related product, and chemokine C-X-C ligand (CXCL) five were consistent markers induced by ozone throughout development. Compared with adults, neonates expressed lower levels of pulmonary TLR4 and responded with increased mucus production, and developed an attenuated response to ozone characterized by reduced albumin leakage and neutrophil influx into the airways, and lower expression of CXCL1 and CXCL2 chemokines. Examination of the responses in tlr4-deficient mice indicated that ozone-mediated airway neutrophilia, but not albumin leakage or mucus production were dependent on TLR4. CONCLUSIONS: Collectively, the data demonstrate that the response to ozone is determined by age and is partially dependent on TLR4 signaling. The reduced responsiveness of the neonatal lung to ozone may be due at least in part to insufficient pulmonary TLR4 expression.


Subject(s)
Air Pollutants/toxicity , Lung/drug effects , Ozone/toxicity , Toll-Like Receptor 4/drug effects , Age Factors , Animals , Antioxidants/metabolism , Biomarkers/metabolism , Capillary Permeability/drug effects , Chemokines/metabolism , Female , Inhalation Exposure , Lung/blood supply , Lung/metabolism , Mice, Inbred BALB C , Mice, Knockout , Mucus/metabolism , Neuropeptides/metabolism , Neutrophil Infiltration/drug effects , Serum Albumin/metabolism , Signal Transduction/drug effects , Time Factors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
5.
Am J Respir Cell Mol Biol ; 51(1): 143-54, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24521403

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract illnesses in infants worldwide. Both RSV-G and RSV-F glycoproteins play pathogenic roles during infection with RSV. The objective of this study was to compare the effects of anti-RSV-G and anti-RSV-F monoclonal antibodies (mAbs) on airway hyperresponsiveness (AHR) and inflammation after primary or secondary RSV infection in mice. In the primary infection model, mice were infected with RSV at 6 weeks of age. Anti-RSV-G or anti-RSV-F mAbs were administered 24 hours before infection or Day +2 postinfection. In a secondary infection model, mice were infected (primary) with RSV at 1 week (neonate) and reinfected (secondary) 5 weeks later. Anti-RSV-G and anti-RSV-F mAbs were administered 24 hours before the primary infection. Both mAbs had comparable effects in preventing airway responses after primary RSV infection. When given 2 days after infection, anti-RSV-G-treated mice showed significantly decreased AHR and airway inflammation, which persisted in anti-RSV-F-treated mice. In the reinfection model, anti-RSV-G but not anti-RSV-F administered during primary RSV infection in neonates resulted in decreased AHR, eosinophilia, and IL-13 but increased levels of IFN-γ in bronchoalveolar lavage on reinfection. These results support the use of anti-RSV-G in the prevention and treatment of RSV-induced disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bronchiolitis, Viral/prevention & control , Inflammation/prevention & control , Respiratory Hypersensitivity/prevention & control , Respiratory Syncytial Virus Infections/prevention & control , Viral Fusion Proteins/immunology , Animals , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Bronchiolitis, Viral/etiology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Inflammation/etiology , Mice , Mice, Inbred BALB C , Respiratory Hypersensitivity/etiology , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity
6.
Ann Allergy Asthma Immunol ; 112(3): 211-216.e1, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24428972

ABSTRACT

BACKGROUND: Numbers of CD8(+) T cells expressing the leukotriene B4 (LTB4) receptor, BLT1, have been correlated with asthma severity. OBJECTIVE: To examine the activation and numbers of BLT1-expressing peripheral blood CD4(+) and CD8(+) T cells from patients with steroid-sensitive (SS) and steroid-resistant (SR) asthma. METHODS: CD4(+) and CD8(+) T cells isolated from peripheral blood of healthy human subjects and patients with SS and SR asthma were stimulated in culture with anti-CD3/anti-CD28 followed by analysis of BLT1 surface expression and cytokine production. Activation of CD8(+) T cells after ligation of BLT1 by LTB4 was monitored by changes in intracellular Ca(2+) concentrations. RESULTS: The number of BLT1-expressing cells was larger in patients with asthma than in controls and larger on activated CD8(+) than on CD4(+) T cells. Addition of LTB4 to activated CD8(+) T cells resulted in increases in intracellular Ca(2+) concentrations. Expansion of activated CD4(+) T cells, unlike CD8(+) T cells, was significantly decreased in the presence of corticosteroid. In patients with SS asthma, numbers of BLT1-expressing CD8(+) T cells were lower in the presence of corticosteroid, unlike in those with SR asthma in whom cell expansion was maintained. Levels of interleukin-13 were highest in cultured CD8(+) T cells, whereas interleukin-10 levels were higher in CD4(+) T cells from controls and patients with SS asthma. Interferon-γ levels were lowest in patients with SR asthma. CONCLUSION: Differences in BLT1 expression, steroid sensitivity, and cytokine production were demonstrated in T lymphocytes from patients with SS and SR asthma. The LTB4-BLT1 pathway in CD8(+) cells may play an important role in asthma and serve as an important target in the treatment of patients with SR asthma.


Subject(s)
Asthma/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dexamethasone/pharmacology , Receptors, Leukotriene B4/metabolism , Adrenal Cortex Hormones/pharmacology , Adult , Antibodies, Monoclonal/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , Calcium/metabolism , Cells, Cultured , Culture Media, Conditioned/chemistry , Female , Humans , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-13/metabolism , Lymphocyte Activation/immunology , Male
7.
J Biol Chem ; 287(21): 17100-17108, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22461627

ABSTRACT

Naturally occurring Foxp3(+)CD4(+)CD25(+) T regulatory cell (nTreg)-mediated suppression of lung allergic responses is abrogated following ligation of glucocorticoid-induced tumor necrosis receptor (GITR) family-related protein. In vitro stimulation of nTregs with GITR ligand increased phosphorylation of c-Jun N-terminal kinase (JNK) but not extracellular signal-regulated protein kinase (ERK) or p38 MAPK. SP600125, a known JNK inhibitor, prevented GITR-mediated phosphorylation of JNK. Activation of JNK was associated with increases in the upstream mitogen-activated protein kinase kinase 7 (MKK7) and the downstream transcription factor NF-κß. Phosphorylated c-Jun (p-c-Jun), indicative of the activation of JNK, was detected in the immunoprecipitates of nTregs from wild-type but not JNK- or GITR-deficient mice. Treatment with an inhibitor of JNK phosphorylation resulted in complete reversal of all GITR-induced changes in nTreg phenotype and function, with full restoration of suppression of in vivo lung allergic responses and in vitro proliferation of activated CD4(+)CD25(-) T cells. Thus, regulation of JNK phosphorylation plays a central role in T regulatory cell function with therapeutic implications for the treatment of asthma and autoimmune diseases.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/metabolism , Immune Tolerance/physiology , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/physiology , T-Lymphocytes, Regulatory/metabolism , Animals , Anthracenes/pharmacology , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Activation/immunology , Female , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/immunology , Immune Tolerance/drug effects , Lung/immunology , Lung/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase 7/immunology , MAP Kinase Kinase 7/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes, Regulatory/immunology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Immunol ; 186(1): 113-20, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21115736

ABSTRACT

Naturally occurring CD4(+)CD25(+)Foxp3(+) T regulatory cells (nTregs) regulate lung allergic responses through production of IL-10 and TGF-ß. nTregs from CD8(-/-) mice failed to suppress lung allergic responses and were characterized by reduced levels of Foxp3, IL-10, and TGF-ß, and high levels of IL-6. Administration of anti-IL-6 or anti-IL-6R to wild-type recipients prior to transfer of CD8(-/-) nTregs restored suppression. nTregs from IL-6(-/-) mice were suppressive, but lost this capability if incubated with IL-6 prior to transfer. The importance of CD8 in regulating the production of IL-6 in nTregs was demonstrated by the loss of suppression and increases in IL-6 following transfer of nTregs from wild-type donors depleted of CD8(+) cells. Transfer of nTregs from CD8(-/-) donors reconstituted with CD8(+) T cells was suppressive, and accordingly, IL-6 levels were reduced. These data identify the critical role of CD8-T regulatory cell interactions in regulating the suppressive phenotype of nTregs through control of IL-6 production.


Subject(s)
CD8 Antigens/physiology , Immunophenotyping , Immunosuppression Therapy , Interleukin-6/biosynthesis , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , CD8 Antigens/genetics , Cells, Cultured , Female , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Interleukin-10/antagonists & inhibitors , Interleukin-6/deficiency , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/physiology , Respiratory Hypersensitivity/genetics , Transforming Growth Factor beta/antagonists & inhibitors
9.
Proc Natl Acad Sci U S A ; 107(1): 320-5, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20080799

ABSTRACT

Respiratory syncytial virus (RSV) is the most common cause of hospitalization for respiratory tract infection in young children. It is also a significant cause of morbidity and mortality in elderly individuals and in persons with asthma and chronic obstructive pulmonary disease. Currently, no reliable vaccine or simple RSV antiviral therapy is available. Recently, we determined that the minor pulmonary surfactant phospholipid, palmitoyl-oleoyl-phosphatidylglycerol (POPG), could markedly attenuate inflammatory responses induced by lipopolysaccharide through direct interactions with the Toll-like receptor 4 (TLR4) interacting proteins CD14 and MD-2. CD14 and TLR4 have been implicated in the host response to RSV. Treatment of bronchial epithelial cells with POPG significantly inhibited interleukin-6 and -8 production, as well as the cytopathic effects induced by RSV. The phospholipid bound RSV with high affinity and inhibited viral attachment to HEp2 cells. POPG blocked viral plaque formation in vitro by 4 log units, and markedly suppressed the expansion of plaques from cells preinfected with the virus. Administration of POPG to mice, concomitant with viral infection, almost completely eliminated the recovery of virus from the lungs at 3 and 5 days after infection, and abrogated IFN-gamma (IFN-gamma) production and the enhanced expression of surfactant protein D (SP-D). These findings demonstrate an important approach to prevention and treatment of RSV infections using exogenous administration of a specific surfactant phospholipid.


Subject(s)
Inflammation/immunology , Phosphatidylglycerols/pharmacology , Pulmonary Surfactants/pharmacology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Animals , Cell Death/physiology , Cells, Cultured , Child , Cytokines/immunology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Female , Humans , Inflammation/drug therapy , Inflammation/virology , Lipopolysaccharide Receptors/immunology , Mice , Mice, Inbred BALB C , Phosphatidylglycerols/therapeutic use , Pulmonary Surfactants/therapeutic use , Respiratory Mucosa/cytology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Toll-Like Receptor 4/immunology
10.
J Allergy Clin Immunol ; 130(5): 1175-1186.e9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23036746

ABSTRACT

BACKGROUND: Recent studies revealed a critical role for thymic stromal lymphopoietin (TSLP) released from epithelial cells and OX40 ligand (OX40L) expressed on dendritic cells (DCs) in T(H)2 priming and polarization. OBJECTIVES: We sought to determine the importance of the TSLP-OX40L axis in neonatal respiratory syncytial virus (RSV) infection. METHODS: Mice were initially infected with RSV as neonates or adults and reinfected 5 weeks later. Anti-OX40L or anti-TSLP were administered during primary or secondary infection. Outcomes included assessment of airway function and inflammation and expression of OX40L, TSLP, and IL-12. RESULTS: OX40L was expressed mainly on CD11c(+)MHC class II (MHCII)(+)CD11b(+) DCs but not CD103(+) DCs. Treatment of neonates with OX40L antibody during primary RSV infection prevented the subsequent enhancement of airway hyperresponsiveness and the development of airway eosinophilia and mucus hyperproduction on reinfection. Administration of anti-TSLP before neonatal RSV infection reduced the accumulation of lung DCs, decreased OX40L expression on lung DCs, and attenuated the enhancement of airway responses after reinfection. CONCLUSIONS: In mice initially infected as neonates, TSLP expression induced by RSV infection is an important upstream event that controls OX40L expression, lung DC migration, and T(H)2 polarization, accounting for the enhanced response on reinfection.


Subject(s)
Bronchial Hyperreactivity/immunology , Cytokines/metabolism , Dendritic Cells/immunology , OX40 Ligand/metabolism , Pulmonary Eosinophilia/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Animals , Animals, Newborn , Antibodies, Blocking/administration & dosage , Bronchial Hyperreactivity/etiology , Cell Movement/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/drug effects , Humans , Immunity/drug effects , Interleukin-12/metabolism , Mice , Mice, Inbred BALB C , Mucus/metabolism , OX40 Ligand/genetics , OX40 Ligand/immunology , Pulmonary Eosinophilia/etiology , Respiratory Syncytial Virus Infections/complications , Thymic Stromal Lymphopoietin
11.
J Immunol ; 185(1): 253-62, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20525882

ABSTRACT

Invariant NKT cells (iNKT cells) play a pivotal role in the development of allergen-induced airway hyperresponsiveness (AHR) and inflammation. However, it is unclear what role they play in the initiation (sensitization) phase as opposed to the effector (challenge) phase. The role of iNKT cells during sensitization was examined by determining the response of mice to intratracheal transfer of OVA-pulsed or OVA-alpha-galactosylceramide (OVA/alphaGalCer)-pulsed bone marrow-derived dendritic cells (BMDCs) prior to allergen challenge. Wild-type (WT) recipients of OVA-BMDCs developed AHR, increased airway eosinophilia, and increased levels of Th2 cytokines in bronchoalveolar lavage fluid, whereas recipients of OVA/alphaGalCer BMDCs failed to do so. In contrast, transfer of these same OVA/alphaGalCer BMDCs into IFN-gamma-deficient (IFN-gamma(-/-)) mice enhanced the development of these lung allergic responses, which was reversed by exogenous IFN-gamma treatment following OVA-BMDC transfer. Further, Jalpha18-deficient recipients, which lack iNKT cells, developed the full spectrum of lung allergic responses following reconstitution with highly purified WT liver or spleen iNKT cells and transfer of OVA-BMDCs, whereas reconstituted recipients of OVA/alphaGalCer BMDCs failed to do so. Transfer of iNKT cells from IFN-gamma(-/-) mice restored the development of these responses in Jalpha18-deficient recipients following OVA-BMDC transfer; the responses were enhanced following OVA/alphaGalCer BMDC transfer. iNKT cells from these IFN-gamma(-/-) mice produced higher levels of IL-13 in vitro compared with WT iNKT cells. These data identify IFN-gamma as playing a critical role in dictating the consequences of iNKT cell activation in the initiation phase of the development of AHR and airway inflammation.


Subject(s)
Allergens/administration & dosage , Interferon-gamma/biosynthesis , Natural Killer T-Cells/immunology , Ovalbumin/immunology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/prevention & control , Adoptive Transfer , Allergens/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/prevention & control , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Female , Galactosylceramides/administration & dosage , Galactosylceramides/immunology , Galactosylceramides/metabolism , Inflammation Mediators/administration & dosage , Inflammation Mediators/metabolism , Interferon-gamma/deficiency , Interferon-gamma/physiology , Intubation, Intratracheal , Ligands , Liver/immunology , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Ovalbumin/administration & dosage , Respiratory Hypersensitivity/pathology
12.
J Immunol ; 183(3): 1821-7, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19592660

ABSTRACT

Naturally occurring Foxp3(+)CD4(+)CD25(+) T cells isolated from lungs of naive mice regulate lung allergic airway hyperresponsiveness, inflammation, levels of Th2 cytokines, and mucus production. OVA-specific (alphabetaTCR(+)) CD4(+)CD25(+) T cells suppressed ragweed-induced airway hyperresponsiveness and inflammation as did anti-TCR-treated OVA-specific CD4(+)CD25(+) T cells, suggesting that Ag-specificity was not required for expression of regulatory activities. Suppression was associated with increased levels of IL-10 and TGF-beta; decreased levels of IL-4, IL-5, and IL-13 in bronchoalveolar lavage fluid; and reduced recruitment and activation of CD8(+) T cells in the airways. Following intratracheal administration, OVA-specific CD4(+)CD25(+) T cells were identified in both the airway lumens and lung parenchyma, and in some instances in close proximity to host CD8(+) T cells. These results demonstrate that the regulatory activities of naturally occurring Foxp3(+)CD4(+)CD25(+) T cells on lung allergic responses are Ag-nonspecific and thus, independent of Ag-specific recognition.


Subject(s)
Epitopes/immunology , Hypersensitivity/immunology , Lung Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Ambrosia/immunology , Animals , CD8-Positive T-Lymphocytes , Cytokines/analysis , Inflammation , Mice , Mice, Inbred Strains , Ovalbumin/immunology
13.
J Immunol ; 183(1): 181-90, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542429

ABSTRACT

Suppressing the abnormalities associated with asthma has been difficult to accomplish using immunotherapy or vaccination once the disease is established. The effector cells necessary for effective immunization/vaccination and immunotherapy of asthma are also not well understood. Therefore, we vaccinated allergen (OVA)-sensitized mice to determine whether therapeutic immunization could suppress airway hyperresponsiveness (AHR) and inflammation and to identify key immune effector cells and cytokines. Mice were immunized with a vaccine comprised of Ag and cationic liposome-DNA complexes (CLDC), a vaccine which has previously been shown to elicit strong CD4(+) and CD8(+) T cell responses and activation of Th1 immunity. We showed that immunization with the OVA-CLDC vaccine significantly suppressed AHR, eosinophilia, goblet cell metaplasia, and Th2 cytokine production. In contrast, immunization with CLDC alone suppressed eosinophilia and Th2 cytokine production, but failed to suppress AHR and goblet cell changes. Using adoptive transfer experiments, we found that suppression of AHR was mediated by Ag-specific CD8(+) T cells and was dependent on IFN-gamma production by the transferred T cells. Thus, we conclude that generation of strong, allergen-specific CD8(+) T cell responses by immunization may be capable of suppressing AHR and allergic airway inflammation, even in previously sensitized and challenged mice.


Subject(s)
Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/prevention & control , CD8-Positive T-Lymphocytes/immunology , Immunosuppressive Agents/administration & dosage , Inflammation Mediators/administration & dosage , Inflammation Mediators/immunology , Vaccines, DNA/immunology , Adoptive Transfer , Animals , Asthma/immunology , Asthma/pathology , Asthma/prevention & control , Bronchial Hyperreactivity/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cations/administration & dosage , Cations/metabolism , Cytokines/physiology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , H-2 Antigens/administration & dosage , H-2 Antigens/genetics , H-2 Antigens/immunology , Humans , Immunosuppressive Agents/immunology , Liposomes , Methacholine Chloride/administration & dosage , Methacholine Chloride/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , Ovalbumin/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
14.
Nat Med ; 10(8): 865-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15258576

ABSTRACT

Allergic asthma is a complex syndrome characterized by airway obstruction, airway inflammation and airway hyper-responsiveness (AHR). Using a mouse model of allergen-induced AHR, we previously demonstrated that CD8-deficient mice develop significantly lower AHR, eosinophilic inflammation and interleukin (IL)-13 levels in bronchoalveolar lavage fluid compared with wild-type mice. These responses were restored by adoptive transfer of antigen-primed CD8(+) T cells. Previously, two distinct populations of antigen-experienced CD8(+) T cells, termed effector (T(EFF)) and central memory (T(CM)) cells, have been described. After adoptive transfer into CD8-deficient mice, T(EFF), but not T(CM), cells restored AHR, eosinophilic inflammation and IL-13 levels. T(EFF), but not T(CM), cells accumulated in the lungs, and intracellular cytokine staining showed that the transferred T(EFF) cells were a source of IL-13. These data suggest an important role for effector CD8(+) T cells in the development of AHR and airway inflammation, which may be associated with their Tc2-type cytokine production and their capacity to migrate into the lung.


Subject(s)
Allergens/immunology , Bronchial Hyperreactivity/immunology , Bronchitis/immunology , CD8-Positive T-Lymphocytes/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Alum Compounds , Analysis of Variance , Animals , Bronchial Hyperreactivity/pathology , Bronchitis/physiopathology , Bronchoalveolar Lavage Fluid/immunology , CD8 Antigens/immunology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukin-13/immunology , Lung/immunology , Lung/pathology , Methacholine Chloride , Mice , Mice, Mutant Strains , Ovalbumin
15.
Am J Respir Crit Care Med ; 182(4): 455-63, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20442434

ABSTRACT

RATIONALE: Respiratory syncytial virus (RSV) bronchiolitis in infants may be followed by the development of asthma-like symptoms. Age at first infection dictates consequences upon reinfection. Reinfection of mice initially exposed as neonates to RSV enhanced development of airway hyperresponsiveness (AHR), eosinophilic inflammation, and mucus hyperproduction. RSV lower respiratory tract disease is associated with activation of the leukotriene pathway. OBJECTIVES: To determine the effects of montelukast (MK), a cysteinyl leukotriene (cysLT) receptor antagonist, in primary and secondary RSV-infected newborn and adult mice. METHODS: BALB/c mice were infected with RSV at 1 week (neonate) or 6 to 8 weeks (adult) of age and reinfected 5 weeks later. MK was administered 1 day before the initial infection and through Day 6 after infection. Seven days after primary or secondary infection, airway function was assessed by lung resistance to increasing doses of inhaled methacholine; lung inflammation, goblet cell metaplasia, and cytokine levels in bronchoalveolar lavage fluid were monitored. MEASUREMENTS AND MAIN RESULTS: RSV infection induced cysLT release in bronchoalveolar lavage fluid. MK decreased RSV-induced AHR, airway inflammation, and increased IFN-gamma production in primary infected adult and neonatal mice. MK, administered during initial infection of neonates but not during secondary infection, prevented subsequent enhancement of AHR, airway eosinophilia, and mucus hyperproduction upon reinfection. CONCLUSIONS: MK attenuated the initial responses to primary RSV infection in both age groups and altered the consequences of RSV reinfection in mice initially infected as neonates. These data support an important role for cysLT in RSV-induced AHR and inflammation.


Subject(s)
Acetates/pharmacology , Anti-Asthmatic Agents/pharmacology , Inflammation/prevention & control , Quinolines/pharmacology , Respiratory Hypersensitivity/prevention & control , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Viruses/drug effects , Animals , Animals, Newborn , Bronchiolitis, Viral/complications , Bronchiolitis, Viral/prevention & control , Bronchiolitis, Viral/virology , Bronchoalveolar Lavage Fluid/virology , Cyclopropanes , Cysteine/antagonists & inhibitors , Disease Models, Animal , Inflammation/complications , Interferon-gamma/drug effects , Leukotriene Antagonists/pharmacology , Leukotrienes , Mice , Mice, Inbred BALB C , Recurrence , Respiratory Hypersensitivity/etiology , Sulfides
16.
J Allergy Clin Immunol ; 126(2): 306-16, 316.e1-12, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20624645

ABSTRACT

BACKGROUND: Although implicated in the disease, the specific contributions of FcepsilonRI and IL-13 to the pathogenesis of peanut-induced intestinal allergy are not well defined. OBJECTIVES: We sought to determine the contributions of FcepsilonRI, IL-13, and mast cells to the development of intestinal mucosal responses in a murine model of peanut-induced intestinal allergy. METHODS: Sensitized wild-type (WT), FcepsilonRI-deficient (FcepsilonRI(-/-)), and mast cell-deficient (Kit(W-sh/W-sh)) mice received peanut orally every day for 1 week. Bone marrow-derived mast cells (BMMCs) from WT, FcepsilonRI(-/-), IL-4(-/-), IL-13(-/-), and IL-4/IL-13(-/-) mice were differentiated and transferred into WT, FcepsilonRI(-/-), and Kit(W-sh/W-sh) recipients. BMMCs from WT and UBI-GFP/BL6 mice were differentiated and transferred into WT and Kit(W-sh/W-sh) mice. Blockade of IL-13 was achieved by using IL-13 receptor alpha2 (IL-13Ralpha2)-IgG fusion protein. RESULTS: FcepsilonRI(-/-) mice showed decreased intestinal inflammation (mast cell and eosinophil numbers) and goblet cell metaplasia and reduced levels of IL4, IL6, IL13, and IL17A mRNA expression in the jejunum. Transfer of WT BMMCs to FcepsilonRI(-/-) recipients restored their ability to develop intestinal allergic responses unlike transfer of FcepsilonRI(-/-), IL-13(-/-), or IL-4/IL-13(-/-) BMMCs. FcepsilonRI(-/-) mice exhibited lower IL-13 levels and treatment of WT mice with IL-13 receptor alpha2 prevented peanut-induced intestinal allergy and inflammation. CONCLUSIONS: These data indicate that the development of peanut-induced intestinal allergy is mediated through a mast cell-dependent IgE-FcepsilonRI-IL-13 pathway. Targeting IL-13 might be a potential treatment for IgE-mediated peanut-induced allergic responses in the intestine.


Subject(s)
Arachis/adverse effects , Immunoglobulin E/immunology , Interleukin-13/immunology , Mast Cells/immunology , Peanut Hypersensitivity/immunology , Receptors, IgE/immunology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Immunity, Mucosal/genetics , Immunity, Mucosal/immunology , Immunoglobulin E/genetics , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/pharmacology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-13/antagonists & inhibitors , Interleukin-13/genetics , Interleukin-13 Receptor alpha2 Subunit/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Intestines/immunology , Intestines/pathology , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Peanut Hypersensitivity/drug therapy , Peanut Hypersensitivity/genetics , Peanut Hypersensitivity/pathology , Receptors, IgE/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology
17.
J Immunol ; 181(2): 1170-8, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606670

ABSTRACT

Dendritic cells (DC) are important APCs that control allergen-induced airway responses by interacting directly with T cells. Leukotriene B(4) (LTB(4)), interacting with its high-affinity receptor, LTB(4) receptor 1 (BLT1), is known to attract and activate leukocytes during inflammation. We have previously shown that BLT1 expression on Ag-primed T cells is required for the development of airway hyperresponsiveness (AHR; Miyahara et al. 2005. Am. J. Respir. Crit. Care Med. 172: 161-167). However, the role for the LTB(4)-BLT1 pathway in DC function in allergen-induced airway responses has not been defined. Bone marrow-derived DCs (BMDC) were generated. Naive BALB/c mice received OVA-pulsed BLT1-deficient (BLT1(-/-)) BMDCs or wild-type BMDCs intratracheally and were then challenged with OVA for 3 days. Airway responses were monitored 48 h after the last allergen challenge. BLT1(-/-) BMDCs showed normal maturation judged from surface expression of CD markers. Compared with recipients of wild-type BMDCs, mice that received BLT1(-/-) BMDCs developed significantly lower AHR to inhaled methacholine, lower goblet cell metaplasia, and eosinophilic infiltration in the airways and decreased levels of Th2 type cytokines in the bronchoalveolar lavage fluid. Migration of BLT1(-/-) BMDCs into peribronchial lymph nodes was significantly impaired compared with BLT1(+/+) BMDCs after intratracheal instillation. These data suggest that BLT1 expression on DCs is required for migration of DCs to regional lymph nodes as well as in the development of AHR and airway inflammation.


Subject(s)
Bronchial Hyperreactivity/immunology , Dendritic Cells/immunology , Lung/immunology , Receptors, Leukotriene B4/metabolism , Th2 Cells/immunology , Allergens/immunology , Animals , Bronchial Hyperreactivity/metabolism , Bronchoalveolar Lavage Fluid/immunology , Bronchoconstrictor Agents/administration & dosage , Cytokines/analysis , Cytokines/immunology , Dendritic Cells/metabolism , Female , Leukotriene A4/immunology , Leukotriene A4/metabolism , Lymph Nodes/immunology , Methacholine Chloride/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/immunology , Receptors, Leukotriene B4/immunology , Th2 Cells/metabolism
18.
J Allergy Clin Immunol ; 123(1): 138-145.e5, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19056111

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV)-specific IgE is a component of the host response to RSV infection, but its role in the subsequent enhancement of altered airway responsiveness is unknown. OBJECTIVE: To define the role of RSV-specific IgE in the enhancement of airway responsiveness on reinfection of newborn mice. METHODS: Mice were infected as newborns with RSV and were reinfected 5 weeks later. The role of IgE was determined by documenting RSV-specific IgE response after neonatal infection, and by assessing airway responsiveness on reinfection. RESULTS: After neonatal infection, wild-type (WT) mice developed an RSV-specific IgE response. On reinfection, these mice developed enhanced airway hyperresponsiveness (AHR), airway eosinophilia, and mucus hyperproduction, and their T-cell cytokine response was skewed toward a T(H)2 phenotype. None of these altered responses developed on reinfection of IL-4(-/-)/IL-13(-/-) mice, and no RSV-specific IgE could be detected after neonatal infection of these mice. Fc epsilon RI(-/-) mice did not develop the enhanced AHR on reinfection, and airway eosinophilia and mucus production were significantly attenuated. These responses could be restored in deficient mice reconstituted with WT mast cells. In RSV-infected newborn WT mice, administration of anti-IgE prevented the enhancement of AHR and attenuated eosinophilia and mucus hyperproduction on reinfection, an effect that was associated with diminished T(H)2 cytokine production and increased IFN-gamma production. CONCLUSION: Respiratory syncytial virus-specific IgE enhances the development of T(H)2-biased airway responsiveness on reinfection of mice initially infected as newborns.


Subject(s)
Antibodies, Viral/immunology , Immunoglobulin E/immunology , Respiratory Hypersensitivity/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Th2 Cells/immunology , Animals , Animals, Newborn , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, IgE/genetics , Receptors, IgE/immunology , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/virology , Respiratory Syncytial Virus Infections/genetics
19.
J Allergy Clin Immunol ; 123(1): 249-57, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19130938

ABSTRACT

BACKGROUND: Ligation of the leukotriene B(4) (LTB(4)) receptor 1 on effector memory CD8(+) T cells by LTB(4) is important for the recruitment of CD8(+) T cells into the airways, which appears central to the induction of airway hyperresponsiveness (AHR) and allergic inflammation. Phosphorylation of extracellular signal-regulated kinase (ERK) is important in activation and cytokine production from many cell types. OBJECTIVE: The roles of ERKs in effector CD8(+) T-cell function and on CD8(+) T cell-mediated AHR were determined. METHODS: Effector CD8(+) T cells were generated from OVA(257-264) (SIINFEKL) peptide-primed mononuclear cells from OT-1 mice. The effects of U0126, an ERK inhibitor, on effector CD8(+) T-cell function and on CD8(+) T cell-mediated AHR and allergic inflammation were examined. RESULTS: Pretreatment of effector CD8(+) T cells with U0126 suppressed anti-CD3/anti-CD28-induced ERK1/2 phosphorylation and cytokine production, but did not affect LTB(4)-induced Ca(2+) mobilization or chemotaxis. Adoptive transfer of U0126-treated CD8(+) T cells into sensitized mice before secondary allergen challenge resulted in significant decreases in AHR, eosinophilic inflammation, goblet cell metaplasia, and IL-5 and IL-13 levels in bronchoalveolar lavage fluid of recipient mice. The number of transferred CD8(+) T cells accumulating in bronchoalveolar lavage fluid or lungs was unaffected by treatment. CONCLUSION: ERK1/2-dependent pathways are essential for the effector functions of CD8(+) T cells, including T(H)2 cytokine production, allergic inflammation, and development of AHR. Inhibition of ERK1/2 signaling has potential therapeutic benefit in preventing CD8(+) T cell-mediated AHR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemotaxis/immunology , MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/immunology , Respiratory Hypersensitivity/immunology , Adoptive Transfer , Allergens/immunology , Allergens/pharmacology , Animals , Bronchoalveolar Lavage Fluid/immunology , Butadienes/pharmacology , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/pathology , Calcium/immunology , Calcium/metabolism , Chemotaxis/drug effects , Enzyme Inhibitors/pharmacology , Eosinophils/enzymology , Eosinophils/immunology , Eosinophils/pathology , Goblet Cells/enzymology , Goblet Cells/immunology , Goblet Cells/pathology , Immunologic Memory/drug effects , Immunologic Memory/immunology , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Leukotriene B4/immunology , Leukotriene B4/metabolism , Leukotriene B4/pharmacology , Lung/enzymology , Lung/immunology , Lung/pathology , MAP Kinase Signaling System/drug effects , Metaplasia/enzymology , Metaplasia/immunology , Metaplasia/pathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Receptors, Leukotriene B4/immunology , Receptors, Leukotriene B4/metabolism , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/pathology , Th2 Cells/enzymology , Th2 Cells/immunology , Th2 Cells/pathology
20.
J Innate Immun ; 12(1): 103-115, 2020.
Article in English | MEDLINE | ID: mdl-30928973

ABSTRACT

The negative immune regulator Tollip inhibits the proinflammatory response to rhinovirus (RV) infection, a contributor to airway neutrophilic inflammation and asthma exacerbations, but the underlying molecular mechanisms are poorly understood. Tollip may inhibit IRAK1, a signaling molecule downstream of ST2, the receptor of IL-33. This study was carried out to determine whether Tollip downregulates ST2 signaling via inhibition of IRAK1, but promotes soluble ST2 (sST2) production, thereby limiting excessive IL-8 production in human airway epithelial cells during RV infection in a type 2 cytokine milieu (e.g., IL-13 and IL-33 stimulation). Tollip- and IRAK1-deficient primary human tracheobronchial epithelial (HTBE) cells and Tollip knockout (KO) HTBE cells were generated using the shRNA knockdown and CRISPR/Cas9 approaches, respectively. Cells were stimulated with IL-13, IL-33, and/or RV16. sST2, activated IRAK1, and IL-8 were measured. A Tollip KO mouse model was utilized to test if Tollip regulates the airway inflammatory response to RV infection in vivo under IL-13 and IL-33 treatment. Following IL-13, IL-33, and RV treatment, Tollip-deficient (vs. -sufficient) HTBE cells produced excessive IL-8, accompanied by decreased sST2 production but increased IRAK1 activation. IL-8 production following IL-13/IL-33/RV exposure was markedly attenuated in IRAK1-deficient HTBE cells, as well as in Tollip KO HTBE cells treated with an IRAK1 inhibitor or a recombinant sST2 protein. Tollip KO (vs. wild-type) mice developed exaggerated airway neutrophilic responses to RV in the context of IL-13 and IL-33 treatment. Collectively, these data demonstrate that Tollip restricts excessive IL-8 production in type 2 cytokine-exposed human airways during RV infection by promoting sST2 production and inhibiting IRAK1 activation. sST2 and IRAK1 may be therapeutic targets for attenuating excessive neutrophilic airway inflammation in asthma, especially during RV infection.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neutrophils/immunology , Picornaviridae Infections/metabolism , Respiratory Mucosa/metabolism , Rhinovirus/physiology , Th2 Cells/immunology , Cells, Cultured , Cytokines/metabolism , Humans , Immunomodulation , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Neutrophil Activation , Picornaviridae Infections/immunology , RNA, Small Interfering/genetics , Respiratory Mucosa/pathology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL