Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Eur Respir Rev ; 33(172)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38657996

ABSTRACT

Common airborne allergens (pollen, animal dander and those from fungi and insects) are the main triggers of type I allergic disorder in the respiratory system and are associated with allergic rhinitis, allergic asthma, as well as immunoglobulin E (IgE)-mediated allergic bronchopulmonary aspergillosis. These allergens promote IgE crosslinking, vasodilation, infiltration of inflammatory cells, mucosal barrier dysfunction, extracellular matrix deposition and smooth muscle spasm, which collectively cause remodelling of the airways. Fungus and insect (house dust mite and cockroaches) indoor allergens are particularly rich in proteases. Indeed, more than 40 different types of aeroallergen proteases, which have both IgE-neutralising and tissue-destructive activities, have been documented in the Allergen Nomenclature database. Of all the inhaled protease allergens, 85% are classed as serine protease activities and include trypsin-like, chymotrypsin-like and collagenolytic serine proteases. In this article, we review and compare the allergenicity and proteolytic effect of allergen serine proteases as listed in the Allergen Nomenclature and MEROPS databases and highlight their contribution to allergic sensitisation, disruption of the epithelial barrier and activation of innate immunity in allergic airways disease. The utility of small-molecule inhibitors of allergen serine proteases as a potential treatment strategy for allergic airways disease will also be discussed.


Subject(s)
Allergens , Immunity, Innate , Serine Proteases , Humans , Allergens/immunology , Serine Proteases/metabolism , Serine Proteases/immunology , Animals , Air Pollution, Indoor/adverse effects , Serine Proteinase Inhibitors/therapeutic use , Inhalation Exposure/adverse effects , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/enzymology
2.
Am J Respir Cell Mol Biol ; 62(5): 657-667, 2020 05.
Article in English | MEDLINE | ID: mdl-31922900

ABSTRACT

Cystic fibrosis (CF) is a lethal genetic disease characterized by progressive lung damage and airway obstruction. The majority of patients demonstrate airway hyperresponsiveness (AHR), which is associated with more rapid lung function decline. Recent studies in the neonatal CF pig demonstrated airway smooth muscle (ASM) dysfunction. These findings, combined with observed CF transmembrane conductance regulator (CFTR) expression in ASM, suggest that a fundamental defect in ASM function contributes to lung function decline in CF. One established driver of AHR and ASM dysfunction is transforming growth factor (TGF) ß1, a genetic modifier of CF lung disease. Prior studies demonstrated that TGFß exposure in CF mice drives features of CF lung disease, including goblet cell hyperplasia and abnormal lung mechanics. CF mice displayed aberrant responses to pulmonary TGFß, with elevated PI3K signaling and greater increases in lung resistance compared with controls. Here, we show that TGFß drives abnormalities in CF ASM structure and function through PI3K signaling that is enhanced in CFTR-deficient lungs. CF and non-CF mice were exposed intratracheally to an adenoviral vector containing the TGFß1 cDNA, empty vector, or PBS only. We assessed methacholine-induced AHR, bronchodilator response, and ASM area in control and CF mice. Notably, CF mice demonstrated enhanced AHR and bronchodilator response with greater ASM area increases compared with non-CF mice. Furthermore, therapeutic inhibition of PI3K signaling mitigated the TGFß-induced AHR and goblet cell hyperplasia in CF mice. These results highlight a latent AHR phenotype in CFTR deficiency that is enhanced through TGFß-induced PI3K signaling.


Subject(s)
Cystic Fibrosis/enzymology , Cystic Fibrosis/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/physiopathology , Signal Transduction , Transforming Growth Factor beta/adverse effects , Adrenergic beta-Agonists/pharmacology , Albuterol/pharmacology , Animals , Bronchoconstriction/drug effects , Goblet Cells/pathology , Hyperplasia , Lung/physiopathology , Mice, Inbred C57BL , Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Signal Transduction/drug effects
3.
Sci Signal ; 12(597)2019 09 03.
Article in English | MEDLINE | ID: mdl-31481522

ABSTRACT

Asthma is a chronic allergic inflammatory airway disease caused by aberrant immune responses to inhaled allergens, which leads to airway hyperresponsiveness (AHR) to contractile stimuli and airway obstruction. Blocking T helper 2 (TH2) differentiation represents a viable therapeutic strategy for allergic asthma, and strong TCR-mediated ERK activation blocks TH2 differentiation. Here, we report that targeting diacylglycerol (DAG) kinase zeta (DGKζ), a negative regulator of DAG-mediated cell signaling, protected against allergic asthma by simultaneously reducing airway inflammation and AHR though independent mechanisms. Targeted deletion of DGKζ in T cells decreased type 2 inflammation without reducing AHR. In contrast, loss of DGKζ in airway smooth muscle cells decreased AHR but not airway inflammation. T cell-specific enhancement of ERK signaling was only sufficient to limit type 2 airway inflammation, not AHR. Pharmacological inhibition of DGK diminished both airway inflammation and AHR in mice and also reduced bronchoconstriction of human airway samples in vitro. These data suggest that DGK is a previously unrecognized therapeutic target for asthma and reveal that the inflammatory and AHR components of asthma are not as interdependent as generally believed.


Subject(s)
Asthma/immunology , Diacylglycerol Kinase/immunology , Inflammation/immunology , Respiratory Hypersensitivity/immunology , Animals , Asthma/enzymology , Asthma/genetics , Bronchoconstriction/drug effects , Bronchoconstriction/genetics , Bronchoconstriction/immunology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Enzyme Inhibitors/pharmacology , Humans , Inflammation/enzymology , Inflammation/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/immunology , Piperidines/pharmacology , Quinazolinones/pharmacology , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Th2 Cells/drug effects , Th2 Cells/enzymology , Th2 Cells/immunology
4.
Int J Chron Obstruct Pulmon Dis ; 13: 1135-1144, 2018.
Article in English | MEDLINE | ID: mdl-29692608

ABSTRACT

BACKGROUND: Airway hyperresponsiveness (AHR) is associated with airway inflammation and a rapid decline in lung function and is a predictor of future risk of COPD among smokers. Alveolar macrophages (AMs) from patients with COPD release a greater amount of matrix metalloproteinase (MMP)-9. We hypothesized that the imbalance between MMP-9 and tissue inhibitor of metalloproteinase-1 (TIMP-1) is related to AHR in smokers. PATIENTS AND METHODS: Healthy smokers with AHR (AHR + S) or smokers without AHR (AHR - S; divided according to a methacholine challenge test) and nonsmokers without AHR (AHR - NS) were enrolled. Spirometry was performed during enrollment and repeated after 5 years. Initially, AMs recovered from bronchoalveolar lavage (BAL) fluid were cultured in the presence of p38 mitogen-activated protein kinase (MAPK) inhibitor (SB203580), MAPK kinase (MEK) 1/2 (the MEK of extracellular signal-regulated kinase [ERK] inhibitor, PD98059), or medium alone for 24 h. The release of MMP-9 and TIMP-1 in culture supernatants was measured by enzyme-linked immunosorbent assay. RESULTS: A greater reduction in forced expiratory volume in 1 s (FEV1)/forced vital capacity (FVC), FEV1 (as a percentage of the predicted value [%pred]), and maximal mid-expiratory flow (MMEF) was observed among AHR + S in the 5-year period. There was a higher proportion of neutrophils and a lower proportion of AMs in BAL fluid recovered from AHR + S. Compared to AMs from AHR - NS and AHR - S, AMs from nonsmokers with AHR (AHR + NS) released more MMP-9 and less TIMP-1, with an increase in MMP-9/TIMP-1 ratios. The MMP-9/TIMP-1 ratio in smokers was positively correlated with the annual decline in FEV1%pred, FVC%pred, and MMEF%pred. Both SB203580 and PD98059 significantly reduced MMP-9, but not TIMP-1, from AMs of smokers. CONCLUSION: AMs of AHR + NS produce excessive MMP-9 over TIMP-1, which may be a predictor of the development of airway obstruction. Inhibition of p38 MAPK and ERK suppresses the generation of MMP-9 by AMs from smokers.


Subject(s)
Lung/enzymology , Macrophages, Alveolar/enzymology , Matrix Metalloproteinase 9/metabolism , Pulmonary Disease, Chronic Obstructive/etiology , Respiratory Hypersensitivity/etiology , Smokers , Smoking/adverse effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , Adult , Aged , Biomarkers/metabolism , Bronchial Provocation Tests , Cells, Cultured , Disease Progression , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Forced Expiratory Volume , Humans , Lung/drug effects , Lung/physiopathology , Macrophages, Alveolar/drug effects , Male , Middle Aged , Protein Kinase Inhibitors/pharmacology , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/physiopathology , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/physiopathology , Smoking/blood , Smoking/physiopathology , Spirometry , Time Factors , Vital Capacity , Young Adult , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
5.
PLoS One ; 13(1): e0190916, 2018.
Article in English | MEDLINE | ID: mdl-29329322

ABSTRACT

RATIONALE: Environmental exposures strongly influence the development and progression of asthma. We have previously demonstrated that mice exposed to a diet enriched with methyl donors during vulnerable periods of fetal development can enhance the heritable risk of allergic airway disease through epigenetic changes. There is conflicting evidence on the role of folate (one of the primary methyl donors) in modifying allergic airway disease. OBJECTIVES: We hypothesized that blocking folate metabolism through the loss of methylene-tetrahydrofolate reductase (Mthfr) activity would reduce the allergic airway disease phenotype through epigenetic mechanisms. METHODS: Allergic airway disease was induced in C57BL/6 and C57BL/6Mthfr-/- mice through house dust mite (HDM) exposure. Airway inflammation and airway hyperresponsiveness (AHR) were measured between the two groups. Gene expression and methylation profiles were generated for whole lung tissue. Disease and molecular outcomes were evaluated in C57BL/6 and C57BL/6Mthfr-/- mice supplemented with betaine. MEASUREMENTS AND MAIN RESULTS: Loss of Mthfr alters single carbon metabolite levels in the lung and serum including elevated homocysteine and cystathionine and reduced methionine. HDM-treated C57BL/6Mthfr-/- mice demonstrated significantly less airway hyperreactivity (AHR) compared to HDM-treated C57BL/6 mice. Furthermore, HDM-treated C57BL/6Mthfr-/- mice compared to HDM-treated C57BL/6 mice have reduced whole lung lavage (WLL) cellularity, eosinophilia, and Il-4/Il-5 cytokine concentrations. Betaine supplementation reversed parts of the HDM-induced allergic airway disease that are modified by Mthfr loss. 737 genes are differentially expressed and 146 regions are differentially methylated in lung tissue from HDM-treated C57BL/6Mthfr-/- mice and HDM-treated C57BL/6 mice. Additionally, analysis of methylation/expression relationships identified 503 significant correlations. CONCLUSION: Collectively, these findings indicate that the loss of folate as a methyl donor is a modifier of allergic airway disease, and that epigenetic and expression changes correlate with this modification. Further investigation into the mechanisms that drive this observation is warranted.


Subject(s)
Methylenetetrahydrofolate Reductase (NADPH2)/physiology , Respiratory Hypersensitivity/enzymology , Animals , Betaine/administration & dosage , DNA Methylation , Gene Expression , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mice , Mice, Inbred C57BL , Quantitative Trait Loci
6.
Allergy ; 72(11): 1744-1752, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28397284

ABSTRACT

BACKGROUND: Fatty acids and lipid mediator signaling play an important role in the pathogenesis of asthma, yet this area remains largely underexplored. The aims of this study were (i) to examine fatty acid levels and their metabolism in obese and nonobese asthma patients and (ii) to determine the functional effects of altered fatty acid metabolism in experimental models. METHODS: Medium- and long-chain fatty acid levels were quantified in serum from 161 human volunteers by LC/MS. Changes in stearoyl-coenzyme A desaturase (SCD) expression and activity were evaluated in the ovalbumin (OVA) and house dust mite (HDM) murine models. Primary human bronchial epithelial cells from asthma patients and controls were evaluated for SCD expression and activity. RESULTS: The serum desaturation index (an indirect measure of SCD) was significantly reduced in nonobese asthma patients and in the OVA murine model. SCD1 gene expression was significantly reduced within the lungs following OVA or HDM challenge. Inhibition of SCD in mice promoted airway hyper-responsiveness. SCD1 expression was suppressed in bronchial epithelial cells from asthma patients. IL-4 and IL-13 reduced epithelial cell SCD1 expression. Inhibition of SCD reduced surfactant protein C expression and suppressed rhinovirus-induced IP-10 secretion, which was associated with increased viral titers. CONCLUSIONS: This is the first study to demonstrate decreased fatty acid desaturase activity in humans with asthma. Experimental models in mice and human epithelial cells suggest that inhibition of desaturase activity leads to airway hyper-responsiveness and reduced antiviral defense. SCD may represent a new target for therapeutic intervention in asthma patients.


Subject(s)
Asthma/metabolism , Fatty Acids/metabolism , Stearoyl-CoA Desaturase/metabolism , Animals , Asthma/enzymology , Bronchi/cytology , Cells, Cultured , Epithelial Cells/enzymology , Fatty Acids/blood , Humans , Lipid Metabolism , Mice , Obesity , Respiratory Hypersensitivity/enzymology
7.
Am J Physiol Lung Cell Mol Physiol ; 309(11): L1344-53, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26453516

ABSTRACT

Most patients with allergic asthma are sensitized to house dust mite (HDM). The allergenicity of HDM largely depends on disruption of the integrity and proinflammatory activation of the airway epithelium. In this study, we hypothesized that Pim1 kinase activity attenuates HDM-induced asthma by preserving airway epithelial integrity. The effects of Pim1 kinase activity on barrier function and release of the proinflammatory mediators IL-1α and CCL20 were studied in vitro in 16HBE and primary bronchial epithelial cells (PBECs). Pim1-proficient and -deficient mice were exposed to a HDM-driven model of allergic asthma, and airway hyperresponsiveness (AHR) was measured upon methacholine challenge. Airway inflammation and proinflammatory mediators in lung tissue and BAL fluid were determined. We observed that inhibition of Pim1 kinase prolongs the HDM-induced loss of barrier function in 16HBE cells and sensitizes PBECs to HDM-induced barrier dysfunction. Additionally, inhibition of Pim1 kinase increased the HDM-induced proinflammatory activity of 16HBE cells as measured by IL-1α secretion. In line herewith, HDM exposure induced an enhanced production of the proinflammatory chemokines CCL17 and CCL20 in Pim1-deficient mice compared with wild-type controls. While we observed a marked increase in eosinophilic and neutrophilic granulocytes as well as mucus cell metaplasia and AHR to methacholine in mice exposed to HDM, these parameters were independent of Pim1 kinase activity. In contrast, levels of the Th2-cytokines IL-5 and IL-10 were significantly augmented in HDM-treated Pim1-deficient mice. Taken together, our study shows that Pim1 kinase activity maintains airway epithelial integrity and protects against HDM-induced proinflammatory activation of the airway epithelium.


Subject(s)
Bronchi/pathology , Epithelial Cells/enzymology , Epithelial Cells/parasitology , Proto-Oncogene Proteins c-pim-1/metabolism , Pyroglyphidae/physiology , Adult , Aged , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Line , Chemokines/metabolism , Epithelial Cells/pathology , Female , Humans , Inflammation/parasitology , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Mice , Middle Aged , Pneumonia/pathology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/deficiency , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/parasitology , Respiratory Hypersensitivity/pathology , Th2 Cells/immunology , Young Adult
8.
Clin Exp Allergy ; 45(2): 457-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25323425

ABSTRACT

BACKGROUND: Major features of allergic asthma include airway hyperresponsiveness (AHR), eosinophilic inflammation, and goblet cell metaplasia. Rho kinase (ROCK) is a serine/threonine protein kinase that regulates the actin cytoskeleton. By doing so, it can modulate airway smooth muscle cell contraction and leucocyte migration and proliferation. This study was designed to determine the contributions of the two ROCK isoforms, ROCK1 and ROCK2, to AHR, inflammation and goblet cell metaplasia in a mast cell-dependent model of allergic airways disease. METHODS AND RESULTS: Repeated intranasal challenges with OVA caused AHR, eosinophilic inflammation, and goblet cell hyperplasia in wild-type (WT) mice. OVA-induced AHR was partially or completely abrogated in mice haploinsufficient for ROCK2 (ROCK2(+/-) ) or ROCK1 (ROCK1(+/-) ), respectively. In contrast, there was no effect of ROCK insufficiency on allergic airways inflammation, although both ROCK1 and ROCK2 insufficiency attenuated mast cell degranulation. Goblet cell hyperplasia, as indicated by PAS staining, was not different in ROCK1(+/-) vs. WT mice. However, in ROCK2(+/-) mice, goblet cell hyperplasia was reduced in medium but not large airways. Maximal acetylcholine-induced force generation was reduced in tracheal rings from ROCK1(+/-) and ROCK2(+/-) vs. WT mice. The ROCK inhibitor, fasudil, also reduced airway responsiveness in OVA-challenged mice, without affecting inflammatory responses. CONCLUSION: In a mast cell model of allergic airways disease, ROCK1 and ROCK2 both contribute to AHR, likely through direct effects on smooth muscle cell and effects on mast cell degranulation. In addition, ROCK2 but not ROCK1 plays a role in allergen-induced goblet cell hyperplasia.


Subject(s)
Respiratory Hypersensitivity/enzymology , rho-Associated Kinases/metabolism , Allergens/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation/genetics , Female , Goblet Cells/metabolism , Goblet Cells/pathology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Inflammation Mediators/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Knockout , Ovalbumin/immunology , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/physiopathology , Th2 Cells/immunology , Th2 Cells/metabolism , rho-Associated Kinases/genetics
9.
DNA Repair (Amst) ; 12(1): 18-26, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23127499

ABSTRACT

Allergic airway inflammation is characterized by increased expression of pro-inflammatory mediators, inflammatory cell infiltration, mucus hypersecretion, and airway hyperresponsiveness, in parallel with oxidative DNA base and strand damage, whose etiological role is not understood. Our goal was to establish the role of 8-oxoguanine (8-oxoG), a common oxidatively damaged base, and its repair by 8-oxoguanine DNA glycosylase 1 (Ogg1) in allergic airway inflammatory processes. Airway inflammation was induced by intranasally administered ragweed (Ambrosia artemisiifolia) pollen grain extract (RWPE) in sensitized BALB/c mice. We utilized siRNA technology to deplete Ogg1 from airway epithelium; 8-oxoG and DNA strand break levels were quantified by Comet assays. Inflammatory cell infiltration and epithelial methaplasia were determined histologically, mucus and cytokines levels biochemically and enhanced pause was used as the main index of airway hyperresponsiveness. Decreased Ogg1 expression and thereby 8-oxoG repair in the airway epithelium conveyed a lower inflammatory response after RWPE challenge of sensitized mice, as determined by expression of Th2 cytokines, eosinophilia, epithelial methaplasia, and airway hyperresponsiveness. In contrast, 8-oxoG repair in Ogg1-proficient airway epithelium was coupled to an increase in DNA single-strand break (SSB) levels and exacerbation of allergen challenge-dependent inflammation. Decreased expression of the Nei-like glycosylases Neil1 and Neil2 that preferentially excise ring-opened purines and 5-hydroxyuracil, respectively, did not alter the above parameters of allergic immune responses to RWPE. These results show that DNA SSBs formed during Ogg1-mediated repair of 8-oxoG augment antigen-driven allergic immune responses. A transient modulation of OGG1 expression/activity in airway epithelial cells could have clinical benefits.


Subject(s)
DNA Glycosylases/genetics , DNA Repair , Respiratory Hypersensitivity/enzymology , Respiratory Mucosa/enzymology , Ambrosia/immunology , Animals , Cell Line , DNA/metabolism , DNA Breaks, Single-Stranded , DNA Glycosylases/metabolism , Down-Regulation , Guanine/analogs & derivatives , Guanine/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Pneumonia/enzymology , Pneumonia/genetics , Pneumonia/immunology , Pollen/immunology , RNA, Small Interfering , Respiratory Hypersensitivity/genetics , Respiratory Mucosa/immunology
10.
Int Arch Allergy Immunol ; 159(2): 157-61, 2012.
Article in English | MEDLINE | ID: mdl-22652554

ABSTRACT

BACKGROUND: Aspirin-exacerbated respiratory disease (AERD) is a clinical syndrome associated with chronic inflammation in the airways coincident with chronic rhinitis, sinusitis, recurrent polyposis and asthma. Eosinophils are the key inflammatory cells in the development of AERD. AERD has been attributed to abnormalities of the arachidonic acid metabolism, but the pathogenesis of AERD is not fully understood. Our aim was to investigate the genetic contribution of the arachidonate 15-lipoxygenase gene (ALOX15) to the development of AERD. METHODS: We enrolled 171 patients with AERD, 229 patients with aspirin-tolerant asthma, and 195 normal healthy controls in a Korean population. Three polymorphisms (-427G/A, -272C/A, -217G/C) in the promoter region of ALOX15 were genotyped. The functional variability of the promoter polymorphisms were analyzed by luciferase reporter activity assay. RESULT: No significant difference in the genotype frequency of the ALOX15 genetic polymorphism was found. Peripheral total eosinophil count was significantly higher in the patients carrying the GG genotype of the -427G/A polymorphism (p = 0.016). Similarly, the patients carrying haplotype 1 (ht1) (GCG) of -427G/A, -272C/A and -217G/C showed a significantly higher total eosinophil count compared to the other haplotypes (p = 0.008) in the AERD group. The promoter activity of the ht1 (GCG) construct was significantly higher compared to that of the ht3 (AGG) construct in A549 and U937 cells (both p < 0.001). CONCLUSION: These results suggest that the promoter polymorphisms of the ALOX15 gene affect ALOX15 activity leading to increased eosinophil infiltration in AERD patients.


Subject(s)
Arachidonate 15-Lipoxygenase/genetics , Aspirin/adverse effects , Polymorphism, Single Nucleotide , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/genetics , Adult , Arachidonate 15-Lipoxygenase/metabolism , Asian People/genetics , Asthma/enzymology , Asthma/etiology , Asthma/genetics , Asthma, Aspirin-Induced/enzymology , Asthma, Aspirin-Induced/etiology , Asthma, Aspirin-Induced/genetics , Base Sequence , Case-Control Studies , DNA Primers/genetics , Eosinophils/enzymology , Eosinophils/immunology , Female , Gene Frequency , Haplotypes , Humans , Leukocyte Count , Male , Middle Aged , Promoter Regions, Genetic , Republic of Korea , Respiratory Hypersensitivity/etiology , Young Adult
11.
Clin Exp Pharmacol Physiol ; 38(4): 208-14, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21261675

ABSTRACT

1. Formaldehyde (FA) has been found to cause toxicity to neurons. However, its neurotoxic mechanisms have not yet been clarified. Increasing evidence has shown that oxidative damage is one of the most critical effects of formaldehyde exposure. Paraoxonase-1 (PON-1) is a pivotal endogenous anti-oxidant. Thus, we hypothesized that FA-mediated downregulation of PON1 is associated with its neurotoxicity. 2. In the present work, we used PC12 cells to study the neurotoxicity of FA and explore whether PON-1 is implicated in FA-induced neurotoxicity. 3. We found that FA has potent cytotoxic and apoptotic effects on PC12 cells. FA induces an accumulation of intracellular reactive oxygen species along with downregulation of Bcl-2 expression, as well as increased cytochrome c release. FA significantly suppressed the expression and activity of PON-1 in PC12 cells. Furthermore, H(2)S, an endogenous anti-oxidant gas, antagonizes FA-induced cytotoxicity as well as 2-hydroxyquinoline, a specific inhibitor of PON-1, which also induces cytotoxicity to PC12 cells. 4. The results of the present study provide, for the first time, evidence that the inhibitory effect on PON-1 expression and activity is involved in the neurotoxicity of FA, and suggest a promising role of PON-1 as a novel therapeutic strategy for FA-mediated toxicity.


Subject(s)
Aryldialkylphosphatase/metabolism , Formaldehyde/toxicity , Neurons/drug effects , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/etiology , Animals , Apoptosis/drug effects , Aryldialkylphosphatase/antagonists & inhibitors , Aryldialkylphosphatase/biosynthesis , Aryldialkylphosphatase/genetics , Carboxylic Ester Hydrolases/metabolism , Cell Line, Tumor , Cytochromes c/genetics , Cytochromes c/metabolism , Down-Regulation/genetics , Formaldehyde/adverse effects , Formaldehyde/metabolism , Hydrogen Sulfide/pharmacology , Hydroxyquinolines/pharmacology , Neurons/enzymology , Neurotoxicity Syndromes/metabolism , Oxidative Stress/drug effects , PC12 Cells , Rats , Reactive Oxygen Species/metabolism , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/metabolism , bcl-Associated Death Protein/antagonists & inhibitors , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism
12.
J Immunol ; 186(5): 3164-72, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270400

ABSTRACT

We have shown that proteinase-activated receptor-2 (PAR(2)) activation in the airways leads to allergic sensitization to concomitantly inhaled Ags, thus implicating PAR(2) in the pathogenesis of asthma. Many aeroallergens with proteinase activity activate PAR(2). To study the role of PAR(2) in allergic sensitization to aeroallergens, we developed a murine model of mucosal sensitization to cockroach proteins. We hypothesized that PAR(2) activation in the airways by natural allergens with serine proteinase activity plays an important role in allergic sensitization. Cockroach extract (CE) was administered to BALB/c mice intranasally on five consecutive days (sensitization phase) and a week later for four more days (challenge phase). Airway hyperresponsiveness (AHR) and allergic airway inflammation were assessed after the last challenge. To study the role of PAR(2), mice were exposed intranasally to a receptor-blocking anti-PAR(2) Ab before each administration of CE during the sensitization phase. Mucosal exposure to CE induced eosinophilic airway inflammation, AHR, and cockroach-specific IgG1. Heat-inactivated or soybean trypsin inhibitor-treated CE failed to induce these effects, indicating that proteinase activity plays an important role. The use of an anti-PAR(2) blocking Ab during the sensitization phase completely inhibited airway inflammation and also decreased AHR and the production of cockroach-specific IgG1. PAR(2) activation by CE acts as an adjuvant for allergic sensitization even in the absence of functional TLR4. We conclude that CE induces PAR(2)-dependent allergic airway sensitization in a mouse model of allergic airway inflammation. PAR(2) activation may be a general mechanism used by aeroallergens to induce allergic sensitization.


Subject(s)
Allergens/immunology , Blattellidae/immunology , Receptor, PAR-2/metabolism , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/physiology , Administration, Intranasal , Allergens/administration & dosage , Animals , Blattellidae/enzymology , Bronchial Hyperreactivity/enzymology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Cell Line, Transformed , Disease Models, Animal , Enzyme Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nasal Mucosa/enzymology , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Rats , Receptor, PAR-2/deficiency , Receptor, PAR-2/immunology , Respiratory Hypersensitivity/enzymology
13.
Int Forum Allergy Rhinol ; 1(2): 106-8, 2011.
Article in English | MEDLINE | ID: mdl-22287327

ABSTRACT

BACKGROUND: Collecting mucosal biopsies is invasive and creates additional inflammation, hampering a better understanding of nasal and sinus disease evolution and response to treatment. We examine whether sinus secretion collection can replace tissue biopsy for protein determination. METHODS: Prior to surgical intervention for chronic rhinosinusitis (CRS), a piece of gelatin foam was used to collect secretions from the ethmoid mucosa. A tissue biopsy was then taken from the same location. Matrix metalloproteinase-9 (MMP-9) protein levels were measured in each sample. RESULTS: MMP-9 protein levels in secretions and tissues were significantly correlated (p = 0.0033, r = 0.52, by Pearson correlation). CONCLUSION: Secretion collection can replace tissue biopsy for MMP-9 determinations, reducing morbidity. Furthermore, secretion collection allows sequential sampling from the same location.


Subject(s)
Ethmoid Sinusitis/enzymology , Matrix Metalloproteinase 9/metabolism , Nasal Mucosa/pathology , Rhinitis, Allergic, Perennial/enzymology , Adolescent , Adult , Aged , Biopsy , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Ethmoid Sinusitis/pathology , Humans , Middle Aged , Nasal Mucosa/enzymology , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/pathology , Rhinitis, Allergic, Perennial/pathology , Young Adult
14.
J Immunol ; 185(6): 3489-97, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20702731

ABSTRACT

Chromatin modifications, such as reversible histone acetylation, play a key role in the regulation of T cell development and function. However, the role of individual histone deacetylases (HDACs) in T cells is less well understood. In this article, we show by conditional gene targeting that T cell-specific loss of HDAC1 led to an increased inflammatory response in an in vivo allergic airway inflammation model. Mice with HDAC1-deficient T cells displayed an increase in all critical parameters in this Th2-type asthma model, such as eosinophil recruitment into the lung, mucus hypersecretion, parenchymal lung inflammation, and enhanced airway resistance. This correlated with enhanced Th2 cytokine production in HDAC1-deficient T cells isolated from diseased mice. In vitro-polarized HDAC1-deficient Th2 cells showed a similar enhancement of IL-4 expression, which was evident already at day 3 of Th2 differentiation cultures and restricted to T cell subsets that underwent several rounds of cell divisions. HDAC1 was recruited to the Il4 gene locus in ex vivo isolated nonstimulated CD4(+) T cells, indicating a direct control of the Il4 gene locus. Our data provide genetic evidence that HDAC1 is an essential HDAC that controls the magnitude of an inflammatory response by modulating cytokine expression in effector T cells.


Subject(s)
Cytokines/biosynthesis , Histone Deacetylase 1/deficiency , Lung/immunology , Lung/pathology , Th1 Cells/immunology , Th2 Cells/immunology , Up-Regulation/immunology , Animals , Cell Polarity/genetics , Cell Polarity/immunology , Cells, Cultured , Disease Models, Animal , Histone Deacetylase 1/genetics , Histone Deacetylase 1/physiology , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Lung/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Th1 Cells/enzymology , Th1 Cells/pathology , Th2 Cells/enzymology , Th2 Cells/pathology , Up-Regulation/genetics
15.
J Med Chem ; 53(16): 6122-8, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20666458

ABSTRACT

Acidic mammalian chitinase (AMCase) is a member of the glycosyl hydrolase 18 family (EC 3.2.1.14) that has been implicated in the pathophysiology of allergic airway disease such as asthma. Small molecule inhibitors of AMCase were identified using a combination of high-throughput screening, fragment screening, and virtual screening techniques and characterized by enzyme inhibition and NMR and Biacore binding experiments. X-ray structures of the inhibitors in complex with AMCase revealed that the larger more potent HTS hits, e.g. 5-(4-(2-(4-bromophenoxy)ethyl)piperazine-1-yl)-1H-1,2,4-triazol-3-amine 1, spanned from the active site pocket to a hydrophobic pocket. Smaller fragments identified by FBS occupy both these pockets independently and suggest potential strategies for linking fragments. Compound 1 is a 200 nM AMCase inhibitor which reduced AMCase enzymatic activity in the bronchoalveolar lavage fluid in allergen-challenged mice after oral dosing.


Subject(s)
Chitinases/antagonists & inhibitors , Models, Molecular , Piperazines/chemical synthesis , Triazoles/chemical synthesis , Allergens/immunology , Animals , Bronchoalveolar Lavage Fluid , Catalytic Domain , Crystallography, X-Ray , Female , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Piperazines/chemistry , Piperazines/pharmacology , Protein Binding , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/immunology , Structure-Activity Relationship , Surface Plasmon Resonance , Triazoles/chemistry , Triazoles/pharmacology
16.
Biochimie ; 92(6): 588-93, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20219624

ABSTRACT

Secreted phospholipases A(2) (sPLA(2)s) are molecules released in plasma and biological fluids of patients with systemic inflammatory, autoimmune and allergic diseases. These molecules exert proinflammatory effects by either enzymatic-mechanisms or through binding to surface molecules expressed on inflammatory cells. sPLA(2)s are released at low levels in the normal airways and tend to increase during respiratory allergies (e.g., rhinitis and bronchial asthma) as the result of local secretion. Several sPLA(2) isoforms are expressed in the human lung and some of them (e.g., group IIA and group X) are released in the airways of patients with rhinitis or asthma. Mast cells play a major role in the pathogenesis of respiratory allergies and other chronic inflammatory lung diseases. Recent evidence indicates that mast cells purified from human lung express most of the sPLA(2) isoforms so far described. IgE-mediated activation of these cells induce the release of sPLA(2)s suggesting that mast cells are a main source of extracellular sPLA(2)s during allergic reactions. Once released, sPLA(2)s may contribute to the generation of eicosanoids (e.g., PGD(2) and LTC(4)) and to the release of preformed mediators (e.g., histamine) by an autocrine loop involving the interaction of sPLA(2)s with surface molecules such as heparan sulphate proteoglycans or the M-type receptor. Thus, mast cell-derived sPLA(2)s may play an important role in the initiation and amplification of the inflammatory reactions in patients with allergic rhinitis and bronchial asthma.


Subject(s)
Mast Cells/enzymology , Mast Cells/immunology , Phospholipases A2, Secretory/metabolism , Respiratory Hypersensitivity/enzymology , Animals , Asthma/enzymology , Asthma/immunology , Humans , Lung/immunology , Mast Cells/metabolism , Models, Biological , Respiratory System/immunology
17.
Clin Exp Allergy ; 40(5): 820-30, 2010 May.
Article in English | MEDLINE | ID: mdl-20337645

ABSTRACT

Background Asthma is a disease characterized by airway inflammation, remodelling and dysfunction. Airway inflammation contributes to remodelling, a term that is used to describe structural changes including goblet cell metaplasia (GCM), matrix deposition, and smooth muscle hyperplasia/hypertrophy. GCM has been implicated in asthma mortality by contributing to mucus plugs and leading to asphyxiation. In animal models, this process is highly dependent on IL-13. Recently, we have described an IL-13-dependent up-regulation of a GABAergic signalling system in airway epithelium that contributes to GCM. The mechanism by which IL-13 up-regulates GABA signalling in airway epithelium is unknown. Objectives To test the hypothesis that IL-4Ralpha signalling is required for allergen induced up-regulation of GABAergic signalling and GCM. Methods BALB/c mice were exposed to an acute house dust mite (HDM) protocol and received vehicle, anti-IL-4Ralpha-monoclonal antibody, or control antibody. Outcomes included airway responses to inhaled methacholine (MCh), histology for eosinophilia and GCM, phosphorylated STAT6 levels using immunohistochemistry and immunoblot, and glutamic acid decarboxylase (GAD) 65/67 and GABA(A)beta(2/3) receptor subunit expression using confocal microscopy. Results Acute HDM exposure resulted in increased airway responses to MCh, lung eosinophilia, STAT6 phosphorylation, elevations in GAD65/67 and GABA(A)beta(2/3) receptor expression, and GCM that were inhibited with anti-IL-4Ralpha-monoclonal treatment. Control antibody had no effect. Conclusion The IL-4Ralpha is required for allergen-induced up-regulation of a GABAergic system in airway epithelium implicated in GCM following acute HDM exposure.


Subject(s)
Glutamate Decarboxylase/metabolism , Interleukin-4 Receptor alpha Subunit/physiology , Pyroglyphidae/immunology , Respiratory Hypersensitivity/enzymology , Respiratory Mucosa/enzymology , Allergens/immunology , Animals , Eosinophils/cytology , Goblet Cells/pathology , Lung/immunology , Metaplasia/pathology , Mice , Mice, Inbred BALB C , Phosphorylation , Receptors, GABA/metabolism , Receptors, GABA-A/metabolism , Respiratory Hypersensitivity/immunology , Respiratory Mucosa/immunology , STAT6 Transcription Factor/metabolism , Up-Regulation
18.
Toxicol Appl Pharmacol ; 242(1): 1-8, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19800904

ABSTRACT

Arginase1 and nitric oxide synthase2 (NOS2) utilize l-arginine as a substrate, with both enzymes expressed at high levels in the asthmatic lung. Inhibition of arginase in ovalbumin-exposed C57BL/6 mice with the transition state inhibitor N(omega)-hydroxy-nor-l-arginine (nor-NOHA) significantly increased total l-arginine content in the airway compartment. We hypothesized that such an increase in l-arginine content would increase the amount of nitric oxide (NO) being produced in the airways and thereby decrease airway hyperreactivity and eosinophilic influx. We further hypothesized that despite arginase inhibition, NOS2 knockout (NOS2-/-) mice would be unable to up-regulate NO production in response to allergen exposure and would demonstrate higher amounts of airway hyperreactivity and eosinophilia under conditions of arginase inhibition than C57BL/6 animals. We found that administration of nor-NOHA significantly decreased airway hyperreactivity and eosinophilic airway inflammation in ovalbumin-exposed C57BL/6 mice, but these parameters were unchanged in ovalbumin-exposed NOS2-/- mice. Arginase1 protein content was increased in mice exposed to ovalbumin, an effect that was reversed upon nor-NOHA treatment in C57BL/6 mice. Arginase1 protein content in the airway compartment directly correlated with the degree of airway hyperreactivity in all treatment groups. NOS2-/- mice had significantly greater arginase1 and arginase2 concentrations compared to their respective C57BL/6 groups, indicating that inhibition of arginase may be dependent upon NOS2 expression. Arginase1 and 2 content were not affected by nor-NOHA administration in the NOS2-/- mice. We conclude that l-arginine metabolism plays an important role in the development of airway hyperreactivity and eosinophilic airway inflammation. Inhibition of arginase early in the allergic inflammatory response decreases the severity of the chronic inflammatory phenotype. These effects appear to be attributable to NOS2, which is a major source of NO production in the inflamed airway, although arginase inhibition may also be affecting the turnover of arginine by the other NOS isoforms, NOS1 and NOS3. The increased l-arginine content in the airway compartment of mice treated with nor-NOHA may directly or indirectly, through NOS2, control arginase expression both in response to OVA exposure and at a basal level.


Subject(s)
Arginase/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/physiology , Ovalbumin/immunology , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/genetics , Aerosols , Airway Resistance/drug effects , Animals , Arginase/biosynthesis , Arginine/analogs & derivatives , Arginine/metabolism , Arginine/pharmacology , Blotting, Western , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/genetics , Lung/pathology , Lung Compliance/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Ovalbumin/administration & dosage , Pneumonia/pathology
19.
BMC Immunol ; 10: 33, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19486531

ABSTRACT

BACKGROUND: Arginase is significantly upregulated in the lungs in murine models of asthma, as well as in human asthma, but its role in allergic airway inflammation has not been fully elucidated in mice. RESULTS: In order to test the hypothesis that arginase has a role in allergic airway inflammation we generated arginase I-deficient bone marrow (BM) chimeric mice. Following transfer of arginase I-deficient BM into irradiated recipient mice, arginase I expression was not required for hematopoietic reconstitution and baseline immunity. Arginase I deficiency in bone marrow-derived cells decreased allergen-induced lung arginase by 85.8 +/- 5.6%. In contrast, arginase II-deficient mice had increased lung arginase activity following allergen challenge to a similar level to wild type mice. BM-derived arginase I was not required for allergen-elicited sensitization, recruitment of inflammatory cells in the lung, and proliferation of cells. Furthermore, allergen-induced airway hyperresponsiveness and collagen deposition were similar in arginase-deficient and wild type mice. Additionally, arginase II-deficient mice respond similarly to their control wild type mice with allergen-induced inflammation, airway hyperresponsiveness, proliferation and collagen deposition. CONCLUSION: Bone marrow cell derived arginase I is the predominant source of allergen-induced lung arginase but is not required for allergen-induced inflammation, airway hyperresponsiveness or collagen deposition.


Subject(s)
Allergens/immunology , Arginase , Bone Marrow Cells/enzymology , Lung/metabolism , Radiation Chimera , Respiratory Hypersensitivity/enzymology , Animals , Arginase/immunology , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Collagen/metabolism , Hyperargininemia , Immunization , Inflammation , Lung/immunology , Lung/pathology , Mice , Respiratory Hypersensitivity/pathology
20.
Clin Exp Allergy ; 39(8): 1179-86, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19400896

ABSTRACT

BACKGROUND: Secretory leucocyte protease inhibitor (SLPI), which is present in many physiological fluids including saliva, sputum and nasal discharge, is the most effective inhibitor of chymase. Previously, we demonstrated that chymase is able to cleave SLPI and that the cleaved portion, cSLPI, is a biomarker of chymase activity. OBJECTIVE: We investigated the potential of cSLPI as a biomarker of chymase activity in subjects with allergic rhinitis (AR) and asthmatic airway disease. METHODS: Baseline sputum samples were collected from atopic asthmatics and healthy controls (HC). Nasal lavages (NAL) were performed in subjects with AR both at baseline and following a nasal challenge with allergen or placebo. Levels of cSLPI and chymase were determined by Western analysis, and tryptase and alpha-2 macroglobulin were measured by immunoassay. RESULTS: As compared with HC, asthmatics showed a significant increase in baseline cSLPI/total SLPI ratios and an increase in chymase levels. There was a high correlation of cSLPI/SLPI ratios to chymase levels in normal individuals and untreated asthmatics. In the NAL of patients with AR, as compared with placebo, allergen challenge increased inflammatory biomarkers, including cSLPI/SLPI ratios, chymase levels, tryptase levels and alpha2-macroglobulin levels. Correlations were observed between cSLPI/SLPI ratios and chymase levels and cSLPI/SLPI ratios and alpha2-macroglobulin levels; no correlation was seen between cSLPI/SLPI ratios and tryptase levels. CONCLUSION: Our data indicate that cSLPI reflects chymase activity in AR and asthma. Hence, cSLPI may serve as a biomarker for disease activity and for monitoring the efficacy of novel anti-inflammatory treatments in chymase-mediated diseases.


Subject(s)
Chymases/metabolism , Respiratory Hypersensitivity/metabolism , Secretory Leukocyte Peptidase Inhibitor/metabolism , Adolescent , Adult , Aged , Biomarkers/metabolism , Female , Humans , Male , Middle Aged , Nasal Lavage Fluid/chemistry , Nasal Lavage Fluid/immunology , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/immunology , Retrospective Studies , Rhinitis, Allergic, Seasonal/immunology , Rhinitis, Allergic, Seasonal/metabolism , Secretory Leukocyte Peptidase Inhibitor/immunology , Sputum/enzymology , Sputum/immunology , Sputum/metabolism , Tryptases/metabolism , alpha-Macroglobulins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL