Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
Chest ; 160(1): 114-134, 2021 07.
Article in English | MEDLINE | ID: mdl-33819471

ABSTRACT

Identification of pathologic changes in early and mild obstructive lung disease has shown the importance of the small airways and their contribution to symptoms. Indeed, significant small airways dysfunction has been found prior to any overt airway obstruction being detectable by conventional spirometry techniques. However, most therapies for the treatment of obstructive lung disease target the physiological changes and associated symptoms that result from chronic lung disease, rather than directly targeting the specific underlying causes of airflow disruption or the drivers of disease progression. In addition, although spirometry is the current standard for diagnosis and monitoring of response to therapy, the most widely used measure, FEV1 , does not align with the pathologic changes in early or mild disease and may not align with symptoms or exacerbation frequency in the individual patient. Newer functional and imaging techniques allow more effective assessment of small airways dysfunction; however, significant gaps in our understanding remain. Improving our knowledge of the role of small airways dysfunction in early disease in the airways, along with the identification of novel end points to measure subclinical changes in this region (ie, those not captured as symptoms or identified through standard FEV1), may lead to the development of novel therapies that directly combat early airways disease processes with a view to slowing disease progression and reversing damage. This expert opinion paper discusses small airways disease in the context of asthma and COPD and highlights gaps in current knowledge that impede earlier identification of obstructive lung disease and the development and standardization of novel small airways-specific end points for use in clinical trials.


Subject(s)
Asthma/therapy , Disease Management , Lung/physiopathology , Pulmonary Disease, Chronic Obstructive/therapy , Asthma/physiopathology , Disease Progression , Humans , Pulmonary Disease, Chronic Obstructive/physiopathology , Respiratory Function Tests/methods
3.
BMC Pulm Med ; 14: 110, 2014 Jul 04.
Article in English | MEDLINE | ID: mdl-24993465

ABSTRACT

BACKGROUND: Improvements in asthma diagnosis and management require deeper understanding of the heterogeneity of the complex airway inflammation. We hypothesise that differences in the two major inflammatory phenotypes of asthma; eosinophilic and neutrophilic asthma, will be reflected in the lung protein expression profile of murine asthma models and can be delineated using proteomics of bronchoalveolar lavage (BAL). METHODS: BAL from mice challenged with ovalbumin (OVA/OVA) alone (standard model of asthma, here considered eosinophilic) or OVA in combination with endotoxin (OVA/LPS, model of neutrophilic asthma) was analysed using liquid chromatography coupled to high resolution mass spectrometry, and compared with steroid-treated animals and healthy controls. In addition, conventional inflammatory markers were analysed using multiplexed ELISA (Bio-Plex™ assay). Multivariate statistics was performed on integrative proteomic fingerprints using principal component analysis. Proteomic data were complemented with lung mechanics and BAL cell counts. RESULTS: Several of the analysed proteins displayed significant differences between the controls and either or both of the two models reflecting eosinophilic and neutrophilic asthma. Most of the proteins found with mass spectrometry analysis displayed a considerable increase in neutrophilic asthma compared with the other groups. Conversely, the larger number of the inflammatory markers analysed with Bio-Plex™ analysis were found to be increased in the eosinophilic model. In addition, major inflammation markers were correlated to peripheral airway closure, while commonly used asthma biomarkers only reflect central inflammation. CONCLUSION: Our data suggest that the commercial markers we are currently relying on to diagnose asthma subtypes are not giving us comprehensive or specific enough information. The analysed protein profiles allowed to discriminate the two models and may add useful information for characterization of different asthma phenotypes.


Subject(s)
Asthma/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Eosinophils , Inflammation Mediators/analysis , Neutrophils , Proteome/analysis , Animals , Anti-Inflammatory Agents/pharmacology , Asthma/chemically induced , Asthma/physiopathology , Biomarkers/analysis , Bronchial Provocation Tests , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Female , Hydrocortisone/analogs & derivatives , Hydrocortisone/pharmacology , Inflammation/chemically induced , Inflammation/metabolism , Leukocyte Count , Lipopolysaccharides , Mass Spectrometry , Methacholine Chloride , Mice , Mice, Inbred BALB C , Ovalbumin , Phenotype , Protein Array Analysis , Proteome/drug effects , Respiratory Mechanics
4.
J Appl Physiol (1985) ; 109(2): 521-31, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20538845

ABSTRACT

Glucocorticoids (GC) remain the first choice of treatment in asthma, but GC therapy is not always effective and is associated with side effects. In a porcine study in our laboratory, simultaneous administration of GC and nitric oxide (NO) attenuated the endotoxin-induced inflammatory response and made GC treatment more effective than inhaled NO or steroids alone. In the present study, we aimed to further investigate the interactions between NO and GC treatment in two murine models of asthma. Inflammation was induced by endotoxin, ovalbumin, or a combination of both. With an animal ventilator and a forced oscillation method (FlexiVent), lung mechanics and airway reactivity to methacholine in response to various treatments were assessed. We also describe histology and glucocorticoid receptor (GR) protein expression in response to inhaled NO treatment [40 ppm NO gas or NO donors sodium nitroprusside (SNP) or diethylamine NONOate (DEA/NO)]. SNP and GC provided protection against bronchoconstriction to a similar degree in the model of severe asthma. When GC-treated mice were given SNP, maximum airway reactivity was further reduced. Similar effects were seen after DEA/NO delivery to GC-treated animals. Using 1-H-[1,2,4]-oxadiazolo-[4,3-a]-quinoxalin-1-one (ODQ), a soluble guanylate cyclase inhibitor, we found this effect of NO donors to be mediated through a cGMP-independent mechanism. In the severe model, prolonged NO treatment restored or even increased the nuclear levels of GR. In conclusion, in our murine model of severe asthma GC treatment provided protection to only a limited degree against bronchoconstriction, while concomitant treatment with a NO donor was markedly more potent than the use of either NO or GC alone.


Subject(s)
Anti-Asthmatic Agents/administration & dosage , Asthma/drug therapy , Bronchoconstriction/drug effects , Glucocorticoids/administration & dosage , Nitric Oxide Donors/administration & dosage , Nitric Oxide/administration & dosage , Pneumonia/drug therapy , Administration, Inhalation , Animals , Asthma/chemically induced , Asthma/metabolism , Asthma/physiopathology , Bronchial Provocation Tests , Bronchoconstrictor Agents , Cyclic GMP/metabolism , Disease Models, Animal , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , Female , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Hydrazines/administration & dosage , Hydrocortisone/administration & dosage , Hydrocortisone/analogs & derivatives , Injections, Intraperitoneal , Lipopolysaccharides , Methacholine Chloride , Mice , Mice, Inbred BALB C , Nitroprusside/administration & dosage , Ovalbumin , Oxadiazoles/pharmacology , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/physiopathology , Quinoxalines/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/metabolism , Respiration, Artificial , Respiratory Mechanics/drug effects , Soluble Guanylyl Cyclase
5.
J Immunol ; 183(10): 6369-76, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19841188

ABSTRACT

It is widely established that mast cells (MCs) have a harmful role in asthma, for example by secreting various proinflammatory substances stored within their secretory granule. However, in this study, we show that one of the substances stored within MC granule, chymase, in fact has a protective role in allergic airway inflammation, indicating that MCs may possess both harmful and protective activities in connection with this type of disease. Wild-type (WT) mice and mice lacking mouse MC protease 4 (mMCP-4), a chymase that is functionally homologous to human chymase, were sensitized and challenged with OVA, followed by the assessment of airway physiology and inflammatory parameters. Our results show that the airway hyperresponsiveness was significantly higher in mMCP-4(-/-) as compared with WT mice. Moreover, the degree of lung tissue inflammation was markedly higher in mice lacking mMCP-4 than in WT controls. Histological analysis revealed that OVA sensitization/challenge resulted in a marked increased in the thickness of the smooth muscle cell (SMC) layer and, notably, that the degree of SMC layer thickening was more pronounced in mMCP-4(-/-) animals than in WT controls, thus indicating that chymase may have an effect on airway SMCs. In support of this, mMCP-4-positive MCs were located in the close vicinity of the SMC layer, mainly in the upper airways, and mMCP-4 was shown to be the major chymase expressed in these MCs. Taken together, our results indicate that chymase present in the upper airways protects against allergic airway responses, possibly by regulating SMCs.


Subject(s)
Hypersensitivity/immunology , Inflammation/immunology , Mast Cells/immunology , Serine Endopeptidases/immunology , Animals , Bronchial Hyperreactivity/enzymology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Hypersensitivity/enzymology , Inflammation/enzymology , Lung/immunology , Lung/pathology , Mast Cells/cytology , Mast Cells/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/physiology , Ovalbumin/immunology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
6.
Respir Physiol Neurobiol ; 165(2-3): 229-36, 2009 Feb 28.
Article in English | MEDLINE | ID: mdl-19136080

ABSTRACT

Airway responses to intravenous (i.v.) and inhaled (i.h.) delivery of methacholine (MCh) in BALB/c and C57BL/6 mouse strains have been compared with and without ovalbumin (OVA)-induced airway inflammation. Bronchial reactivity to MCh was assessed in anaesthetised and tracheostomised animals by using an animal ventilator (flexiVent). We partitioned the response of the lungs into airway and parenchymal components in order to compare the contributions of the airways with those of the lung parenchyma to the pulmonary mechanical responses resulting from different routes of MCh administration. Our results indicate disparate physiological responses. Intravenous MCh delivery induced a higher maximum lung resistance than i.h. MCh in OVA-treated BALB/c mice but not in C57BL/6 mice. Inhaled MCh delivery led to a significantly larger fall in lung compliance and a greater impact on peripheral airways than i.v. MCh in both strains. In conclusion, i.v. and i.h. MCh produced disparate effects in different murine strains and variant responses in inflamed airways and healthy controls. The two methods of MCh delivery have important advantages but also certain limitations with regard to measuring airway reactivity in a murine model of allergic asthma.


Subject(s)
Airway Resistance/drug effects , Asthma/physiopathology , Bronchoconstrictor Agents/pharmacology , Lung Compliance/drug effects , Methacholine Chloride/pharmacology , Administration, Inhalation , Airway Resistance/physiology , Animals , Asthma/chemically induced , Disease Models, Animal , Female , Injections, Intravenous , Lung Compliance/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/pharmacology , Pneumonia/chemically induced , Pneumonia/physiopathology , Respiration, Artificial
7.
Article in English | MEDLINE | ID: mdl-19081714

ABSTRACT

F(2)-isoprostanes have been associated with various forms of oxidant stress. The levels of F(2)-isoprostanes in a murine asthma model were studied both in situ and in vivo and further investigated whether the formation of F(2)-isoprostanes was associated with increased ovalbumin (OVA)-induced airway inflammation after a 17-day (OVA-17) or a 24-day (OVA-24) protocol. Bronchial reactivity was assessed by using a ventilator (FlexiVent). OVA-treated animals had higher lung resistance and lung compliance compared to control groups (P<0.001). 8-Iso-PGF(2)(alpha) levels in bronchoalveolar lavage (BAL) and 8-iso-PGF(2)(alpha) immunoreactivity in lung tissue were analyzed. OVA-17 mice showed a 2.5-fold increased level of 8-iso-PGF(2)(alpha) in BAL compared to PBS-17 mice (P=0.023). Lung tissue from OVA-24 mice had more intense 8-iso-PGF(2)(alpha) staining compared to OVA-17 mice. This study showed an accumulation of F(2)-isoprostanes in acute airway inflammation and a markedly increased tissue damage caused by oxidative stress in an ongoing inflammation.


Subject(s)
Allergens/immunology , Asthma/immunology , Bronchial Hyperreactivity/immunology , F2-Isoprostanes/immunology , Inflammation/immunology , Oxidative Stress/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Ovalbumin/immunology
8.
Respir Res ; 9: 23, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-18307760

ABSTRACT

BACKGROUND: Deep inspirations (DI) have bronchodilatory and bronchoprotective effects in healthy human subjects, but these effects appear to be absent in asthmatic lungs. We have characterized the effects of DI on lung mechanics during mechanical ventilation in healthy mice and in a murine model of acute and chronic airway inflammation. METHODS: Balb/c mice were sensitized to ovalbumin (OVA) and exposed to nebulized OVA for 1 week or 12 weeks. Control mice were challenged with PBS. Mice were randomly selected to receive DI, which were given twice during the minute before assessment of lung mechanics. RESULTS: DI protected against bronchoconstriction of central airways in healthy mice and in mice with acute airway inflammation, but not when OVA-induced chronic inflammation was present. DI reduced lung resistance induced by methacholine from 3.8 +/- 0.3 to 2.8 +/- 0.1 cmH2O.s.mL-1 in healthy mice and 5.1 +/- 0.3 to 3.5 +/- 0.3 cmH2O.s.mL-1 in acute airway inflammation (both P < 0.001). In healthy mice, DI reduced the maximum decrease in lung compliance from 15.9 +/- 1.5% to 5.6 +/- 0.6% (P < 0.0001). This protective effect was even more pronounced in mice with chronic inflammation where DI attenuated maximum decrease in compliance from 44.1 +/- 6.6% to 14.3 +/- 1.3% (P < 0.001). DI largely prevented increased peripheral tissue damping (G) and tissue elastance (H) in both healthy (G and H both P < 0.0001) and chronic allergen-treated animals (G and H both P < 0.0001). CONCLUSION: We have tested a mouse model of potential value for defining mechanisms and sites of action of DI in healthy and asthmatic human subjects. Our current results point to potent protective effects of DI on peripheral parts of chronically inflamed murine lungs and that the presence of DI may blunt airway hyperreactivity.


Subject(s)
Bronchitis/physiopathology , Immunization , Inhalation , Ovalbumin/immunology , Respiratory Mechanics , Respiratory System/physiopathology , Acute Disease , Airway Resistance , Animals , Bronchitis/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoconstriction , Bronchoconstrictor Agents/pharmacology , Chronic Disease , Elasticity , Lung/drug effects , Lung/physiopathology , Lung Compliance , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C
9.
Nitric Oxide ; 16(4): 419-24, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17459737

ABSTRACT

BACKGROUND: We have previously shown that the protective effect of inhaled nitric oxide (iNO) against methacholine-induced bronchoconstriction is negated in airways subjected to hyperosmotic stress. In this study, hypothesizing that the impaired efficiency of iNO was caused by release of reactive oxygen radicals, we examined the effect of the radical scavenging enzyme superoxide dismutase (SOD). METHODS: Hemodynamic and respiratory measurements were performed on anesthetized rabbits after (1) inhalation of methacholine (MCh), (2) iNO (80ppm), followed by MCh, (3) inhalation of hypertonic saline (HS), followed by iNO and MCh and (4) pre-treatment with inhalation of SOD, followed by HS, iNO and MCh. We analyzed plasma for a marker of oxidative stress, 8-iso-prostaglandin (PG)F(2alpha) and for a marker of activation of COX-mediated inflammatory cascades, PGF(2alpha) metabolite. RESULTS: Pre-treatment with SOD restored the bronchoprotective response to iNO in hyperosmotic airways. No direct effect was seen by SOD treatment on levels of 8-iso-PGF(2alpha), but this marker of oxidative stress correlated positively with increased bronchoconstriction. Hyperosmotic challenge elevated levels of PGF(2alpha) metabolite, and pre-treatment with SOD protected against this activation of the inflammatory cascade. CONCLUSION: SOD pre-treatment restores the relaxant effects of iNO in hyperosmotically challenged airways by attenuating oxidative stress and activation of COX-mediated inflammatory cascades.


Subject(s)
Bronchoconstriction/drug effects , Bronchodilator Agents/pharmacology , Nitric Oxide/pharmacology , Superoxide Dismutase/pharmacology , Administration, Inhalation , Animals , Biomarkers/blood , Dinoprost/analogs & derivatives , Dinoprost/blood , Female , Methacholine Chloride/pharmacology , Oxidative Stress/drug effects , Rabbits , Signal Transduction/drug effects , Superoxide Dismutase/blood
10.
J Pharmacol Exp Ther ; 315(1): 458-65, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16024733

ABSTRACT

Nitric oxide (NO) in exhaled air is a biomarker of airway inflammation. However, the role of NO in the peripheral lung is not known. The aim of this study was to determine the role of endogenous NO in antigen-induced contractions of ovalbumin (OVA)-sensitized guinea pig lung parenchyma (GPLP). The contraction in this in vitro model of the peripheral lung closely resembles the corresponding response in human airways. Cumulatively increasing concentrations (10-10,000 microg/l) of OVA induced concentration-dependent contractions of the GPLP that were enhanced by the NO synthase (NOS) inhibitors N(omega)-nitro-L-arginine (L-NOARG; 100 microM), N(omega)-monomethyl-L-arginine (100 microM), N(omega)-nitro-L-arginine methyl ester (100 microM), and N-(3-(aminomethyl)benzyl)acetamidine (1400W; 1 microM). The enhancement induced by L-NOARG was reversed by coadministration with the 5-lipoxygenase inhibitor (R)-2-[4-(quinolin-2-yl-methoxy)phenyl]-2-cyclopentyl acetic acid (BAY x1005; 3 microM), whereas coadministration of L-NOARG with the cyclooxygenase inhibitor indomethacin (10 microM) did not change the effect of L-NOARG alone. L-NOARG (100 microM) did not affect the cumulative concentration-response relations for either leukotriene (LT) D4 (0.1-100 nM) or histamine (1-30 microM). The NO donor NONOate (0.001-100 microM) was ineffective in GPLP but potently relaxed precontracted guinea pig pulmonary artery. Furthermore, L-NOARG enhanced the release of LTE4 and decreased the release of prostaglandin E2 induced by OVA. In conclusion, endogenous NO exerts an inhibitory effect on antigen-induced contractions in the peripheral lung. The action of NO apparently involves inhibition of the release of mediators rather than direct relaxation of airway smooth muscle. The findings support the belief that endogenous NO has a protective anti-inflammatory effect in the airways.


Subject(s)
Cysteine/metabolism , Enzyme Inhibitors/pharmacology , Leukotrienes/metabolism , Lung/immunology , Muscle Contraction , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/physiology , Ovalbumin/immunology , Animals , Arachidonate 5-Lipoxygenase/physiology , Dinoprostone/pharmacology , Guinea Pigs , In Vitro Techniques , Lung/drug effects , Lung/physiology , Male , Nitroarginine/pharmacology
11.
Respir Physiol Neurobiol ; 146(2-3): 239-46, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15766912

ABSTRACT

Increased airway surface osmolarity has been shown to abolish the airway relaxant effects of inhaled nitric oxide. We have investigated the effects of increased airway surface osmolarity on airway relaxation induced by nitric oxide. The physiological responses, obtained by a guinea pig tracheal perfusion method, were compared to the ion content of the tracheal tissues, and the effects of amiloride or furosemide were studied. Hyperosmolarity decreased the ability of sodium nitroprusside (SNP) to relax carbachol-constricted trachea. Light microscopy showed shrinkage of the epithelial cells and X-ray microanalysis showed increased epithelial ion content under conditions of intraluminal hyperosmolarity, suggesting dehydration of the epithelium. Amiloride treatment reduced the increase in epithelial ion content but had no effect on shrinkage or SNP-induced relaxation. Furosemide had no effect on the altered ion content, shrinkage, or on SNP-induced relaxation. In conclusion, neither amiloride nor furosemide can counteract the shrinkage of the airway wall induced by increased osmolarity in the lumen of guinea pig trachea in vitro, nor can they affect the reduction in relaxing response to the NO-donor SNP.


Subject(s)
Endothelium-Dependent Relaxing Factors/pharmacology , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Nitric Oxide/pharmacology , Osmolar Concentration , Trachea/drug effects , Amiloride/pharmacology , Analysis of Variance , Animals , Connective Tissue/drug effects , Connective Tissue/physiology , Drug Interactions , Epithelium/drug effects , Epithelium/physiology , Furosemide , Guinea Pigs , In Vitro Techniques , Male , Muscle Relaxation/physiology , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Statistics, Nonparametric , Trachea/cytology , Trachea/physiology , X-Rays
12.
J Appl Physiol (1985) ; 97(1): 249-59, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15020581

ABSTRACT

Individuals with asthma have increased levels of nitric oxide in their exhaled air. To explore its role, we have developed a regulatable transgenic mouse capable of overexpressing inducible nitric oxide synthase in a lung-specific fashion. The CC10-rtTA-NOS-2 mouse contains two transgenes, a reverse tetracycline transactivator under the control of the Clara cell protein promoter and the mouse nitric oxide synthase-2 (NOS-2) coding region under control of a tetracycline operator. Addition of doxycycline to the drinking water of CC10-rtTA-NOS-2 mice causes an increase in nitric oxide synthase-2 that is largely confined to the airway epithelium. The fraction of expired nitric oxide increases over the first 24 h from approximately 10 parts per billion to a plateau of approximately 20 parts per billion. There were no obvious differences between CC10-rtTA-NOS-2 mice, with or without doxycycline, and wild-type mice in lung histology, bronchoalveolar protein, total cell count, or count differentials. However, airway resistance was lower in CC10-rtTA-NOS-2 mice with doxycycline than in CC10-rtTA-NOS-2 mice without doxycycline or wild-type mice with doxycycline. Moreover, doxycycline-treated CC10-rtTA-NOS-2 mice were hyporesponsive to methacholine compared with other groups. These data suggest that increased nitric oxide in the airways has no proinflammatory effects per se and may have beneficial effects on pulmonary function.


Subject(s)
Airway Resistance/genetics , Airway Resistance/physiology , Lung/enzymology , Lung/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Asthma/enzymology , Asthma/metabolism , Blotting, Northern , Blotting, Western , Bronchoalveolar Lavage Fluid/cytology , Bronchodilator Agents/pharmacology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Doxycycline/metabolism , Immunohistochemistry , Methacholine Chloride/pharmacology , Mice , Mice, Transgenic , Nitric Oxide/metabolism , Nitric Oxide/physiology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Respiratory Mechanics/genetics , Respiratory Mechanics/physiology , Respiratory Mucosa/metabolism , Respiratory Mucosa/physiology , Reverse Transcriptase Polymerase Chain Reaction , Tetracycline/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transgenes , Uteroglobin/genetics
13.
Am J Physiol Lung Cell Mol Physiol ; 286(4): L817-25, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15003938

ABSTRACT

Platelet-derived growth factors (PDGF) may contribute to the activation and growth of smooth muscle that is characteristic of airway remodeling in asthmatic patients. Early growth response 1 (EGR-1) is a transcription factor that is induced in several cell types by PDGF and may mediate some of the effects of PDGF. We show that human airway smooth muscle cells in cell culture express EGR-1 1 h after addition of PDGF. Analysis of the EGR-1 promoter indicates that a serum response element located between 663 and 654 bp 5' to the ATG start site is essential for this induction. Serum response factor, E26 transcription factor-like protein 1, and serum protein 1 bind to this region. PDGF causes phosphorylation of ERK1/2 and is temporally associated with E26 transcription factor-like protein 1 phosphorylation. Finally, the specific ERK1/2 inhibitor U-0126 abolishes PDGF-induced expression of EGR-1 in these cells. On the basis of these data, we speculated that EGR-1 would be increased in airway smooth muscle of asthmatic patients compared with nonasthmatic controls. Using immunohistochemistry, we found that EGR-1 protein was expressed in airway smooth muscle cells and epithelial cells of asthmatic patients and nonasthmatic controls; however, there was no significant difference in the intensity of staining between groups. EGR-1 was similarly expressed in the lungs of mice with and without ovalbumin-induced airway inflammation; however, there was no difference between groups by immunohistochemistry and quantitative PCR. Although EGR-1 is induced by PDGF in human airway smooth muscle cells in cell culture, the role of EGR-1 in airway remodeling and asthma remains to be established.


Subject(s)
DNA-Binding Proteins/genetics , Immediate-Early Proteins , Lung/cytology , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Platelet-Derived Growth Factor/pharmacology , Transcription Factors/genetics , 3T3 Cells , Animals , Asthma/physiopathology , DNA-Binding Proteins/metabolism , Early Growth Response Protein 1 , Genes, Reporter , Humans , MAP Kinase Signaling System/physiology , Mice , Muscle, Smooth/drug effects , Ovalbumin , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , RNA, Messenger/analysis , Serum Response Factor/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , ets-Domain Protein Elk-1
14.
Am J Physiol Lung Cell Mol Physiol ; 286(6): L1302-10, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14966082

ABSTRACT

During infection, inflammation is essential for host defense, but it can injure tissues and compromise organ function. TNF-alpha and IL-1 (alpha and beta) are early response cytokines that facilitate inflammation. To determine the roles of these cytokines with overlapping functions, we generated mice deficient in all of the three receptors mediating their effects (TNFR1, TNFR2, and IL-1RI). During Escherichia coli pneumonia, receptor deficiency decreased neutrophil recruitment and edema accumulation to half of the levels observed in wild-type mice. Thus these receptors contributed to maximal responses, but substantial inflammation progressed independently of them. Receptor deficiency compromised antibacterial efficacy for some infectious doses. Decreased ventilation during E. coli pneumonia was not affected by receptor deficiency. However, the loss of lung compliance during pneumonia was substantially attenuated by receptor deficiency. Thus during E. coli pneumonia in mice, the lack of signaling from TNF-alpha and IL-1 decreases inflammation and preserves lung compliance.


Subject(s)
Antineoplastic Agents/immunology , Escherichia coli Infections/immunology , Interleukin-1/immunology , Pneumonia, Bacterial/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antigens, CD/genetics , Antineoplastic Agents/pharmacology , Escherichia coli Infections/metabolism , Escherichia coli Infections/physiopathology , Female , Interleukin-1/pharmacology , Lung Compliance/immunology , Male , Mice , Mice, Mutant Strains , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/physiopathology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1 Type I , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Respiratory Mechanics/immunology , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/pharmacology
15.
Am J Respir Cell Mol Biol ; 26(4): 475-83, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11919084

ABSTRACT

5-lipoxygenase (ALOX5), an enzyme essential for the formation of all leukotrienes, is highly regulated at multiple levels, including gene transcription. The human ALOX5 promoter sequence has been cloned and is well characterized. Several important cis-acting elements have been identified including a G+C-rich sequence approximately 145-179 base pairs (bp) upstream from the ATG start codon. This region contains consensus-binding sites for the transcription factor serum protein 1, a zinc-finger transcription factor (SP1) and early growth-response protein 1, a zinc-finger transcription factor (EGR-1) and is unique in that functionally significant polymorphisms alter these sequences. To further understand the significance of these polymorphisms and other regulatory sequences in the promoter we cloned approximately 2,000 bp of the mouse promoter sequence from a 129/SvJ BAC library for direct comparison with the human gene. Like the human promoter, the mouse Alox5 promoter lacks a TATA box and has multiple start sites. The first 292 bp immediately upstream of the translational start site function as a core promoter that is capable of mediating high basal transcription in RAW cells but not 3T3 cells. There are vast differences in the distribution of consensus cis elements between human and mouse genes; however, three areas of strong homology exist and they contain consensus-binding sites for the SP1, GATA, GGAGA, and ETS family of transcription factors. We show that Sp1/Sp3 is essential for constitutive promoter-reporter activity.


Subject(s)
Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Promoter Regions, Genetic , 3T3 Cells , Animals , Base Sequence , Binding Sites , Cells, Cultured , Cloning, Molecular , DNA-Binding Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Macrophages/metabolism , Mice , Molecular Sequence Data , Organ Specificity , Peptides/metabolism , Sequence Deletion , Sequence Homology, Nucleic Acid , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor , Transcription Factors/metabolism , Transcription Initiation Site
SELECTION OF CITATIONS
SEARCH DETAIL
...