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










Publication year range
1.
J Immunol ; 182(12): 8037-46, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494329

ABSTRACT

Adenosine is a signaling nucleoside that is generated in response to cellular injury and orchestrates the balance between tissue protection and the progression to pathological tissue remodeling. Adenosine deaminase (ADA)-deficient mice develop progressive airway inflammation and remodeling in association with adenosine elevations, suggesting that adenosine can promote features of chronic lung disease. Furthermore, pharmacological studies in ADA-deficient mice demonstrate that A(2B)R antagonism can attenuate features of chronic lung disease, implicating this receptor in the progression of chronic lung disease. This study examines the contribution of A(2B)R signaling in this model by generating ADA/A(2B)R double-knockout mice. Our hypothesis was that genetic removal of the A(2B)R from ADA-deficient mice would lead to diminished pulmonary inflammation and damage. Unexpectedly, ADA/A(2B)R double-knockout mice exhibited enhanced pulmonary inflammation and airway destruction. Marked loss of pulmonary barrier function and excessive airway neutrophilia are thought to contribute to the enhanced tissue damage observed. These findings support an important protective role for A(2B)R signaling during acute stages of lung disease.


Subject(s)
Adenosine Deaminase/deficiency , Adenosine Deaminase/metabolism , Pneumonia/immunology , Pneumonia/metabolism , Receptor, Adenosine A2B/metabolism , Adenosine Deaminase/genetics , Animals , Cell Adhesion Molecules/metabolism , Collagen/biosynthesis , Cytokines/biosynthesis , Cytokines/immunology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/genetics , Pneumonia/pathology , Receptor, Adenosine A2B/deficiency , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/immunology
2.
Am J Respir Cell Mol Biol ; 38(6): 629-38, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18096867

ABSTRACT

Nontypeable Haemophilus influenzae (NTHi) commonly colonizes the lower airways of patients with chronic obstructive pulmonary disease (COPD). Whether it contributes to COPD progression is unknown. Here, we determined which aspects of the COPD phenotype can be induced by repetitive exposure to NTHi products. Mice were exposed weekly to an aerosolized NTHi lysate, and inflammation was evaluated by measurement of cells and cytokines in bronchoalveolar lavage fluid (BALF) and immunohistochemical staining; structural changes were evaluated histochemically by periodic acid fluorescent Schiff's reagent, Masson's trichrome, and Picrosirius red staining; mucin gene expression was measured by quantitative RT-PCR; and the role of TNF-alpha was examined by transgenic airway overexpression and use of an inhibitory antibody. NTHi lysate induced rapid activation of NF-kappaB in airway cells and increases of inflammatory cytokines and neutrophils in BALF. Repetitive exposure induced infiltration of macrophages, CD8+ T cells, and B cells around airways and blood vessels, and collagen deposition in airway and alveolar walls, but airway mucin staining and gel-forming mucin transcripts were not increased. Transgenic overexpression of TNF-alpha caused BALF neutrophilia and inflammatory cell infiltration around airways, but not fibrosis, and TNF-alpha neutralization did not reduce BALF neutrophilia in response to NTHi lysate. In conclusion, NTHi products elicit airway inflammation in mice with a cellular and cytokine profile similar to that in COPD, and cause airway wall fibrosis but not mucous metaplasia. TNF-alpha is neither required for inflammatory cell recruitment nor sufficient for airway fibrosis. Colonization by NTHi may contribute to the pathogenesis of small airways disease in patients with COPD.


Subject(s)
Haemophilus influenzae , Inflammation , Pulmonary Disease, Chronic Obstructive , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Female , Haemophilus influenzae/chemistry , Haemophilus influenzae/immunology , Humans , Inflammation/immunology , Inflammation/virology , Leukocytes/immunology , Lung/cytology , Lung/immunology , Lung/pathology , Metaplasia/metabolism , Metaplasia/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucins/genetics , Mucins/metabolism , NF-kappa B/metabolism , Phenotype , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Uteroglobin/genetics , Uteroglobin/metabolism
3.
Am J Respir Cell Mol Biol ; 37(3): 273-90, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17463395

ABSTRACT

Mucus hypersecretion contributes to morbidity and mortality in many obstructive lung diseases. Gel-forming mucins are the chief glycoprotein components of airway mucus, and elevated expression of these during mucous metaplasia precedes the hypersecretory phenotype. Five orthologous genes (MUC2, MUC5AC, MUC5B, MUC6, and MUC19) encode the mammalian gel-forming mucin family, and several have been implicated in asthma, cystic fibrosis, and chronic obstructive pulmonary disease pathologies. However, in the absence of a comprehensive analysis, their relative contributions remain unclear. Here, we assess the expression of the entire gel-forming mucin gene family in allergic mouse airways and show that Muc5ac is the predominant gel-forming mucin induced. We previously showed that the induction of mucous metaplasia in ovalbumin-sensitized and -challenged mouse lungs occurs within bronchial Clara cells. The temporal induction and localization of Muc5ac transcripts correlate with the induced expression and localization of mucin glycoproteins in bronchial airways. To better understand the tight regulation of Muc5ac expression, we analyzed all available 5'-flanking sequences of mammalian MUC5AC orthologs and identified evolutionarily conserved regions within domains proximal to the mRNA coding region. Analysis of luciferase reporter gene activity in a mouse transformed Clara cell line demonstrates that this region possesses strong promoter activity and harbors multiple conserved transcription factor-binding motifs. In particular, SMAD4 and HIF-1alpha bind to the promoter, and mutation of their recognition motifs abolishes promoter function. In conclusion, Muc5ac expression is the central event in antigen-induced mucous metaplasia, and phylogenetically conserved 5' noncoding domains control its regulation.


Subject(s)
Lung/pathology , Lung/physiopathology , Mucins/genetics , Mucins/physiology , 3T3 Cells , Amino Acid Sequence , Animals , Antigens/administration & dosage , Base Sequence , Cell Line , Conserved Sequence , DNA Primers/genetics , Female , Humans , Lung/immunology , Metaplasia , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mucin 5AC , Mucous Membrane/pathology , Mucous Membrane/physiopathology , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Up-Regulation
4.
Am J Respir Cell Mol Biol ; 35(5): 549-58, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16763221

ABSTRACT

Mucin hypersecretion is a prominent feature of obstructive airway diseases such as asthma. Clara cells conditionally produce mucin in response to inflammatory signals in a process termed mucous metaplasia. This can be followed by mucin secretion stimulated by various signaling molecules. The cellular and molecular mechanisms that regulate mucin production and secretion are not well understood. Adenosine is a signaling nucleoside that has been implicated in airway diseases in which mucus obstruction is prominent. Furthermore, the A(3) adenosine receptor (A(3)AR) is upregulated in mucin-producing goblet cells of the airway, thereby implicating it in processes involved in mucous cell biology. Here we use genetic approaches to investigate the contribution of A(3)AR signaling to mucus production and secretion in a mouse model of allergen-induced pulmonary disease. We found that the degree of mucin production in response to allergen is similar in wild-type and A(3)AR-deficient mice, and that overexpression of this receptor in Clara cells neither induces mucin production itself, nor enhances mucin production in response to allergen challenge. Collectively, these experiments demonstrate that the A(3)AR is neither necessary nor sufficient for mucous cell metaplasia. In contrast to the lack of effect on mucin production, agonist-induced mucin secretion was increased in goblet cells overexpressing the A(3)AR, and was absent in A(3)AR-deficient mice. Thus, the A(3)AR contributes to mucin secretion in allergen-induced metaplasia. Signaling through this receptor may contribute to mucus airway obstruction seen in pulmonary disorders in which adenosine levels are elevated.


Subject(s)
Bronchial Provocation Tests , Mucins/metabolism , Receptor, Adenosine A3/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Signal Transduction/physiology , Animals , Cell Shape , Metaplasia/immunology , Metaplasia/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Receptor, Adenosine A3/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/pathology
5.
J Clin Invest ; 116(5): 1274-83, 2006 May.
Article in English | MEDLINE | ID: mdl-16670768

ABSTRACT

To define the factors that control the tissue effects of IL-4, we compared the effects of Tg IL-4 in Balb/c and C57BL/6 mice. In the former, IL-4 caused modest eosinophilic inflammation and mild airway fibrosis and did not shorten survival. In C57BL/6 mice, IL-4 caused profound eosinophilic inflammation, airway fibrosis, emphysematous alveolar destruction, and premature death. These differences could not be accounted for by changes in Th2 or Th1 cytokines, receptor components, STAT6 activation, MMPs, or cathepsins. In contrast, in C57BL/6 mice, alveolar remodeling was associated with decreased levels of tissue inhibitors of metalloproteinase 2, -3, and -4 and alpha1-antitrypsin, and fibrosis was associated with increased levels of total and bioactive TGF-beta1. Impressive differences in adenosine metabolism were also appreciated, with increased tissue adenosine levels and A(1), A(2B), and A(3) adenosine receptor expression and decreased adenosine deaminase (ADA) activity in C57BL/6 animals. Treatment with ADA also reduced the inflammation, fibrosis, and emphysematous destruction and improved the survival of C57BL/6 Tg animals. These studies demonstrate that genetic influences control IL-4 effector pathways in the murine lung. They also demonstrate that IL-4 has different effects on adenosine metabolism in Balb/c and C57BL/6 mice and that these differences contribute to the different responses that IL-4 induces in these inbred animals.


Subject(s)
Adenosine/metabolism , Emphysema/pathology , Fibrosis/pathology , Interleukin-4/physiology , Animals , Female , Inflammation , Interleukin-4/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pulmonary Alveoli/metabolism , Rats , Species Specificity , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
6.
Arch Pathol Lab Med ; 130(4): 440-6, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16594735

ABSTRACT

CONTEXT: During an asthmatic episode, leukotriene C4 (LTC4) and interleukin 13 (IL-13) are released into the airways and are thought to be central mediators of the asthmatic response. However, little is known about how these molecules interact or affect each other's signaling pathway. OBJECTIVE: To determine if the LTC4 and IL-13 signaling pathways interact with each other's pathways. DESIGN: We examined airway responsiveness, cysteinyl LTs (Cys-LTs), and Cys-LT and IL-13 receptor transcript levels in wild-type mice and in mice that were deficient in gamma-glutamyl leukotrienase (an enzyme that converts LTC4 to LTD4), STAT6 (signal transducer and activator of transcription 6 [a critical molecule in IL-13 signaling]), and IL-4Ralpha (a subunit of the IL-13 receptor). RESULTS: Wild-type (C57BL/129SvEv) and gamma-glutamyl leukotrienase-deficient mice showed increased airway responsiveness after intranasal instillation of IL-13; similar results were observed after intranasal instillation of IL-13 or LTC4 in a second wild-type strain (BALB/c). Interleukin 13 treatment reduced levels of Cys-LTs in bronchoalveolar lavage fluid. This change was unaccompanied by changes in other arachidonic acid metabolites or in RNA transcript levels of enzymes associated with Cys-LT synthesis. Interleukin 13 treatment also increased transcript levels of the Cys-LT 1 and Cys-LT 2 receptors, while LTC4 increased transcript levels of the alpha1 chain of the IL-13 receptor. Furthermore, IL-4Ralpha-deficient mice had increased airway responsiveness to LTC4 but not to IL-13, whereas STAT6-deficient mice failed to respond to either agonist. CONCLUSIONS: These findings indicate that LTC4 and IL-13 are dependent on or signal through STAT6 to increase airway responsiveness and that both agonists regulate expression of each other's receptors.


Subject(s)
Airway Resistance/physiology , Asthma/metabolism , Disease Models, Animal , Interleukin-13/metabolism , Leukotriene C4/metabolism , Signal Transduction , Airway Resistance/drug effects , Animals , Asthma/genetics , Asthma/immunology , Bronchoalveolar Lavage Fluid/chemistry , Dipeptidases/deficiency , Dipeptidases/genetics , Humans , Interleukin-13/pharmacology , Interleukin-13 Receptor alpha1 Subunit , Leukotriene C4/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Receptors, Interleukin-13 , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/metabolism , Receptors, Leukotriene/genetics , Receptors, Leukotriene/metabolism , Recombinant Proteins/pharmacology , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/genetics
7.
Am J Physiol Lung Cell Mol Physiol ; 290(3): L579-87, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16258000

ABSTRACT

Adenosine, a signaling nucleoside, exhibits tissue-protective and tissue-destructive effects. Adenosine levels in tissues are controlled in part by the enzyme adenosine deaminase (ADA). ADA-deficient mice accumulate adenosine levels in multiple tissues, including the lung, where adenosine contributes to the development of pulmonary inflammation and chronic airway remodeling. The present study describes the development of pulmonary fibrosis in mice that have been genetically engineered to possess partial ADA enzyme activity and, thus, accumulate adenosine over a prolonged period of time. These partially ADA-deficient mice live for up to 5 mo and die from apparent respiratory distress. Detailed investigations of the lung histopathology of partially ADA-deficient mice revealed progressive pulmonary fibrosis marked by an increase in the number of pulmonary myofibroblasts and an increase in collagen deposition. In addition, in regions of the distal airways that did not exhibit fibrosis, an increase in the number of large foamy macrophages and a substantial enlargement of the alveolar air spaces suggest emphysemic changes. Furthermore, important proinflammatory and profibrotic signaling pathways, including IL-13 and transforming growth factor-beta1, were activated. Increases in tissue fibrosis were also seen in the liver and kidneys of these mice. These changes occurred in association with pronounced elevations of lung adenosine concentrations and alterations in lung adenosine receptor levels, supporting the hypothesis that elevation of endogenous adenosine is a proinflammatory and profibrotic signal in this model.


Subject(s)
Adenosine Deaminase/physiology , Adenosine/metabolism , Pulmonary Fibrosis/etiology , Respiratory System/metabolism , Adenosine Deaminase/genetics , Animals , Collagen/metabolism , Fibroblasts/metabolism , Homozygote , Interleukin-13/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Pulmonary Alveoli/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
8.
J Clin Invest ; 115(1): 35-43, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15630442

ABSTRACT

Adenosine is a signaling nucleoside that has been implicated in the regulation of asthma and chronic obstructive pulmonary disease. Adenosine signaling can serve both pro- and anti-inflammatory functions in tissues and cells. In this study we examined the contribution of A(1) adenosine receptor (A(1)AR) signaling to the pulmonary inflammation and injury seen in adenosine deaminase-deficient (ADA-deficient) mice, which exhibit elevated adenosine levels. Experiments revealed that transcript levels for the A(1)AR were elevated in the lungs of ADA-deficient mice, in which expression was localized predominantly to alveolar macrophages. Genetic removal of the A(1)AR from ADA-deficient mice resulted in enhanced pulmonary inflammation along with increased mucus metaplasia and alveolar destruction. These changes were associated with the exaggerated expression of the Th2 cytokines IL-4 and IL-13 in the lungs, together with increased expression of chemokines and matrix metalloproteinases. These findings demonstrate that the A(1)AR plays an anti-inflammatory and/or protective role in the pulmonary phenotype seen in ADA-deficient mice, which suggests that A(1)AR signaling may serve to regulate the severity of pulmonary inflammation and remodeling seen in chronic lung diseases by controlling the levels of important mediators of pulmonary inflammation and damage.


Subject(s)
Adenosine/metabolism , Lung Injury , Lung/metabolism , Receptor, Adenosine A1/metabolism , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Chemokines/genetics , Cytokines/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lung/pathology , Metaplasia/genetics , Metaplasia/metabolism , Metaplasia/pathology , Mice , Mice, Knockout , Mucus/metabolism , RNA, Messenger/genetics , Receptor, Adenosine A1/deficiency , Receptor, Adenosine A1/genetics , Transcription, Genetic/genetics
9.
J Immunol ; 173(2): 1380-9, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15240734

ABSTRACT

Adenosine signaling has been implicated in chronic lung diseases such as asthma and chronic obstructive pulmonary disease; however, the specific roles of the various adenosine receptors in processes central to these disorders are not well understood. In this study, we have investigated the role(s) of the A(3) adenosine receptor in adenosine-dependent pulmonary inflammation observed in adenosine deaminase (ADA)-deficient mice. The A(3) receptor (A(3)R) was found to be expressed in eosinophils and mucus-producing cells in the airways of ADA-deficient mice. Treatment of ADA-deficient mice with MRS 1523, a selective A(3)R antagonist, prevented airway eosinophilia and mucus production. Similar findings were seen in the lungs of ADA/A(3) double knockout mice. Although eosinophils were decreased in the airways of ADA-deficient mice following antagonism or removal of the A(3)R, elevations in circulating and lung interstitial eosinophils persisted, suggesting signaling through the A(3)R is needed for the migration of eosinophils into the airways. These findings identify an important role for the A(3)R in regulating lung eosinophilia and mucus production in an environment of elevated adenosine.


Subject(s)
Adenosine Deaminase/deficiency , Mucus/metabolism , Receptor, Adenosine A3/immunology , Respiratory System/immunology , Signal Transduction/immunology , Adenosine Deaminase/genetics , Animals , Eosinophils/metabolism , Inflammation/immunology , Mice , Mice, Knockout , RNA, Messenger/metabolism , Receptor, Adenosine A3/genetics , Respiratory System/metabolism
10.
Biol Reprod ; 70(4): 919-24, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14627551

ABSTRACT

External Ca2+ entry into myometrial smooth-muscle cells is important to uterine contraction and hence to labor progression and parturition. Proteins of the transient receptor potential (Trp) channel family are putative capacitative Ca2+ entry channels that respond to contractant-generated signals and intracellular Ca2+ store depletion. Quantitative reverse transcription-polymerase chain reaction was used to examine the relative expression of TrpC mRNAs in rat myometrium and determine their expression pattern during pregnancy and labor. rTrpC1, rTrpC2, rTrpC4, rTrpC5, rTrpC6, and rTrpC7 mRNAs, but not rTrpC3 mRNA, were expressed in nonpregnant rat myometrium. With the exception of rTrpC7, the resulting products were sequenced and found to be identical with published sequences; new rTrpC7 sequence exhibited >88% homology to mouse and human TrpC7 coding regions. Relative to beta-actin mRNA, rTrpC4 mRNA was expressed in the greatest abundance. rTrpC1, 5, and 6 mRNAs were expressed at lower levels, whereas rTrpC2 and 7 mRNAs were barely detectable. This relative expression pattern was also observed throughout the course of gestation. There were no major differences in expression of rTrpC1, 2, 4, or 7 mRNAs between Day 13 and Day 21 of gestation or labor. Rat TrpC5 and TrpC6 mRNA expression decreased in pregnancy but was not altered between Day 13 and Day 21 or in labor. Western blot analysis generally confirmed these observations with respect to protein expression. These data suggest that rTrpC4 may play a major role in regulated Ca2+ entry in myometrial cells and throughout pregnancy but do not rule out contributions from other Trp proteins.


Subject(s)
Calcium Channels/metabolism , Myometrium/metabolism , Pregnancy, Animal/metabolism , Animals , Blotting, Western , Female , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , TRPC Cation Channels
11.
Am J Respir Cell Mol Biol ; 30(1): 38-50, 2004 Jan.
Article in English | MEDLINE | ID: mdl-12855405

ABSTRACT

Adenosine signaling has been characterized in various physiologic systems, but little is known about the role of adenosine signaling in lung development. Alveogenesis and microvascular maturation are the final stages in lung development in mammals. Alveogenesis in the mouse begins on Postnatal Day 5, when the process of secondary septation plays a pivotal role in the expansion of the alveolar sacs and microvascular maturation. Adenosine deaminase null mice (ADA-/-) exhibit abnormalities in alveogenesis in association with elevated lung adenosine levels. Large-scale gene expression analysis of ADA-/- lungs using oligonucleotide-based microarrays revealed novel relationships between gene expression patterns and elevated lung adenosine during the stages of alveolar maturation. Genes regulating apoptosis, proliferation, and vascular development were shown to be altered, and decreased cell proliferation in association with increased alveolar type II cell apoptosis was shown to contribute to abnormal secondary septation in these mice. ADA enzyme therapy allowed for normal patterns of apoptosis, proliferation, and alveolar development in association with prevention of adenosine elevations. These findings were correlated with the presence of adenosine receptors in the developing lung, suggesting the involvement of receptor signaling. These studies provide evidence that elevated lung adenosine can lead to abnormal alveogenesis by disrupting patterns of cell proliferation and apoptosis.


Subject(s)
Adenine Nucleotides/metabolism , Pulmonary Alveoli/growth & development , Animals , Apoptosis , Base Sequence , DNA Primers , Gene Expression , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Receptors, Purinergic P1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
J Clin Invest ; 112(3): 332-44, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12897202

ABSTRACT

IL-13 is an important mediator of inflammation and remodeling. We hypothesized that adenosine accumulation, alterations in adenosine receptors, and adenosine-IL-13 autoinduction are critical events in IL-13-induced pathologies. To test this, we characterized the effects of IL-13 overexpression on the levels of adenosine, adenosine deaminase (ADA) activity, and adenosine receptors in the murine lung. We also determined whether adenosine induced IL-13 in lungs from ADA-null mice. IL-13 induced an inflammatory and remodeling response that caused respiratory failure and death. During this response, IL-13 caused a progressive increase in adenosine accumulation, inhibited ADA activity and mRNA accumulation, and augmented the expression of the A1, A2B, and A3 but not the A2A adenosine receptors. ADA enzyme therapy diminished the IL-13-induced increase in adenosine, inhibited IL-13-induced inflammation, chemokine elaboration, fibrosis, and alveolar destruction, and prolonged the survival of IL-13-transgenic animals. In addition, IL-13 was strongly induced by adenosine in ADA-null mice. These findings demonstrate that adenosine and adenosine signaling contribute to and influence the severity of IL-13-induced tissue responses. They also demonstrate that IL-13 and adenosine stimulate one another in an amplification pathway that may contribute to the nature, severity, progression, and/or chronicity of IL-13 and/or Th2-mediated disorders.


Subject(s)
Adenosine/physiology , Interleukin-13/physiology , Lung/physiopathology , Adenosine Deaminase/administration & dosage , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Animals , Inflammation/drug therapy , Inflammation/genetics , Inflammation/physiopathology , Interleukin-13/genetics , Lung/drug effects , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/physiology , Signal Transduction
13.
Am J Physiol Lung Cell Mol Physiol ; 282(2): L169-82, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11792620

ABSTRACT

Adenosine has been implicated as a modulator of inflammatory processes central to asthma. However, the molecular mechanisms involved are poorly understood. We used Atlas mouse cDNA arrays to analyze differential gene expression in association with lung inflammation resulting from elevated adenosine in adenosine deaminase (ADA)-deficient mice. We report that of the 1,176 genes on the array, the expression patterns of 280 genes were consistently altered. Of these genes, the steady-state levels of 93 genes were upregulated and 29 were downregulated. We also show that lowering adenosine levels with ADA enzyme therapy has striking effects on gene expression that may be associated with resolution of pulmonary eosinophilia. In addition, we confirmed the nucleic acid and protein expression of vascular endothelial growth factor and monocyte chemoattractant protein-3, two candidate genes that may be regulated by adenosine. In conclusion, high-throughput profiling of gene expression by cDNA array hybridization has provided an overview of critical regulatory genes involved in airway inflammation in ADA-deficient mice. These mice will serve as a useful in vivo model for characterizing molecular mechanisms of adenosine-mediated lung damage.


Subject(s)
Adenosine Deaminase/genetics , Adenosine/metabolism , Asthma/metabolism , Asthma/physiopathology , Cytokines , Adenosine Deaminase/pharmacology , Animals , Asthma/immunology , Chemokine CCL7 , Endothelial Growth Factors/analysis , Eosinophils/immunology , Gene Expression Regulation, Enzymologic , Immunohistochemistry , Lung/chemistry , Lung/enzymology , Lung/immunology , Lymphokines/analysis , Mice , Mice, Transgenic , Monocyte Chemoattractant Proteins/analysis , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...