Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Eur J Biochem ; 268(23): 6083-96, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733002

ABSTRACT

The siglecs (sialic acid-binding Ig-like lectins) are a distinct subset of the Ig superfamily with adhesion-molecule-like structure. We describe here a novel member of the siglec protein family that shares a similar structure including five Ig-like domains, a transmembrane domain, and a cytoplasmic tail containing two ITIM-signaling motifs. Siglec-10 was identified through database mining of an asthmatic eosinophil EST library. Using the Stanford G3 radiation hybrid panel we were able to localize the genomic sequence of siglec-10 within the cluster of genes on chromosome 19q13.3-4 that encode other siglec family members. We have demonstrated that siglec-10 is an immune system-restricted membrane-bound protein that is highly expressed in peripheral blood leukocytes as demonstrated by Northern, RT-PCR and flow cytometry. Binding assays determined that the extracellular domain of siglec-10 was capable of binding to peripheral blood leukocytes. The cytoplasmic tail of siglec-10 contains four tyrosines, two of which are embedded in ITIM-signaling motifs (Y597 and Y667) and are likely involved in intracellular signaling. The ability of tyrosine kinases to phosphorylate the cytoplasmic tyrosines was evaluated by kinase assay using wild-type siglec-10 cytoplasmic domain and Y-->F mutants. The majority of the phosphorylation could be attributed to Y597 andY667. Further experiments with cell extracts suggest that SHP-1 interacts with Y667 and SHP-2 interacts with Y667 in addition to another tyrosine. This is very similar to CD33, which also binds the phosphatases SHP-1 and SHP-2, therefore siglec-10, as CD33, may be characterized as an inhibitory receptor.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Immune System/metabolism , Lectins/genetics , Lectins/metabolism , Receptors, Cell Surface , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , Binding Sites/genetics , Cell Line , Chromosome Mapping , DNA/genetics , Gene Expression , Humans , Intracellular Signaling Peptides and Proteins , Lectins/chemistry , Molecular Sequence Data , Multigene Family , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Sialic Acid Binding Ig-like Lectin 3 , Signal Transduction
2.
J Appl Physiol (1985) ; 90(3): 1111-7, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181627

ABSTRACT

The objective of the study was to develop a scintigraphic method for measurement of airway mucociliary clearance in small laboratory rodents such as the mouse. Previous investigations have characterized the secretory cell types present in the mouse airway, but analysis of the mucus transport system has been limited to in vitro examination of tissue explants or invasive in vivo measures of a single airway, the trachea. Three methods were used to deposit insoluble, radioisotopic colloidal particles: oropharyngeal aspiration, intratracheal instillation, and nose-only aerosol inhalation. The initial distribution of particles within the lower respiratory tract was visualized by gamma-camera, and clearance of particles was followed intermittently over 6 h and at the conclusion, 24 h postdelivery. Subsets of mice underwent lavage for evidence of tissue inflammation, and others were restudied for reproducibility of the methods. The aspiration and instillation methods of delivery led to greater distributions of deposited activity within the lungs, i.e., approximately 60--80% of the total respiratory tract radioactivity, whereas the nose-only aerosol technique attained a distribution of 32% to the lungs. However, the aerosol technique maximized the fraction of particles that cleared the airway over a 24-h period, i.e, deposited onto airway epithelial surfaces and cleared by mucociliary function such that lung retention at 24 h averaged 57% for delivery by aerosol inhalation and > or =80% for the aspiration or intratracheal instillation techniques. Particle delivery methods did not cause lung inflammation/injury with use of inflammatory cells and chemoattractant cytokines as criteria. Scintigraphy can discern particle deposition and clearance from the lower respiratory tract in the mouse, is noninvasive and reproducible, and includes the capability for restudy and lung lavage when time course or chronic treatments are being considered.


Subject(s)
Mucociliary Clearance/physiology , Respiratory Mucosa/physiology , Respiratory Physiological Phenomena , Technetium Tc 99m Sulfur Colloid/pharmacokinetics , Animals , Animals, Laboratory , Bronchoalveolar Lavage Fluid/cytology , Gamma Cameras , Half-Life , Leukocytes/cytology , Lung/diagnostic imaging , Lung/physiology , Male , Mice , Mice, Inbred A , Oropharynx/diagnostic imaging , Oropharynx/physiology , Radionuclide Imaging/methods , Radiopharmaceuticals/pharmacokinetics , Respiratory Mucosa/diagnostic imaging , Time Factors , Trachea/diagnostic imaging , Trachea/physiology
3.
J Appl Physiol (1985) ; 90(2): 713-23, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160073

ABSTRACT

Airways inflammation and epithelial injury induced by chronic ozone (O(3)) in genetically mast cell-deficient mice (Kit(W)/Kit(W-v)) were compared with those in mast cell-sufficient mice (+/+) and Kit(W)/Kit(W-v) mice repleted of mast cells (Kit(W)/Kit(W-v)-BMT). Mice were exposed to 0.26 ppm O(3) 8 h/day, 5 days/wk, for 1-90 days. Background was 0.06 ppm O(3). Age-matched mice were exposed to filtered air for O(3) controls. Reversibility of lesions was evaluated 35 days after exposure. Compared with Kit(W)/Kit(W-v), O(3) caused greater increases in lavageable macrophages, epithelial cells, and polymorphonuclear leukocytes in +/+ and Kit(W)/Kit(W-v)-BMT mice. O(3) also caused lung hyperpermeability, but the genotypic groups were not different. Cells and permeability returned to air control levels after O(3). O(3) induced lung cell proliferation only in +/+ and Kit(W)/Kit(W-v)-BMT mice; proliferation remained elevated or increased in +/+ and Kit(W)/Kit(W-v)-BMT mice after O(3). Greater O(3)-induced cell proliferation was found in nasal epithelium of +/+ and Kit(W)/Kit(W-v)-BMT mice compared with Kit(W)/Kit(W-v) mice. Results are consistent with the hypothesis that mast cells affect airway responses induced by chronic O(3) exposure.


Subject(s)
Inflammation/immunology , Mast Cells/immunology , Ozone/adverse effects , Respiratory Mucosa/immunology , Air , Animals , Blood Cell Count , Bronchi/cytology , Bronchi/immunology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Cell Division , Epithelial Cells/cytology , Inflammation/etiology , Leukocyte Count , Macrophages/cytology , Male , Mast Cells/cytology , Mice , Mice, Mutant Strains , Nasal Mucosa/cytology , Neutrophils/cytology , Respiratory Mucosa/cytology , Trachea/cytology , Trachea/immunology
4.
Toxicol Appl Pharmacol ; 162(2): 86-92, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10637131

ABSTRACT

Human and animal exposure to particulate air pollution is correlated with airway mucus hypersecretion and increased susceptibility to infection. Seeking clues to the mechanisms underlying this pathology, we examined the effect of the particulate air pollutant residual oil fly ash (ROFA) on production of the major component of mucus, mucin, and the major antibacterial protein of the respiratory tract, lysozyme. We found that following in vitro exposure to ROFA, epithelial cells showed an increase in mucin (MUC5AC) and lysozyme (LYS) steady state mRNA. This upregulation was controlled at least partly at the level of transcription as shown by reporter assays. Experiments testing the ability of the major components of ROFA to mimic these effects showed that vanadium, a metal making up 18.8% by weight, accounted for the bulk of the response. A screen of signaling inhibitors showed that MUC5AC and LYS induction by ROFA are mediated by dissimilar signaling pathways, both of which are, however, phosphotyrosine dependent. Recognizing that the ROFA constituent vanadium is a potent tyrosine phosphatase inhibitor and that mucin induction by pathogens is phophotyrosine dependent, we suggest that vanadium-containing air pollutants trigger disease-like conditions by unmasking phosphorylation-dependent pathogen resistance pathways.


Subject(s)
Air Pollutants/toxicity , Fuel Oils/toxicity , Lung/drug effects , Lung/metabolism , Mucins/biosynthesis , Vanadium/toxicity , Bronchi/cytology , Bronchi/drug effects , Cell Line , Cytoplasm/drug effects , Cytoplasm/metabolism , Cytoplasm/physiology , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Humans , Lung Neoplasms , Particle Size , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Tumor Cells, Cultured
5.
Am J Respir Crit Care Med ; 160(5 Pt 2): S44-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10556169

ABSTRACT

Mucin production is an evolutionarily ancient defense mechanism that is retained in mammals and operates at all mucosal surfaces to protect the host against pathogens and irritants. As in lower organisms, the mammalian mucosa (epithelium) produces mucin in response to diverse insults. Our studies aim to understand the intracellular signaling and gene regulation mechanisms mediating mucin production in response to clinically important insults. To date, we find that the signaling pathway triggered by each type of insult is distinct. Relatively common, however, is the involvement of the protein tyrosine kinase c-Src, the MAP kinase kinase MEK 1/2, and the transcription factor NF-kappaB. Basbaum C, Lemjabbar H, Longphre M, Li D, Gensch E, McNamara N. Control of mucin transcription by diverse injury-induced signaling pathways.


Subject(s)
Lung Diseases, Obstructive/genetics , Mucins/genetics , Signal Transduction/genetics , Transcription, Genetic/genetics , Animals , CSK Tyrosine-Protein Kinase , Gene Expression Regulation/physiology , Humans , Lung Diseases, Obstructive/immunology , MAP Kinase Signaling System/physiology , NF-kappa B/physiology , Protein-Tyrosine Kinases/physiology , Respiratory Mucosa/immunology , src-Family Kinases
6.
J Clin Invest ; 104(10): 1375-82, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10562299

ABSTRACT

A hallmark of asthma is mucin overproduction, a condition that contributes to airway obstruction. The events responsible for mucin overproduction are not known but are thought to be associated with mediators of chronic inflammation. Others have shown that T-helper 2 (Th2) lymphocytes are required for mucous cell metaplasia, which then leads to mucin overproduction in animal models of allergy. We hypothesized that Th2 cell mediators are present in asthmatic airway fluid and directly stimulate mucin synthesis in airway epithelial cells. Results in cultured airway epithelial cells showed that samples of asthmatic fluid stimulated mucin (MUC5AC) synthesis severalfold more potently than non-asthmatic fluid. Consistent with this, lavage fluid from the airways of allergen-challenged dogs stimulated mucin synthesis severalfold more potently than that from non-allergen-challenged dogs. Fractionation of dog samples revealed 2 active fractions at <10 kDa and 30-100 kDa. Th2 cytokines in these molecular weight ranges are IL-9 (36 kDa), IL-5 (56 kDa), and IL-13 (10 kDa). Antibody blockade of ligand-receptor interaction for IL-9 (but not IL-5 or IL-13) inhibited mucin stimulation by dog airway fluid. Furthermore, recombinant IL-9, but not IL-5 or IL-13, stimulated mucin synthesis. These results indicate that IL-9 may account for as much as 50-60% of the mucin-stimulating activity of lung fluids in allergic airway disease.


Subject(s)
Allergens , Asthma/physiopathology , Interleukin-9/physiology , Mucins/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Th2 Cells/immunology , Transcription, Genetic , Adult , Animals , Asthma/immunology , Asthma/metabolism , Asthma/pathology , Bronchi/cytology , Bronchi/pathology , Cells, Cultured , Cytokines/analysis , Dogs , Female , Gene Expression Regulation/drug effects , Humans , Interleukin-9/genetics , Interleukins/analysis , Interleukins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mucin 5AC , Mucins/biosynthesis , Receptors, Interleukin/analysis , Receptors, Interleukin/genetics , Receptors, Interleukin-9 , Recombinant Proteins/pharmacology , Respiratory Mucosa/pathology , Trachea/cytology , Trachea/pathology , Transcription, Genetic/drug effects , Tumor Cells, Cultured
7.
Res Rep Health Eff Inst ; (85): 1-30; discussion 31-6, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10349676

ABSTRACT

Acute and subacute exposure to ozone (O3) induces lung inflammation and hyperpermeability and causes epithelial injury of both upper (nasal) and lower airways. Mast cells are important regulatory cells in mice for each of these effects. Subacute and chronic O3 exposures cause epithelial injury and inflammation in terminal bronchioles and proximal alveoli. Little is known, however, about the mechanisms of injury. Because inflammatory processes may be linked to the pathogenesis of many airway diseases, it is critical to understand the underlying mechanisms that initiate and propagate these processes. We tested the hypothesis that mast cells mediate airway injury induced by chronic O3 exposure by comparing regional airway inflammation and epithelial injury as well as ventilatory responses in genetically mast cell-deficient mice (WBB6F1-KitW/KitW-v [KitW/KitW-v]) with those in (1) normal, mast cell-sufficient, congenic littermates (WBB6F1(-)+/+ [+/+]) and those in (2) KitW/KitW-v mice that were repleted with mast cells by bone marrow transplantation (BMT) from +/+ donors (KitW/KitW-v-BMT). Thus, three (different) groups of mice were used. The following experimental protocol was utilized to test this hypothesis. Animals from each treatment group (n = 4-6/group) were exposed to 0.26 parts per million (ppm) O3 8 hours/day and 5 days/week for durations of 1, 3, 14, 30, and 90 days. Between 8-hour exposures, mice were exposed continuously to 0.06 ppm O3. Age-matched mice were simultaneously exposed to filtered air (0.0 ppm O3) to serve as O3 controls. To evaluate reversibility of exposure-induced lesions, a set of mice from each genotypic group was exposed to air or O3 for 90 days and then placed in HEPA-filtered air for 35 days. After each period of exposure and after 35-day recovery, the nasal cavity and lungs of O3- and air-exposed mice from each group were evaluated for regional inflammation and permeability, epithelial proliferation, and ventilation pattern. Estimates of airway inflammation and hyperpermeability were obtained by analysis of cell differentials and total protein concentrations, respectively, in fluids obtained through use of bronchoalveolar lavage (BAL). Ozone exposure caused significantly greater increases in lung macrophages, epithelial cells, and polymorphonuclear leukocytes (PMNs) in mast cell-sufficient +/+ and KitW/KitW-v-BMT mice than in mast cell-deficient KitW/KitW-v mice. Comparable ozone exposure also elicited increases in lung lymphocytes and in total protein, but there were no significant differences in these two genotypic groups. Cell and total-protein responses in BAL fluid returned to control levels (that is, air exposure only) in all three groups of mice after a 35-day recovery period. The effects of O3 exposure on cell proliferation in the nose and lung were evaluated in the genotypic groups by counting the number of cells that incorporated bromodeoxyuridine (BrdU, a thymidine analog) into DNA. In the centriacinar region of the lung, DNA synthesis was increased significantly in O3-exposed +/+ and KitW/KitW-v-BMT mice, but not in KitW/KitW-v mice, compared with DNA synthesis in air controls. Epithelial proliferation remained significantly elevated or even increased in +/+ and KitW/KitW-v-BMT mice after O3 exposure. Nasal responses to O3 were also evaluated in these three genotypic groups of mice, and there were slight, although statistically significant, O3-exposure effects on the transitional epithelium. However, there were no differences among the groups up to an exposure of 90 days in duration. After a 35-day recovery period, epithelial cell proliferation in +/+ and KitW/KitW-v-BMT mice was greater than that in KitW/KitW-v mice. There were no significant exposure, genotype, or duration effects on baseline ventilation or responses to hypercapnic hypoxia in the three groups of mice exposed to air or O3. (ABSTRACT TRUNCATED)


Subject(s)
Air Pollutants/toxicity , Mast Cells/drug effects , Oxidants, Photochemical/toxicity , Ozone/toxicity , Respiratory System/drug effects , Analysis of Variance , Animals , Bronchoalveolar Lavage Fluid/cytology , Dose-Response Relationship, Drug , Inflammation/chemically induced , Male , Mice , Mice, Congenic , Respiratory Mechanics
8.
J Appl Physiol (1985) ; 86(1): 341-9, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9887149

ABSTRACT

Ozone (O3) exposure stimulates airway inflammation and epithelial sloughing in a number of species, including mice. Platelet-activating factor (PAF) is a lipid mediator released by activated mast cells, macrophages, and epithelial cells and causes pulmonary inflammation and hyperpermeability. We hypothesized that the activation of PAF receptors is central to the development of inflammation and epithelial injury induced by acute O3 exposure in mice. To test this hypothesis, O3-susceptible C57BL/6J mice were treated with a PAF-receptor antagonist, UK-74505, or vehicle either before or immediately after 3-h exposure to O3 (2 parts/million) or filtered air. Bronchoalveolar lavage (BAL) fluids were collected 6 and 24 h after exposure. Differential cell counts and protein content of the lavage were used as indicators of inflammation in the airways. O3-induced epithelial injury was assessed by light microscopy, and DNA synthesis in epithelium of terminal bronchioles was estimated by using a bromodeoxyuridine-labeling index. Intercellular adhesion molecule 1 (ICAM-1) expression was also examined in the lung by immunohistochemical localization. O3 caused significant increases in polymorphonuclear leukocytes and protein in the BAL fluid, increased pulmonary epithelial proliferation, and increased epithelial expression of ICAM-1 compared with air-exposed, vehicle-treated control mice. Relative to O3-exposed, vehicle-treated control mice, UK-74505 before exposure significantly (P < 0.05) decreased BAL protein, polymorphonuclear leukocytes, and epithelial cells. O3-induced inflammation was similarly attenuated in mice treated with UK-74505 after exposure. These experiments thus support the hypothesis that O3-induced airways inflammation and epithelial damage in mice are partially mediated by activation of PAF receptors, possibly through modulation of ICAM-1 expression.


Subject(s)
Epithelial Cells/physiology , Oxidants, Photochemical/toxicity , Ozone/toxicity , Platelet Activating Factor/pharmacology , Pneumonia/chemically induced , Pneumonia/pathology , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Division/drug effects , Epithelial Cells/drug effects , Lung/cytology , Lung/drug effects , Male , Mice , Mice, Inbred C57BL , Platelet Membrane Glycoproteins/antagonists & inhibitors
9.
Nat Genet ; 17(4): 475-8, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9398854

ABSTRACT

Exposures to the common air pollutant ozone (O3) cause decrements in pulmonary function and induce airway inflammation that is characterized by infiltration of polymorphonuclear neutrophils (PMNs; refs 1-4). Because of the impact that O3 may have on public health, it is critical to identify susceptibility factors. Highly reproducible, significant inter-individual variations in human pulmonary function responses to O3 support the hypothesis that genetic background is an important determinant. Initial analysis of PMN responses to O3 exposure in segregant populations derived from inflammation-prone (susceptible) C57BL/6J (B6) and inflammation-resistant C3H/HeJ (C3) inbred mice indicated that susceptibility was controlled by a locus we termed Inf2 (ref. 7). Subsequent analyses with recombinant inbred strains suggested that a more complex interaction of genes is involved. In this report, we identify a quantitative trait locus (QTL) for O3 susceptibility on chromosome 17. Candidate genes for the locus include Tnf, the gene encoding the pro-inflammatory cytokine tumour necrosis factor-alpha (Tnf). Antibody neutralization of the protein product of this putative candidate gene significantly protected against O3 injury in susceptible mice. These results strongly support linkage of O3 susceptibility to a QTL on chromosome 17 and Tnf as a candidate gene.


Subject(s)
Genetic Linkage , Lung/pathology , Ozone/toxicity , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/genetics , Animals , Antibodies/pharmacology , Crosses, Genetic , Disease Susceptibility , Female , Inflammation/chemically induced , Inflammation/genetics , Lung/drug effects , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Quantitative Trait, Heritable , Respiratory Hypersensitivity/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
10.
Toxicol Lett ; 88(1-3): 243-8, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8920744

ABSTRACT

A flow-past nose-only inhalation system was used for the co-exposure of mice to carbon black aerosols (CBA) and sulfur dioxide (SO2) at varying relative humidities (RH). The conversion of SO2 to sulfate (SO4(-2)) on the CBA, at a fixed aerosol concentration, was dependent on RH and SO2 concentration. The effect of the aerosol-gas mixture on alveolar macrophage (AM) phagocytosis was assessed three days following exposure for 4 h. Exposure to 10 mg/m3 CBA alone at low RH (10%) and high RH (85%), to 10 ppm SO2 alone at both RH, and to the mixture at low RH had no effect on AM phagocytosis. In contrast, AM phagocytosis was significantly suppressed following co-exposure at 85% RH, the only circumstance in which significant chemisorption of the gas by the aerosol and oxidation to SO4(-2) occurred. The results suggest that fine carbon particles can be an effective vector for the delivery of toxic amounts of SO4(-2) to the periphery of the lung.


Subject(s)
Carbon/toxicity , Macrophages, Alveolar/drug effects , Phagocytosis/drug effects , Sulfur Dioxide/toxicity , Administration, Inhalation , Aerosols/toxicity , Animals , Atmosphere Exposure Chambers , Bronchoalveolar Lavage Fluid/cytology , Carbon/administration & dosage , Cell Count/drug effects , Cell Survival/drug effects , Female , Macrophages, Alveolar/physiology , Mice , Receptors, Fc/drug effects , Receptors, Fc/physiology , Sulfates/analysis , Sulfur Dioxide/administration & dosage
11.
Am J Respir Cell Mol Biol ; 14(5): 461-9, 1996 May.
Article in English | MEDLINE | ID: mdl-8624251

ABSTRACT

We tested the hypothesis that mast cells contribute to platelet-activating factor (PAF)-induced airways hyperreactivity and hyperpermeability in mice. Airways reactivity to acetylcholine (ACh) and lung permeability to Evans blue (EB) dye were measured before and after PAF challenge in genetically mast cell-deficient (WBB6F1 W/Wv) and normal congenic (WBB6F1 +/+) mice, as well as mast cell-reconstituted (BMT W/Wv) mice. In addition, prostaglandin D2 (PGD2), a mast cell-specific mediator, was measured in the bronchoalveolar lavage (BAL) from +/+ and W/Wv mice to determine if lung mast cell activation was a consequence of PAF challenge. Genetically PAF-sensitive AKR/J mice were also treated with the mast cell stabilizer nedocromil prior to assessment of PAF effects on ACh reactivity. Intravenous PAF (10 micrograms/kg) induced a significant (P < 0.05) increase in airways reactivity to ACh (25 micrograms/kg) in both +/+ (371 +/- 52%) and W/Wv (122 +/- 24%) mice. There was a significantly greater increase in +/+ compared with W/Wv mice. PAF-induced hyperreactivity to ACh in BMT W/Wv mice (191 +/- 44%) was significantly (P < 0.05) greater than age-matched W/Wv mice (80 +/- 16%), but not significantly different from age-matched +/+ mice (153 +/- 44%). PAF (10 micrograms/kg) also significantly (P < 0.5) increased lung permeability in +/+ and W/Wv mice, but there was no significant difference between groups. BAL PGD2 increased significantly in +/+ mice following PAF challenge (559 +/- 24 ng/ml) compared with vehicle controls (152 +/- 8 pg/ml). There was no significant increase in BAL PGD2 from W/Wv mice. Nedocromil pretreatment significantly (P < 0.05) decreased PAF-induced hyperreactivity in AKR/J mice but not in W/Wv mice (P > 0.05). We conclude that mast cells contribute significantly to PAF-induced hyperreactivity but not hyperpermeability in mice.


Subject(s)
Bronchial Hyperreactivity/physiopathology , Mast Cells/physiology , Platelet Activating Factor/pharmacology , Acetylcholine/pharmacology , Analysis of Variance , Animals , Azepines/pharmacology , Blood Cell Count , Bone Marrow Transplantation/physiology , Bronchi/drug effects , Bronchi/physiology , Bronchi/physiopathology , Bronchoalveolar Lavage Fluid/chemistry , Evans Blue , Hematocrit , Lung/drug effects , Lung/physiology , Lung/physiopathology , Male , Mast Cells/drug effects , Mice , Mice, Inbred AKR , Mice, Mutant Strains , Nedocromil/pharmacology , Permeability , Platelet Activating Factor/antagonists & inhibitors , Prostaglandin D2/analysis , Species Specificity , Trachea/drug effects , Trachea/physiology , Trachea/physiopathology , Triazoles/pharmacology
12.
J Appl Physiol (1985) ; 80(4): 1322-30, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8926262

ABSTRACT

Ozone (O3) exposure produces inflammation in the airways of humans and animal models. However, the mechanism by which O3 affects these changes is uncertain. Mast cells are strategically located below the epithelium of the airways and are capable of releasing a number of proinflammatory mediators. We tested the hypothesis that mast cells contribute to inflammation, epithelial sloughing, and epithelial proliferation in the nasal and terminal bronchiolar murine airways after O3 exposure. Mast cell-sufficient (+/+), mast cell-deficient (W/Wv), and mast cell-repleted [bone marrow-transplanted (BMT) W/Wv] mice were exposed to 2 ppm O3 or filtered air for 3 h. Nasal and bronchoalveolar lavage fluids were collected 6 and 24 h after exposure. Differential cell counts and protein content of the lavage fluids were used as indicators of inflammation and permeability changes in the airways. O3-induced epithelial injury was assessed by light microscopy, and O3-induced DNA synthesis in airway epithelium was estimated by using a 5-bromo-2'-deoxyuridine-labeling index in the nasal and terminal bronchiolar epithelia. Relative to air control mice, O3 caused significant increases in inflammation, epithelial injury, and epithelial DNA synthesis in +/+ mice. There was no significant effect of O3 exposure on any measured parameter in the W/Wv mice. To further assess the role of mast cells in O3-induced epithelial damage, mast cells were restored in W/Wv mice by BMT from +/+ congeners. Relative to sham-transplanted W/Wv mice, O3 caused significant increases in epithelial damage and DNA synthesis as well as inflammatory indicators in BMT W/Wv mice. These observations are consistent with the hypothesis that mast cells significantly modulate the inflammatory and proliferative responses of the murine airways to O3.


Subject(s)
Bronchi/drug effects , Cell Division/physiology , Mast Cells/physiology , Nose/drug effects , Oxygen/pharmacology , Animals , Cell Count , Epithelium/metabolism , Lung/ultrastructure , Male , Mice , Mice, Inbred Strains
13.
Am J Respir Cell Mol Biol ; 13(5): 586-94, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7576695

ABSTRACT

Platelet-activating factor (PAF) is a proinflammatory mediator known to elicit changes in airway reactivity and vascular permeability, and it may also have a role in the development and progression of acute respiratory distress syndrome and asthma. We have developed a mouse model to test the hypothesis that these traits were controlled by a single gene and were mechanistically related. We further hypothesized that there was a relationship between PAF-induced hyperreactivity and baseline reactivity to acetylcholine (ACh). Among eight inbred strains of mice that exhibited significant interstrain variation in ACh reactivity, intravenous PAF induced 16 to 278% increases in reactivity to ACh (25 micrograms/kg). PAF also elicited 95 to 307% increases in lung permeability as measured by Evans blue extravasation. Both reactivity and permeability changes induced by PAF were blocked by a PAF receptor antagonist (L-659,989). Strain distribution patterns for baseline reactivity to ACh and PAF-induced hyperreactivity and lung permeability were not significantly concordant, and suggest that the variables were not interdependent. Progeny derived from AKR/J (PAF hyperresponsive) and C3H/HeJ (PAF hyporesponsive) mice were characterized for their PAF responsiveness as determined by PAF-induced hyperreactivity and hyperpermeability. The ratios of hyperresponsive and hyporesponsive phenotypes in outcross progeny were compared to those predicted for Mendelian inheritance and assessed for relatedness by chi 2 and cosegregation analyses. Results suggested that PAF-induced hyperreactivity was controlled by a single gene, but PAF-induced hyperpermeability was controlled by a more complicated interaction of factors.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Airway Resistance/drug effects , Capillary Permeability/drug effects , Platelet Activating Factor/pharmacology , Pulmonary Circulation/drug effects , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Acetylcholine/pharmacology , Animals , Female , Furans/pharmacology , Genes , Male , Mice , Mice, Inbred AKR , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Platelet Membrane Glycoproteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...