Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Allergy Clin Immunol ; 148(5): 1227-1235.e6, 2021 11.
Article in English | MEDLINE | ID: mdl-33453288

ABSTRACT

BACKGROUND: Phenotypes and endotypes predicting optimal response to bronchial thermoplasty (BT) in patients with severe asthma remain elusive. OBJECTIVE: Our aim was to compare the clinical characteristics and hallmarks of airway inflammation and remodeling before and after BT in responder and partial responder patients with severe asthma refractory to oral steroids and to omalizumab. METHODS: In all, 23 patients with severe refractory asthma were divided into BT responders (n = 15) and BT partial responders (n = 8), according to the decrease in asthma exacerbations at 12 months after BT. Clinical parameters were compared at baseline and 12 months after BT, and hallmarks of airway inflammation and remodeling were analyzed by immunohistochemistry in bronchial biopsy specimens before and 3 months after BT. RESULTS: At baseline, the BT responders were around 8 years younger than the BT partial responders (P = .02) and they had a greater incidence of atopy, higher numbers of blood eosinophils (both P = .03) and IgE levels, higher epithelial IFN-α expression, and higher numbers of mucosal eosinophils and IL-33-positive cells (P ≤ .05). A reduction in blood eosinophil count, serum IgE level, type 2 airway inflammation, and numbers of mucosal IL-33-positive cells and mast cells associated with augmented epithelial MUC5AC and IFN-α/ß immunostaining was noted after BT in responders, whereas the numbers of mucosal IL-33-positive cells were augmented in BT partial responders. Most of these changes were correlated with clinical parameters. Subepithelial membrane thickening and airway smooth muscle area were similar in the 2 patient groups at baseline and after BT. CONCLUSION: By reducing allergic type 2 inflammation and increasing epithelial MUC5AC and anti-viral IFN-α/ß expression, BT may enhance host immune responses and thus attenuate exacerbations and symptoms in BT responders. Instead, targeting IL-33 may provide a clinical benefit in BT partial responders.


Subject(s)
Asthma/diagnosis , Bronchial Thermoplasty/methods , Th2 Cells/immunology , Adult , Anti-Asthmatic Agents/therapeutic use , Asthma/immunology , Asthma/therapy , Biomarkers , Disease Progression , Drug Resistance , Female , Humans , Interferons/metabolism , Interleukin-33/metabolism , Male , Middle Aged , Mucin 5AC/metabolism , Omalizumab/therapeutic use , Prognosis , Steroids/therapeutic use
3.
Respir Res ; 14: 34, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23496774

ABSTRACT

BACKGROUND: Subepithelial fibrosis is one of the most critical structural changes affecting bronchial airway function during asthma. Eosinophils have been shown to contribute to the production of pro-fibrotic cytokines, TGF-ß and IL-11, however, the mechanism regulating this process is not fully understood. OBJECTIVE: In this report, we investigated whether cytokines associated with inflammation during asthma may induce eosinophils to produce pro-fibrotic cytokines. METHODS: Eosinophils were isolated from peripheral blood of 10 asthmatics and 10 normal control subjects. Eosinophils were stimulated with Th1, Th2 and Th17 cytokines and the production of TGF-ß and IL-11 was determined using real time PCR and ELISA assays. RESULTS: The basal expression levels of eosinophil derived TGF-ß and IL-11 cytokines were comparable between asthmatic and healthy individuals. Stimulating eosinophils with Th1 and Th2 cytokines did not induce expression of pro-fibrotic cytokines. However, stimulating eosinophils with Th17 cytokines resulted in the enhancement of TGF-ß and IL-11 expression in asthmatic but not healthy individuals. This effect of IL-17 on eosinophils was dependent on p38 MAPK activation as inhibiting the phosphorylation of p38 MAPK, but not other kinases, inhibited IL-17 induced pro-fibrotic cytokine release. CONCLUSIONS: Th17 cytokines might contribute to airway fibrosis during asthma by enhancing production of eosinophil derived pro-fibrotic cytokines. Preventing the release of pro-fibrotic cytokines by blocking the effect of Th17 cytokines on eosinophils may prove to be beneficial in controlling fibrosis for disorders with IL-17 driven inflammation such as allergic and autoimmune diseases.


Subject(s)
Asthma/immunology , Bronchi/immunology , Cytokines/immunology , Eosinophils/immunology , Interleukin-17/immunology , Th2 Cells/immunology , Adult , Asthma/physiopathology , Bronchi/pathology , Bronchi/physiopathology , Cytokines/pharmacology , Enzyme-Linked Immunosorbent Assay , Eosinophils/metabolism , Female , Fibrosis , Humans , Inflammation/immunology , Interleukin-11/metabolism , Male , Polymerase Chain Reaction , Transforming Growth Factor beta/metabolism
4.
Am J Pathol ; 176(2): 638-49, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20042671

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterized by chronic airway inflammation and emphysematous alveolar destruction. In this study, we have investigated whether chitotriosidase (ChTRase) and acidic mammalian chitinase, two chitinases with chitinolytic activity, are selectively augmented in COPD and contribute to its pathogenesis. We found that smokers with COPD, but not asthmatics, had higher chitinolytic activity and increased levels of ChTRase in bronchoalveolar lavage, more ChTRase-positive cells in bronchial biopsies, and an elevated proportion of alveolar macrophages expressing ChTRase than smokers without COPD or never-smokers. ChTRase accounted for approximately 80% of bronchoalveolar lavage chitinolytic activity, while acidic mammalian chitinase was undetectable. Bronchoalveolar lavage chitinolytic activity and ChTRase were associated with airflow obstruction and emphysema and with the levels of interleukin (IL)-1beta, IL-8, tumor-necrosis factor (TNF)-alpha, and its type II soluble receptor. Tumor necrosis factor-alpha stimulated ChTRase release only from alveolar macrophages from smokers with COPD, and exposure of these cells to ChTRase promoted the release of IL-8, monocyte-chemoattractant protein-1, and metalloproteinase-9. Finally, ChTRase overexpression in the lung of normal mice promoted macrophage recruitment and the synthesis of the murine homologue of IL-8, keratinocyte-derived cytokine, and of monocyte-chemoattractant protein-1. We conclude that pulmonary ChTRase overexpression may represent a novel important mechanism involved in COPD onset and progression.


Subject(s)
Chitinases/metabolism , Hexosaminidases/metabolism , Lung/enzymology , Pneumonia/etiology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Animals , Asthma/metabolism , Asthma/pathology , Bronchoalveolar Lavage Fluid/chemistry , Cells, Cultured , Chitinases/physiology , Cytokines/analysis , Cytokines/metabolism , Female , Hexosaminidases/physiology , Humans , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Pneumonia/metabolism , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/immunology , Receptors, Cytokine/analysis , Receptors, Cytokine/metabolism , Smoking/metabolism , Validation Studies as Topic
5.
J Immunol ; 182(9): 5740-7, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380821

ABSTRACT

Bid, a proapoptotic member of Bcl-2 family, is involved in Fas receptor signaling. Fas activation promotes human eosinophil cell death and is believed to accelerate the resolution of pulmonary Th2-driven allergic reaction in mice. We hypothesized that Bid would regulate eosinophil apoptosis and Ag-induced airway inflammation, particularly eosinophilia. C57BL/6 Bid(-/-) and wild-type mice were immunized and repeatedly challenged with OVA, and bronchoalveolar lavage (BAL) fluid, lung, and spleen were collected 4-240 h after the final challenge. Cultured BAL eosinophils from Bid-deficient mice showed resistance to Fas-mediated apoptotic DNA fragmentation, phosphatidylserine exposure, mitochondria depolarization, and caspase-3 activity. In addition, OVA-challenged Bid(-/-) mice had higher BAL eosinophilia and a lower proportion of BAL apoptotic eosinophils than Bid(+/+) mice. This was accompanied by augmented BAL levels of the eosinophilotactic cytokine, IL-5, and of the eosinophil-associated mediators, TGF-beta1 and fibronectin. Finally, cultured OVA-stimulated lung mononuclear cells and splenocytes from Bid-deficient mice showed increased release of the Th2-type cytokines, IL-4 and IL-5, but no change in cell number. We conclude that Bid modulates BAL eosinophilia by regulating both eosinophil apoptosis and Th2-type cytokine production.


Subject(s)
Apoptosis/immunology , BH3 Interacting Domain Death Agonist Protein/physiology , Eosinophils/immunology , Eosinophils/pathology , Lung/pathology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Animals , Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein/deficiency , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Cytokines/biosynthesis , Eosinophils/metabolism , Inflammation Mediators/physiology , Lung/immunology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Hypersensitivity/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
6.
Am J Respir Crit Care Med ; 181(9): 917-27, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20133931

ABSTRACT

RATIONALE: Chronic obstructive pulmonary disease (COPD) is characterized by airway inflammation and remodeling. High-mobility group box 1 (HMGB1), a nuclear protein that is released during inflammation and repair, interacts with proinflammatory cytokines and with the receptor for advanced glycation end products (RAGE), which is highly expressed in the lung. OBJECTIVES: To determine whether HMGB1 is augmented in COPD and is associated with IL-1beta and RAGE. METHODS: HMGB1 was assessed in the bronchoalveolar lavage (BAL) of 20 never-smokers, 20 smokers, and 30 smokers with COPD and it was correlated with inflammatory and clinical parameters. In parallel, HMGB1 and RAGE immunolocalization was determined in bronchial and lung tissues. Last, binding of HMGB1 to IL-1beta in human macrophages and in BAL fluid was examined. MEASUREMENTS AND MAIN RESULTS: BAL levels of HMGB1 were higher in smokers with COPD than in smokers and never-smokers (P < 0.0001 for both comparisons), and similar differences were observed in epithelial cells and alveolar macrophages. BAL HMGB1 correlated positively with IL-1beta (r(s) = 0.438; P = 0.0006) and negatively with FEV(1) (r(s) = -0.570; P < 0.0001) and transfer factor of the lung for carbon monoxide (r(s) = -0.382; P = 0.0026). HMGB1-IL-1beta complexes were found in BAL supernatant and alveolar macrophages from smokers and patients with COPD, as well as in the human macrophage cell line, THP-1, where they enhanced the synthesis of tumor-necrosis factor-alpha. RAGE was overexpressed in the airway epithelium and smooth muscle of patients with COPD and it colocalized with HMGB1. CONCLUSIONS: Elevated HMGB1 expression in COPD airways may sustain inflammation and remodeling through its interaction with IL-1beta and RAGE.


Subject(s)
HMGB1 Protein/metabolism , Interleukin-1beta/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Receptors, Immunologic/metabolism , Airway Remodeling/physiology , Bronchi/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Cell Line , Female , Fluorescent Antibody Technique , Forced Expiratory Flow Rates , Humans , Immunohistochemistry , Lung/metabolism , Macrophages, Alveolar/metabolism , Male , Middle Aged , Receptor for Advanced Glycation End Products , Smoking/metabolism
7.
J Immunol ; 181(7): 5167-73, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802121

ABSTRACT

YKL-40 is a chitin-binding protein that is elevated in patients with various inflammatory conditions associated with ongoing remodeling. We investigated whether the levels of YKL-40 were up-regulated in the circulation and the airways of patients with chronic obstructive pulmonary disease (COPD), and whether it promoted the production of inflammatory mediators from macrophages. Serum, bronchoalveolar lavage (BAL), bronchial biopsies, lung tissue specimens, and alveolar macrophages from never-smokers (n = 15), smokers without COPD (n = 20), and smokers with COPD (n = 30) were assessed for YKL-40 levels and immunolocalization. In addition, YKL-40-induced mediator release from alveolar macrophages was examined. We found that smokers with COPD had elevated levels of YKL-40 in serum (p

Subject(s)
Glycoproteins/biosynthesis , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Adipokines , Aged , Bronchi/immunology , Bronchi/metabolism , Bronchi/pathology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Chitinase-3-Like Protein 1 , Female , Glycoproteins/blood , Glycoproteins/physiology , Humans , Lectins , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/pathology , Severity of Illness Index
8.
J Allergy Clin Immunol ; 120(6): 1425-32, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17761275

ABSTRACT

BACKGROUND: T(H)2 cytokines play crucial roles in driving human B lymphocytes to produce IgE. However, it is unclear whether IL-4 and IL-13 have parallel or sequential roles in the development of B lymphocytes. OBJECTIVE: We investigated IL-13 receptor (IL-13R) expression and regulation in mature and immature human B cells. METHODS: Purified B cells were isolated from human tonsils. We evaluated IL-13Ralpha1 mRNA expression using real-time PCR and IL-13Ralpha1 and IL-4 receptor (IL-4R) alpha expression using flow cytometry and microscopy. Signal transduction was assessed on the basis of signal transducer and activator of transcription 6 phosphorylation. RESULTS: IL13Ralpha1 mRNA was induced after stimulation with anti-CD40 antibodies, anti-CD40 plus IL-4, or anti-IgM/IgG. Baseline surface IL13Ralpha1 levels were low in unstimulated B cells but increased significantly at 24 hours and were sustained for 5 to 14 days. In contrast, IL4R alpha was constitutively expressed on tonsillar B cells, and levels did not significantly vary after stimulation. B cells activated by CD40 ligation or B-cell receptor cross-linking, but not resting B cells, showed significant increases in signal transducer and activator of transcription 6 phosphorylation in response to IL-13. IL-13Ralpha1 expression was induced on mature and memory B cells, as well as on naive subsets. CONCLUSIONS: There is lower constitutive expression and signaling of IL13Ralpha1 in resting tonsillar B lymphocytes compared with that of IL4R alpha. IL-13 is induced on both immature and mature B lymphocytes. CLINICAL IMPLICATIONS: This implies different roles for IL-4 and IL-13 in B-cell development, which would allow for specific targeting of IL-13 in IgE-mediated diseases.


Subject(s)
B-Lymphocyte Subsets/immunology , Gene Expression Regulation/immunology , Interleukin-13 Receptor alpha1 Subunit/biosynthesis , Palatine Tonsil/immunology , Signal Transduction/immunology , B-Lymphocyte Subsets/metabolism , Cells, Cultured , Humans , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/physiology , Interleukin-4/physiology , Lymphocyte Activation/immunology , Palatine Tonsil/cytology , Palatine Tonsil/metabolism , Phosphorylation , STAT6 Transcription Factor/metabolism , Up-Regulation/immunology
9.
Transplantation ; 101(9): 2192-2199, 2017 09.
Article in English | MEDLINE | ID: mdl-27820781

ABSTRACT

BACKGROUND: Bronchiolitis obliterans syndrome (BOS) after lung transplantation (LTx) results from bronchial epithelial cell (BECs) damages, thought to be orchestrated by T cells primed by antigen-presenting cell presenting alloantigens. In this cell cross-talk, BECs are also suspected to play a pivotal immunosuppressive role in T cell alloreactivity. We studied the immunomodulating role of BECs in a human ex vivo model of allogeneic T cell response, both in healthy subjects and LTx recipients. METHODS: BECs from 35 LTx recipients (n = 22 stable, n = 13 BOS) and healthy controls (n = 25) were cultured as primary cell cultures. Their inhibitory capacities through the involvement of tolerogenic molecules (HLA-G, TGF-ß, and IL-10) were tested on a mixed lymphocyte reaction between antigen-presenting cells and recipient T cells. RESULTS: Control BECs inhibited T cell alloproliferation by a mean of 53 ± 7%. This inhibitory effect of BECs was significantly reduced in the stable LTx group (24 ± 8%, P = 0.009), but not in the BOS TxP group (53 ± 10%, P = 0.97). Neutralization of HLA-G, TGF-ß, and IL-10 partially restored T cell alloproliferation, arguing for their involvement in the immunosuppressive effect of BECs. BECs culture supernatant from stable LTx patients with impaired BEC properties showed a skewed Th2-type secretion profile (high IL-4/IFN-γ ratio). CONCLUSIONS: The inhibitory properties of BECs are dysregulated in stable LTx recipients, which could suggest their instrumental role in the initiation of BOS process and potential targeted therapies.


Subject(s)
Bronchiolitis Obliterans/etiology , Epithelial Cells/metabolism , HLA-G Antigens/metabolism , Immunity, Mucosal , Interleukin-10/metabolism , Lung Transplantation/adverse effects , Respiratory Mucosa/metabolism , T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism , Bronchiolitis Obliterans/immunology , Bronchiolitis Obliterans/metabolism , Bronchiolitis Obliterans/pathology , Case-Control Studies , Cell Proliferation , Cells, Cultured , Coculture Techniques , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , HLA-G Antigens/immunology , Humans , Interleukin-10/immunology , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Male , Middle Aged , Primary Cell Culture , Prospective Studies , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transforming Growth Factor beta/immunology , Treatment Outcome
10.
J Allergy Clin Immunol ; 117(3): 590-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16522458

ABSTRACT

BACKGROUND: IL-17E is a new TH2 cytokine that promotes airway eosinophilia in mice. IL-17E proinflammatory activity has been proposed to involve induction of cytokine and chemokine production. Recruitment of inflammatory cells may be mediated by tissue-resident cells. OBJECTIVE: This study aimed to evaluate whether fibroblasts represent a target of IL-17E for the production of eosinophil active mediators in the lung. METHODS: Expression of IL-17B receptor (IL-17BR), a receptor for IL-17E, was evaluated by immunofluorescent staining, Western blot, and real-time PCR in human primary lung fibroblasts. Mediator production was analyzed by using real-time PCR and ELISA after stimulation of fibroblasts with IL-17E alone or in combination with TNF-alpha and TGF-beta1. Expression of IL-17E and of eosinophil major basic protein was evaluated by immunohistochemistry in bronchial biopsies from subjects with asthma. RESULTS: Human primary lung fibroblasts constitutively expressed IL-17BR. IL-17BR mRNA levels were increased in cells stimulated with TNF-alpha and decreased with TGF-beta1. IL-17E slightly upregulated CC chemokine ligand (CCL)-5, CCL-11, GM-CSF, and CXC chemokine ligand (CXCL)-8 mRNA in fibroblasts. Moreover, IL-17E and TNF-alpha synergistically induced GM-CSF and CXCL-8 mRNA. IL-17E also potentiated the upregulation of CXCL-8 transcripts observed with TGF-beta1. In contrast, TGF-beta1 decreased IL-17E-induced CCL-11 mRNA. The capacity of IL-17E to enhance GM-CSF and CXCL-8 responses to TNF-alpha was accompanied by production and secretion of both proteins by lung fibroblasts. Finally, IL-17E was detected in asthma in eosinophil-infiltrated bronchial submucosa. CONCLUSION: IL-17E may contribute to the induction and maintenance of eosinophilic inflammation in the airways by acting on lung fibroblasts. This study supports a role for IL-17E in asthma pathophysiology.


Subject(s)
Asthma/immunology , Cytokines/immunology , Eosinophils/immunology , Fibroblasts/immunology , Interleukin-17/physiology , Asthma/physiopathology , Biopsy , Bronchi/immunology , Bronchi/pathology , Cells, Cultured , Chemokine CCL11 , Chemokine CCL5 , Chemokines, CC/biosynthesis , Chemokines, CC/immunology , Chemokines, CXC/biosynthesis , Chemokines, CXC/immunology , Chemotactic Factors, Eosinophil/biosynthesis , Chemotactic Factors, Eosinophil/immunology , Chemotaxis, Leukocyte , Cytokines/biosynthesis , Cytokines/physiology , Eosinophil Major Basic Protein/biosynthesis , Eosinophil Major Basic Protein/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Inflammation , Interleukin-17/biosynthesis , Lung/immunology , Receptors, Interleukin/biosynthesis , Receptors, Interleukin/immunology , Receptors, Interleukin-17 , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1 , Tumor Necrosis Factor-alpha/physiology , Up-Regulation
11.
Am J Physiol Lung Cell Mol Physiol ; 290(6): L1238-46, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16428271

ABSTRACT

The interleukin-17B receptor (IL-17BR) is expressed in a variety of tissues and is upregulated under inflammatory conditions. This receptor binds both its cognate ligand IL-17B and IL-17E/IL-25, a novel cytokine known to promote Th2 responses. The present study shows that airway smooth muscle cells express IL-17BR in vitro and that its expression is upregulated by TNF-alpha and downregulated by IFN-gamma. Our data indicate that TNF-alpha upregulates IL-17BR mainly through nuclear factor-kappaB as assessed with the IkappaB kinase 2 inhibitor AS-602868. In addition, both IFN-gamma and dexamethasone are able to antagonize a TNF-alpha-induced IL-17BR increase in mRNA expression. The mitogen-activated protein kinase kinase inhibitor U0126 totally reversed the inhibition observed with IFN-gamma, suggesting the involvement of the extracellular signal-regulated kinase pathway in this effect. In addition, on stimulation with IL-17E, airway smooth muscle cells increase their expression of ECM components, namely procollagen-alphaI and lumican mRNA. Furthermore, immunohistochemical analysis of biopsies from asthmatic subjects reveals that this receptor is abundant in smooth muscle layers. This is the first report showing IL-17BR receptor in structural cells of the airways. Our results suggest a potential proremodeling effect of IL-17E on airway smooth muscle cells through the induction of ECM and that its receptor is upregulated by proinflammatory conditions.


Subject(s)
Cytokines/physiology , Gene Expression Regulation/physiology , Interferon-gamma/pharmacology , Interleukin-17/physiology , Muscle, Smooth/physiology , Receptors, Interleukin/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cytokines/pharmacology , Gene Expression Regulation/drug effects , Humans , Mice , RNA, Messenger/genetics , Receptors, Interleukin/drug effects , Receptors, Interleukin/genetics , Recombinant Proteins/pharmacology , Respiratory Physiological Phenomena , Transcription, Genetic
12.
Can J Physiol Pharmacol ; 83(1): 117-22, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15759058

ABSTRACT

Whole-thorax irradiation results in the development of the diffuse inflammatory response alveolitis in C3H/HeJ (C3H) mice and a milder alveolitis with fibrosis in C57BL/6J (B6) mice. In this study, we investigate if this mouse strain difference in response to radiation is due to differences in lung inflammatory cell apoptosis. Mice of the C3H and B6 strains were given a radiation dose of 18 Gy to the thorax and the animals were sacrificed at 11 or 18 weeks following exposure or when they were moribund. Active caspase-3 staining was used to identify apoptotic cells in the alveolar space of histological lung sections from the mice. The apoptotic index of B6 mice was greater than that of C3H mice at 11 weeks postirradiation (17.8% of airspace cells vs. 7.8%, p = 0.028) and in mice sacrificed because of illness (27.3% vs. 14.4%, p = 0.036). No C3H mice survived to the later time point. The inflammatory cells undergoing apoptosis in the mouse lungs were morphologically consistent with alveolar macrophages. We conclude that a difference in inflammatory cell apoptosis may contribute to the disparate pulmonary radiation response of these mouse strains.


Subject(s)
Apoptosis/radiation effects , Gamma Rays , Pulmonary Alveoli/radiation effects , Radiation Injuries, Experimental/pathology , Radiation Pneumonitis/pathology , Animals , Female , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Pulmonary Alveoli/pathology , Radiation Dosage , Radiation Injuries, Experimental/etiology , Radiation Pneumonitis/etiology , Species Specificity
13.
Am J Respir Cell Mol Biol ; 26(5): 565-71, 2002 May.
Article in English | MEDLINE | ID: mdl-11970908

ABSTRACT

Glucocorticosteroids are potent anti-inflammatory drugs used in the treatment of eosinophilic disorders. These molecules directly promote eosinophil apoptosis, yet the molecular mechanisms regulating this process remain ill-defined. We show here that stimulation of human peripheral blood eosinophils with dexamethasone induced DNA fragmentation, chromatin and cytoplasm condensation, and caspase-3 activation, as assessed by the proteolysis of its zymogen form and by the increase of caspase-3-like activity in eosinophil lysates. These phenomena were accompanied by a reduced uptake of the mitochondrial potential-sensitive marker DiOC(6)(3), suggestive of mitochondrial membrane permeabilization. Eosinophil incubation with the caspase-3 inhibitor, Z-Asp-Glu-Val-Asp-fluromethylketone, or with the broad spectrum caspase inhibitor, Z-Val-Ala-Asp-fluromethylketone, inhibited caspase-3-like activity generation but failed to modify dexamethasone-mediated loss in mitochondrial transmembrane potential and eosinophil apoptosis. In contrast, bongkrekic acid, a ligand of the mitochondrial permeability transition pore component, adenine nucleotide translocator, prevented both dexamethasone-induced mitochondrial disruption and apoptosis. We conclude that the mitochondrial permeability transition pore, rather than the caspase cascade, plays a critical role in the propagation of glucocorticosteroid-mediated apoptotic signals in human eosinophils.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Dexamethasone/pharmacology , Eosinophils/metabolism , Glucocorticoids/pharmacology , Mitochondria/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Anti-Bacterial Agents/pharmacology , Bongkrekic Acid/pharmacology , Caspase 3 , Caspase Inhibitors , Cells, Cultured , DNA Fragmentation/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Eosinophils/cytology , Eosinophils/drug effects , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Ion Channels/drug effects , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondrial ADP, ATP Translocases/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Oligopeptides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL