Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
PLoS One ; 8(2): e56172, 2013.
Article in English | MEDLINE | ID: mdl-23451035

ABSTRACT

BACKGROUND: Previous work has shown that disruption of the gene for group X secreted phospholipase A2 (sPLA2-X) markedly diminishes airway hyperresponsiveness and remodeling in a mouse asthma model. With the large number of additional sPLA2s in the mammalian genome, the involvement of other sPLA2s in the asthma model is possible - in particular, the group V sPLA2 (sPLA2-V) that like sPLA2-X is highly active at hydrolyzing membranes of mammalian cells. METHODOLOGY AND PRINCIPAL FINDINGS: The allergen-driven asthma phenotype was significantly reduced in sPLA2-V-deficient mice but to a lesser extent than observed previously in sPLA2-X-deficient mice. The most striking difference observed between the sPLA2-V and sPLA2-X knockouts was the significant impairment of the primary immune response to the allergen ovalbumin (OVA) in the sPLA2-V(-/-) mice. The impairment in eicosanoid generation and dendritic cell activation in sPLA2-V(-/-) mice diminishes Th2 cytokine responses in the airways. CONCLUSIONS: This paper illustrates the diverse roles of sPLA2s in the immunopathogenesis of the asthma phenotype and directs attention to developing specific inhibitors of sPLA2-V as a potential new therapy to treat asthma and other allergic disorders.


Subject(s)
Asthma/enzymology , Asthma/immunology , Group V Phospholipases A2/metabolism , Group X Phospholipases A2/metabolism , Animals , Asthma/genetics , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Group V Phospholipases A2/genetics , Group X Phospholipases A2/genetics , Immunohistochemistry , Mice , Mice, Knockout , Ovalbumin/immunology , Polymerase Chain Reaction , Th2 Cells/metabolism
2.
Proc Natl Acad Sci U S A ; 107(32): 14309-14, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660310

ABSTRACT

Idiopathic pulmonary fibrosis (IPF)/usual interstitial pneumonia is a ravaging condition of progressive lung scarring and destruction. Anti-inflammatory therapies including corticosteroids have limited efficacy in this ultimately fatal disorder. An important unmet need is to identify new agents that interact with key molecular pathways involved in the pathogenesis of pulmonary fibrosis to prevent progression or reverse fibrosis in these patients. Because aberrant activation of the Wnt/beta-catenin signaling cascade occurs in lungs of patients with IPF, we have targeted this pathway for intervention in pulmonary fibrosis using ICG-001, a small molecule that specifically inhibits T-cell factor/beta-catenin transcription in a cyclic AMP response-element binding protein binding protein (CBP)-dependent fashion. ICG-001 selectively blocks the beta-catenin/CBP interaction without interfering with the beta-catenin/p300 interaction. We report here that ICG-001 (5 mg/kg per day) significantly inhibits beta-catenin signaling and attenuates bleomycin-induced lung fibrosis in mice, while concurrently preserving the epithelium. Administration of ICG-001 concurrent with bleomycin prevents fibrosis, and late administration is able to reverse established fibrosis and significantly improve survival. Because no effective treatment for IPF exists, selective inhibition of Wnt/beta-catenin-dependent transcription suggests a potential unique therapeutic approach for pulmonary fibrosis.


Subject(s)
CREB-Binding Protein/metabolism , Pulmonary Fibrosis/prevention & control , Signal Transduction/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Bleomycin/administration & dosage , Bleomycin/toxicity , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Mice , Pulmonary Fibrosis/chemically induced , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Transcription, Genetic/drug effects
3.
Immunity ; 28(4): 546-58, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18387831

ABSTRACT

The regulatory T (Treg) cells restrain immune responses through suppressor-function elaboration that is dependent upon expression of the transcription factor Foxp3. Despite a critical role for Treg cells in maintaining lympho-myeloid homeostasis, it remains unclear whether a single mechanism or multiple mechanisms of Treg cell-mediated suppression are operating in vivo and how redundant such mechanisms might be. Here we addressed these questions by examining the role of the immunomodulatory cytokine IL-10 in Treg cell-mediated suppression. Analyses of mice in which the Treg cell-specific ablation of a conditional IL-10 allele was induced by Cre recombinase knocked into the Foxp3 gene locus showed that although IL-10 production by Treg cells was not required for the control of systemic autoimmunity, it was essential for keeping immune responses in check at environmental interfaces such as the colon and lungs. Our study suggests that Treg cells utilize multiple means to limit immune responses. Furthermore, these mechanisms are likely to be nonredundant, in that a distinct suppressor mechanism most likely plays a prominent and identifiable role at a particular tissue and inflammatory setting.


Subject(s)
Inflammation Mediators/physiology , Interleukin-10/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Colitis/genetics , Colitis/immunology , Colitis/prevention & control , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Inflammation Mediators/metabolism , Integrases/genetics , Interleukin-10/deficiency , Interleukin-10/genetics , Luminescent Proteins/genetics , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity/genetics , Organ Specificity/immunology , Skin/immunology , Skin/metabolism , Skin/pathology , T-Lymphocytes, Regulatory/pathology
4.
Am J Respir Crit Care Med ; 176(11): 1072-8, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17901411

ABSTRACT

RATIONALE: Secreted phospholipase A(2) enzymes (sPLA(2)s) play key regulatory roles in the biosynthesis of eicosanoids, such as the cysteinyl leukotrienes, but the role of these enzymes in the pathogenesis of asthma is not known. OBJECTIVES: To establish if sPLA(2)s are overexpressed in the airways of patients with asthma, and to determine if these enzymes may play a role in the generation of eicosanoids in exercise-induced bronchoconstriction. METHODS: Induced sputum samples were obtained from subjects with asthma with exercise-induced bronchoconstriction and nonasthmatic control subjects at baseline, and on a separate day 30 minutes after exercise challenge. The expression of the PLA(2)s in induced sputum cells and supernatant was determined by quantitative polymerase chain reaction, immunocytochemistry, and Western blot. MEASUREMENTS AND MAIN RESULTS: The sPLA(2)s expressed at the highest levels in airway cells of subjects with asthma were groups X and XIIA. Group X sPLA(2) (sPLA(2)-X) was differentially overexpressed in asthma and localized to airway epithelial cells and bronchial macrophages. The gene expression, immunostaining in airway epithelial cells and bronchial macrophages, and the level of the extracellular sPLA(2)-X protein in the airways increased in response to exercise challenge in the asthma group, whereas the levels were lower and unchanged after challenge in nonasthmatic control subjects. CONCLUSIONS: Increased expression of sPLA(2)-X may play a key role in the dysregulated eicosanoid synthesis in asthma.


Subject(s)
Asthma/enzymology , Bronchial Hyperreactivity/enzymology , Phospholipases A2, Secretory/metabolism , Adolescent , Adult , Asthma/metabolism , Asthma/physiopathology , Bronchi/enzymology , Bronchial Hyperreactivity/etiology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/physiopathology , Bronchoconstriction , Cohort Studies , Eicosanoids/analysis , Epithelial Cells/enzymology , Exercise , Female , Humans , Isoenzymes/metabolism , Male , Middle Aged , Phospholipases A2, Secretory/classification , Respiratory System/enzymology , Sputum/chemistry , Sputum/cytology
5.
Am J Physiol Heart Circ Physiol ; 293(4): H2119-28, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17660400

ABSTRACT

Hypothermia preserves myocardial function, promotes signaling for cell survival, and inhibits apoptotic pathways during 45-min reperfusion. We tested the hypothesis that signaling at the transcriptional level is followed by corresponding proteomic response and maintenance of structural integrity after 3-h reperfusion. Isolated hearts were Langendorff perfused and exposed to mild (I group; n = 6, 34 degrees C) or moderate (H group; n = 6, 30 degrees C) hypothermia during 120-min total ischemia with cardioplegic arrest and 180-min 37 degrees C reperfusion. Moderate hypothermia suppressed anaerobic metabolism during ischemia and significantly diminished left ventricular end-diastolic pressure at the end of ischemia from 52.7 +/- 3.3 (I group) to 1.8 +/- 0.9 (H group) mmHg. Unlike the I group, which showed poor cardiac function and high left ventricular pressure, the H group showed preservation of myocardial function, coronary flow, and oxygen consumption. Compared with normal control hearts without ischemia (n = 5), histological staining in the I group showed marked disarray and fragmentation of collagen network (score 4-5), while the H group showed preserved collagen integrity (score 0-1). The apoptosis-linked tumor suppressor protein p53 was expressed throughout the I group only (score 4-5). The H group produced elevated expression for hypoxia-inducible factor 1alpha and heme oxygenase 1, but minimally affected vascular endothelial growth factor expression. The H group also elevated expression for survival proteins peroxisomal proliferator-activated receptor-beta and Akt-1. These results show in a constant left ventricular volume model that moderate hypothermia (30 degrees C) decreases myocardial energy utilization during ischemia and subsequently promotes expression of proteins involved in cell survival, while inhibiting induction of p53 protein. These data also show that 34 degrees C proffers less protection and loss of myocardial integrity.


Subject(s)
Hypothermia, Induced , Intracellular Signaling Peptides and Proteins/metabolism , Myocardial Ischemia/therapy , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Signal Transduction , Animals , Cell Survival , Collagen/metabolism , Coronary Circulation , Disease Models, Animal , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Myocardial Contraction , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/pathology , Oxygen Consumption , PPAR-beta/genetics , PPAR-beta/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rabbits , Signal Transduction/genetics , Time Factors , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Ventricular Function, Left , Ventricular Pressure
6.
J Exp Med ; 204(4): 865-77, 2007 Apr 16.
Article in English | MEDLINE | ID: mdl-17403936

ABSTRACT

Arachidonic acid metabolites, the eicosanoids, are key mediators of allergen-induced airway inflammation and remodeling in asthma. The availability of free arachidonate in cells for subsequent eicosanoid biosynthesis is controlled by phospholipase A(2)s (PLA(2)s), most notably cytosolic PLA(2)-alpha. 10 secreted PLA(2)s (sPLA(2)s) have also been identified, but their function in eicosanoid generation is poorly understood. We investigated the role of group X sPLA(2) (sPLA(2)-X), the sPLA(2) with the highest in vitro cellular phospholipolysis activity, in acute and chronic mouse asthma models in vivo. The lungs of sPLA(2)-X(-/-) mice, compared with those of sPLA(2)-X(+/+) littermates, had significant reduction in ovalbumin-induced infiltration by CD4(+) and CD8(+) T cells and eosinophils, goblet cell metaplasia, smooth muscle cell layer thickening, subepithelial fibrosis, and levels of T helper type 2 cell cytokines and eicosanoids. These data direct attention to sPLA(2)-X as a novel therapeutic target for asthma.


Subject(s)
Allergens/immunology , Asthma/enzymology , Asthma/immunology , Disease Models, Animal , Phospholipases A/metabolism , Animals , Asthma/genetics , Asthma/pathology , Cytokines/metabolism , Eicosanoids/metabolism , Gene Expression Regulation, Enzymologic , Group X Phospholipases A2 , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Metaplasia/enzymology , Metaplasia/pathology , Mice , Mice, Knockout , Phospholipases A/deficiency , Phospholipases A/genetics , Phospholipases A2 , Th2 Cells/enzymology
7.
Am J Respir Crit Care Med ; 173(7): 718-28, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16387808

ABSTRACT

RATIONALE: Airway inflammation in asthma is accompanied by structural changes, including goblet cell metaplasia, smooth muscle cell layer thickening, and subepithelial fibrosis. This allergen-induced airway remodeling can be replicated in a mouse asthma model. OBJECTIVES: The study goal was to determine whether established airway remodeling in a mouse asthma model is reversible by administration of the cysteinyl leukotriene (CysLT)1 receptor antagonist montelukast, the corticosteroid dexamethasone, or the combination montelukast + dexamethasone. METHODS: BALB/c mice, sensitized by intraperitoneal ovalbumin (OVA) as allergen, received intranasal OVA periodically Days 14-73 and montelukast or dexamethasone or placebo from Days 73-163. MEASUREMENTS AND MAIN RESULTS: Allergen-induced trafficking of eosinophils into the bronchoalveolar lavage fluid and lung interstitium and airway goblet cell metaplasia, smooth muscle cell layer thickening, and subepithelial fibrosis present on Day 73 persisted at Day 163, 3 mo after the last allergen challenge. Airway hyperreactivity to methacholine observed on Day 73 in OVA-treated mice was absent on Day 163. In OVA-treated mice, airway eosinophil infiltration and goblet cell metaplasia were reduced by either montelukast or dexamethasone alone. Montelukast, but not dexamethasone, reversed the established increase in airway smooth muscle mass and subepithelial collagen deposition. By immunocytochemistry, CysLT1 receptor expression was significantly increased in airway smooth muscle cells in allergen-treated mice compared with saline-treated controls and was reduced by montelukast, but not dexamethasone, administration. CONCLUSIONS: These data indicate that established airway smooth muscle cell layer thickening and subepithelial fibrosis, key allergen-induced airway structural changes not modulated by corticosteroids, are reversible by CysLT1 receptor blockade therapy.


Subject(s)
Acetates/therapeutic use , Asthma/pathology , Leukotriene Antagonists/therapeutic use , Lung/pathology , Membrane Proteins/antagonists & inhibitors , Quinolines/therapeutic use , Animals , Asthma/drug therapy , Asthma/metabolism , Cyclopropanes , Disease Models, Animal , Female , Follow-Up Studies , Glucocorticoids/therapeutic use , Lung/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Receptors, Leukotriene/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Sulfides , Treatment Outcome
8.
J Allergy Clin Immunol ; 116(2): 332-40, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16083788

ABSTRACT

BACKGROUND: (R)- and (S)-Enantiomers of albuterol likely exert differential effects in patients with asthma. The (R)-enantiomer binds to the beta2-adrenergic receptor with greater affinity than the (S)-enantiomer and is responsible for albuterol's bronchodilating activity. (S)-Albuterol augments bronchospasm and has proinflammatory actions. OBJECTIVE: The study aim was to determine whether the (S)-enantiomer, in contrast to the (R)-enantiomer, has adverse effects on allergic airway inflammation and hyperresponsiveness in a mouse asthma model. METHODS: Mice sensitized to ovalbumin (OVA) intraperitoneally on days 0 and 14 were challenged with OVA intranasally on days 14, 25, and 35. On day 36, 24 hours after the final allergen challenge, the effect of the (R)- and (S)-enantiomers of albuterol (1 mg x kg(-1) x d(-1) administered by means of a miniosmotic pump from days 13-36) on airway inflammation and hyperreactivity was determined. RESULTS: In OVA-sensitized/OVA-challenged mice, (R)-albuterol significantly reduced the influx of eosinophils into the bronchoalveolar lavage fluid and airway tissue. (R)-Albuterol also significantly decreased airway goblet cell hyperplasia and mucus occlusion and levels of IL-4 in bronchoalveolar lavage fluid and OVA-specific IgE in plasma. Although (S)-albuterol significantly reduced airway eosinophil infiltration, goblet cell hyperplasia, and mucus occlusion, it increased airway edema and responsiveness to methacholine in OVA-sensitized/OVA-challenged mice. Allergen-induced airway edema and pulmonary mechanics were unaffected by (R)-albuterol. CONCLUSION: Both (R)- and (S)-enantiomers of albuterol reduce airway eosinophil trafficking and mucus hypersecretion in a mouse model of asthma. However, (S)-albuterol increases allergen-induced airway edema and hyperresponsiveness. These adverse effects of the (S)-enantiomer on lung function might limit the clinical efficacy of racemic albuterol.


Subject(s)
Adrenergic beta-Agonists/therapeutic use , Albuterol/therapeutic use , Asthma/drug therapy , Animals , Apoptosis/drug effects , Asthma/immunology , Bronchial Hyperreactivity/drug therapy , Disease Models, Animal , Female , Interleukin-13/physiology , Interleukin-4/physiology , Mice , Mice, Inbred BALB C , Mucus/metabolism , Ovalbumin/immunology , Stereoisomerism
9.
Crit Care Med ; 32(12): 2496-501, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15599157

ABSTRACT

OBJECTIVE: Mechanical overdistension and hyperoxia can independently cause lung injury, yet little is known about their combined effects. We hypothesized that hyperoxia exacerbates lung injury caused by large tidal volume ventilation. DESIGN: Experimental study. SETTING: University laboratory. SUBJECTS: Anesthetized, paralyzed rabbits. INTERVENTIONS: In experiment 1, 12 rabbits were ventilated with 25 mL/kg tidal volumes at positive end-expiratory pressure of 0 cm H2O for 4 hrs with either hyperoxia (HO; FiO2 = 0.5) or normoxia (NO; FiO2 = 0.21). In experiment 2, a separate group of animals were randomized to one of four groups to assess the interaction of tidal volume and inspired oxygen concentration on potential mediators of injury after 2 hrs of ventilation, before significant injury occurs: a) NO+normal tidal volume (NV; VT = 10 mL/kg); b) HO+NV; c) NO+high tidal volume (HV; VT = 25 mL/kg); d) HO+HV (n = 3 per group). MEASUREMENTS AND MAIN RESULTS: : In the first study, HO compared with the NO group had significantly reduced PaO2/FiO2 ratio (320 +/- 110 vs. 498 +/- 98, p = .014) and increased lung injury scores at 4 hrs. Hyperoxia also significantly increased polymorphonuclear leukocytes, growth-related oncogene-alpha (2073 +/- 535 vs. 463 +/- 236 pg/mL, p = .02), and monocyte chemotactic protein-1 (7517 +/- 1612 vs. 2983 +/- 1289 pg/mL, p = .05) concentrations in bronchoalveolar lavage fluid. The second study showed increased alveolar-capillary permeability to a 70-kD fluorescent-labeled dextran only in response to the combination of both HO and HV. Chemokines and bronchoalveolar lavage fluid neutrophils were elevated in both HV groups; however, hyperoxia did not further increase chemokine or neutrophil counts over normoxia. No difference in lipid peroxidation was seen between groups. CONCLUSIONS: Moderate hyperoxia exacerbates lung injury in a large tidal volume model of ventilator-induced lung injury. The mechanism by which this occurs is not mediated by increased production of CXC chemokines or lipid peroxidation.


Subject(s)
Hyperoxia/complications , Respiration, Artificial/adverse effects , Respiratory Distress Syndrome/etiology , Animals , Cytokines/analysis , Cytokines/metabolism , Disease Models, Animal , Extravascular Lung Water/metabolism , Female , Male , Positive-Pressure Respiration/adverse effects , Positive-Pressure Respiration/methods , Rabbits , Respiration, Artificial/methods , Respiratory Distress Syndrome/physiopathology , Respiratory Mechanics , Risk Factors , Sensitivity and Specificity , Tidal Volume/physiology
10.
J Ultrasound Med ; 23(2): 217-25, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14992358

ABSTRACT

OBJECTIVE: Previous studies have shown that high-intensity focused ultrasound can effectively control bleeding from injuries of liver, spleen, and blood vessels. This study investigated long-term hemostasis and tissue repair after high-intensity focused ultrasound treatment in liver. METHODS: A total of 21 rabbits were randomly assigned to 2 groups: high-intensity focused ultrasound treatment (n = 14) and sham treatment (n = 7). All animals had sterile laparotomy and liver exposure. The high-intensity focused ultrasound-treated animals received liver incisions, 20 to 25 mm long and 4 to 6 mm deep, followed immediately by high-intensity focused ultrasound application until complete hemostasis was achieved. After recovery, sonographic images, blood samples, and histologic samples were collected immediately and on days 1, 3, 7, 14, 28, and 60 after treatment. RESULTS: All 14 liver injuries were hemostatic after an average +/- SD of 78 +/- 44 seconds of high-intensity focused ultrasound application, with no rebleeding at any time point after the treatment. Subsequent blood analysis showed no significant difference in serial hematologic or coagulation measures between the high-intensity focused ultrasound and sham groups. Alanine aminotransferase and aspartate aminotransferase levels increased immediately after surgery by as much as 285% up to day 3 and returned to normal values by day 7. Hematocrit and white blood cell counts showed no statistically significant difference from normal values at all time points. Histologic examination up to 60 days after treatment revealed scarring and liver tissue regeneration at the treatment site. CONCLUSIONS: High-intensity focused ultrasound appears to provide long-lasting hemostasis of acute liver injury. Healing and repair mechanisms after high-intensity focused ultrasound application appear to be intact.


Subject(s)
Liver Diseases/therapy , Ultrasonic Therapy/methods , Animals , Disease Models, Animal , Hemostatic Techniques , Liver Diseases/diagnostic imaging , Rabbits , Ultrasonography , Wound Healing/physiology
11.
Comb Chem High Throughput Screen ; 6(7): 611-21, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14683490

ABSTRACT

There is increasing evidence that redox regulation of transcription, particularly activator protein-1 (AP-1) and nuclear factor kappa B (NF-kappaB), is important in inflammatory diseases. Human thioredoxin (TRX), a member of the oxidoreductase superfamily, was initially identified, as a factor which augments the production of interleukin-2 receptor alpha (IL-2R alpha) in human T-cell lymphotropic virus type 1 (HTLV-1) infected patient T-cells. Substrates for the redox activity of TRX bind the active site cleft in extended strand structure. The rapid generation of high numbers of peptide secondary structure mimetics through solid-phase synthesis is a key technology for the identification of pharmaceutical leads based on such protein-peptide interactions. In this manuscript, we describe a chemogenomic approach utilizing an extended strand templated library to develop small molecule inhibitors to validate oxidoreductase molecular targets in a murine asthma model.


Subject(s)
Molecular Mimicry , Peptides/chemistry , Peptides/pharmacology , Animals , Asthma/drug therapy , Combinatorial Chemistry Techniques , Enzyme Inhibitors , Humans , Mice , Peptide Library , Peptides/therapeutic use , Protein Structure, Secondary , Receptors, G-Protein-Coupled/agonists , Transcription, Genetic/drug effects
12.
J Immunol ; 170(6): 3386-91, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12626599

ABSTRACT

Asthma is characterized by acute and chronic airway inflammation, and the severity of the airway hyperreactivity correlates with the degree of inflammation. Many of the features of lung inflammation observed in human asthma are reproduced in OVA-sensitized/challenged mice. T lymphocytes, particularly Th2 cells, are critically involved in the genesis of the allergic response to inhaled Ag. In addition to antiapoptotic effects, broad-spectrum caspase inhibitors inhibit T cell activation in vitro. We investigated the effect of the broad-spectrum caspase inhibitor, N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (z-VAD-fmk), on airway inflammation in OVA-sensitized/challenged mice. OVA-sensitized mice treated with z-VAD-fmk immediately before allergen challenge showed marked reduction in inflammatory cell infiltration in the airways and pulmonary blood vessels, mucus production, and Th2 cytokine production. We hypothesized that the caspase inhibitor prevented T cell activation, resulting in the reduction of cytokine production and eosinophil infiltration. Treatment with z-VAD-fmk in vivo prevented subsequent T cell activation ex vivo. We propose that caspase inhibitors may offer a novel therapeutic approach to T cell-dependent inflammatory airway diseases.


Subject(s)
Allergens/immunology , Amino Acid Chloromethyl Ketones/pharmacology , Asthma/enzymology , Asthma/immunology , Caspase Inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Lung/immunology , Lung/pathology , Aerosols , Allergens/administration & dosage , Amino Acid Chloromethyl Ketones/therapeutic use , Animals , Asthma/pathology , Bronchial Hyperreactivity/enzymology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Movement/drug effects , Cell Movement/immunology , Disease Models, Animal , Inflammation/enzymology , Inflammation/immunology , Inflammation/prevention & control , Interleukin-4/metabolism , Interleukin-5/metabolism , Intubation, Intratracheal , Leukocytes/pathology , Lung/enzymology , Lymphocyte Activation/drug effects , Methacholine Chloride/administration & dosage , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Ovalbumin/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
Proc Natl Acad Sci U S A ; 100(3): 1169-73, 2003 Feb 04.
Article in English | MEDLINE | ID: mdl-12552119

ABSTRACT

Asthma is characterized by an oxidantantioxidant imbalance in the lungs leading to activation of redox-sensitive transcription factors, nuclear factor kappaB (NF-kappaB), and activator protein-1 (AP-1). To develop therapeutic strategies for asthma, we used a chemogenomics approach to screen for small molecule inhibitor(s) of AP-1 transcription. We developed a beta-strand mimetic template that acts as a reversible inhibitor (pseudosubstrate) of redox proteins. This template incorporates an enedione moiety to trap reactive cysteine nucleophiles in the active sites of redox proteins. Specificity for individual redox factors was achieved through variations in X and Y functionality by using a combinatorial library approach. A limited array (2 x 6) was constructed where X was either NHCH(3) or NHCH(2) Ph and Y was methyl, phenyl, m-cyanophenyl, m-nitrophenyl, m-acetylaniline, or m-methylbenzoate. These analogs were evaluated for their ability to inhibit transcription in transiently transfected human lung epithelial A549 cells from either an AP-1 or NF-kappaB reporter. A small-molecule inhibitor, PNRI-299, was identified that selectively inhibited AP-1 transcription (IC(50) of 20 microM) without affecting NF-kappaB transcription (up to 200 microM) or thioredoxin (up to 200 microM). The molecular target of PNRI-299 was determined to be the oxidoreductase, redox effector factor-1 by an affinity chromatography approach. The selective redox effector factor-1 inhibitor, PNRI-299, significantly reduced airway eosinophil infiltration, mucus hypersecretion, edema, and IL-4 levels in a mouse asthma model. These data validate AP-1 as an important therapeutic target in allergic airway inflammation.


Subject(s)
Asthma/drug therapy , Carbon-Oxygen Lyases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase , Transcription Factor AP-1/metabolism , Animals , Binding Sites , Bronchoalveolar Lavage Fluid , Cell Nucleus/metabolism , Chromatography , Cysteine/chemistry , Cytosol/metabolism , Female , Genes, Reporter , Humans , Inhibitory Concentration 50 , Interleukin-4/metabolism , Lung/drug effects , Mice , Mice, Inbred BALB C , Models, Chemical , Models, Molecular , NF-kappa B/antagonists & inhibitors , Oxidation-Reduction , Peptide Library , Thioredoxins/antagonists & inhibitors , Tumor Cells, Cultured
14.
J Trauma ; 53(6): 1115-20, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12478037

ABSTRACT

BACKGROUND: Previous studies have shown that high-intensity focused ultrasound (HIFU) can effectively control bleeding of incised livers and spleens and punctured vessels. This current study investigated the long-term safety of HIFU in splenic hemostasis. METHODS: A total of 21 rabbits were randomly assigned to two groups: HIFU treatment (n = 14), and sham treatment (n = 7). All animals underwent sterile laparotomy and splenic exposure. The HIFU-treated animals received splenic incisions, 8 to 10 mm long and 4 to 5 mm deep, and immediate 9.6-MHz HIFU until hemostasis was achieved. After recovery, ultrasound images, blood samples, and histologic samples were collected on days 0, 1, 3, 7, 14, 28, and 60. RESULTS: All 14 splenic injuries were hemostatic after an average of 96 seconds of HIFU application. There was evidence of rebleeding in one animal between days 3 and 7 posttreatment. Subsequent blood analysis showed no significant difference in serial hematologic or coagulation measures between HIFU and sham groups. Histologic examination up to 60 days posttreatment revealed scarring and spleen tissue regeneration at the treatment site. CONCLUSION: HIFU provides an effective and safe method of achieving hemostasis after acute splenic injury.


Subject(s)
Hemorrhage/therapy , Splenic Diseases/therapy , Ultrasonic Therapy/methods , Animals , Disease Models, Animal , Female , Hemorrhage/diagnostic imaging , Hemorrhage/mortality , Hemostatic Techniques , Male , Rabbits , Random Allocation , Reference Values , Regeneration/physiology , Sensitivity and Specificity , Splenic Diseases/diagnostic imaging , Splenic Diseases/mortality , Survival Analysis , Treatment Outcome , Ultrasonography
15.
J Immunol ; 169(9): 5294-9, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391249

ABSTRACT

An oxidant/antioxidant imbalance is seen in the lungs of patients with asthma. This oxidative stress in asthmatic airways may lead to activation of redox-sensitive transcription factors, NF-kappaB and AP-1. We examined the effect of the small molecule inhibitor of redox-regulated NF-kappaB and AP-1 transcription, MOL 294 on airway inflammation and airway hyperreactivity (AHR) in a mouse model of asthma. MOL 294 is a potent nonpeptide inhibitor of NF-kappaB and AP-1 based upon a beta-strand template that binds to and inhibits the cellular redox protein thioredoxin. BALB/c mice after i.p. OVA sensitization (day 0) were challenged with intranasal OVA on days 14, 25, 26, and 27. MOL 294, administered intranasal on days 25-27, blocked the airway inflammatory response to OVA assessed 24 h after the last OVA challenge on day 28. MOL 294 reduced eosinophil, IL-13, and eotaxin levels in bronchoalveolar lavage fluid and airway tissue eosinophilia and mucus hypersecretion. MOL 294 also decreased AHR in vivo to methacholine. These results support redox-regulated transcription as a therapeutic target in asthma and demonstrate that selective inhibitors can reduce allergic airway inflammation and AHR.


Subject(s)
Allergens/administration & dosage , Asthma/metabolism , Asthma/prevention & control , Lung/pathology , NF-kappa B/antagonists & inhibitors , Pyridazines/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Triazoles/pharmacology , Administration, Intranasal , Animals , Asthma/pathology , Bronchial Hyperreactivity/prevention & control , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Movement/drug effects , Cell Movement/immunology , Chemokine CCL11 , Chemokines, CC/biosynthesis , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/pathology , Female , Humans , Inflammation/metabolism , Inflammation/prevention & control , Interleukin-13/biosynthesis , Lung/drug effects , Lung/immunology , Mice , Mice, Inbred BALB C , Mucus/drug effects , Mucus/immunology , Mucus/metabolism , NF-kappa B/metabolism , Ovalbumin/administration & dosage , Ovalbumin/immunology , Oxidation-Reduction/drug effects , Pyridazines/therapeutic use , Thioredoxins/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Triazoles/therapeutic use , Tumor Cells, Cultured
16.
Gynecol Oncol ; 86(3): 344-50, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12217758

ABSTRACT

OBJECTIVE: The objective was to test the efficacy of high-intensity focused ultrasound (HIFU) for treatment of uterine leiomyosarcoma in a Xenograft nude mouse model. METHODS: A total of 65 athymic nude mice were inoculated subcutaneously with 5 to 7 x 10(6) ELT-5B cells, a uterine leiomyosarcoma cell line derived from the Eker rat. Thirty animals showed tumor growth. The tumor volume was measured transcutaneously once a week. Animals were randomly assigned to three groups: HIFU treatment (n = 17), sham treatment (n = 7), and control (n = 6). A HIFU device, operating at a frequency of 2.0 MHz and an intensity of 2000 W/cm(2), was used for treatment. RESULTS: Within 3 weeks of a single HIFU treatment, 100% reduction in tumor volume was observed in all animals, except one. A second HIFU treatment was applied to that animal, resulting in 100% reduction in tumor volume. The tumors in the sham-treated animals continued to grow at a similar rate to that of the control group to approximately 500% of the tumor volume at the time of treatment. All animals were monitored for a maximum of 3 months. No metastasis was observed in the HIFU-treated animals. Histological examination confirmed a complete tumor disappearance after HIFU treatment. CONCLUSION: We have shown that HIFU can effectively treat uterine leiomyosarcoma tumors inoculated in Xenograft nude mice, demonstrating HIFU's potential use for treatment of recurrent uterine leiomyosarcoma.


Subject(s)
Leiomyosarcoma/therapy , Neoplasm Recurrence, Local/therapy , Ultrasonic Therapy/methods , Uterine Neoplasms/therapy , Animals , Female , Mice , Mice, Nude , Rats , Xenograft Model Antitumor Assays
17.
Am J Respir Crit Care Med ; 166(3): 403-8, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12153979

ABSTRACT

To investigate whether hypercapnic acidosis protects against ventilator-induced lung injury (VILI) in vivo, we subjected 12 anesthetized, paralyzed rabbits to high tidal volume ventilation (25 cc/kg) at 32 breaths per minute and zero positive end-expiratory pressure for 4 hours. Each rabbit was randomized to receive either an FI(CO(2)) to achieve eucapnia (Pa(CO(2)) approximately 40 mm Hg; n = 6) or hypercapnic acidosis (Pa(CO(2)) 80-100 mm Hg; n = 6). Injury was assessed by measuring differences between the two groups' respiratory mechanics, gas exchange, wet:dry weight, bronchoalveolar lavage fluid protein concentration and cell count, and injury score. The eucapnic group showed significantly higher plateau pressures (27.0 +/- 2.5 versus 20.9 +/- 3.0; p = 0.016), change in Pa(O(2)) (165.2 +/- 19.4 versus 77.3 +/- 87.9 mm Hg; p = 0.02), wet:dry weight (9.7 +/- 2.3 versus 6.6 +/- 1.8; p = 0.04), bronchoalveolar lavage protein concentration (1,350 +/- 228 versus 656 +/- 511 micro g/ml; p = 0.03), cell count (6.86 x 10(5) +/- 0.18 x 10(5) versus 2.84 x 10(5) +/- 0.28 x 10(5) nucleated cells/ml; p = 0.021), and injury score (7.0 +/- 3.3 versus 0.7 +/- 0.9; p < 0.0001). We conclude that hypercapnic acidosis is protective against VILI in this model.


Subject(s)
Acidosis, Respiratory/physiopathology , Hypercapnia/physiopathology , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/physiopathology , Ventilators, Mechanical/adverse effects , Acidosis, Respiratory/complications , Acidosis, Respiratory/pathology , Animals , Disease Models, Animal , Hemodynamics/physiology , Hypercapnia/complications , Hypercapnia/pathology , Pulmonary Gas Exchange/physiology , Rabbits , Random Allocation , Respiratory Distress Syndrome/complications , Respiratory Mechanics/physiology
18.
J Immunol ; 168(4): 1992-2000, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11823536

ABSTRACT

Release of human lung mast cell tryptase may be important in the pathophysiology of asthma. We examined the effect of the reversible, nonelectrophilic tryptase inhibitor MOL 6131 on airway inflammation and hyper-reactivity in a murine model of asthma. MOL 6131 is a potent selective nonpeptide inhibitor of human lung mast cell tryptase based upon a beta-strand template (K(i) = 45 nM) that does not inhibit trypsin (K(i) = 1,061 nM), thrombin (K(i) = 23, 640 nM), or other serine proteases. BALB/c mice after i.p. OVA sensitization (day 0) were challenged intratracheally with OVA on days 8, 15, 18, and 21. MOL 6131, administered days 18-21, blocked the airway inflammatory response to OVA assessed 24 h after the last OVA challenge on day 22; intranasal delivery (10 mg/kg) had a greater anti-inflammatory effect than oral delivery (10 or 25 mg/kg) of MOL 6131. MOL 6131 reduced total cells and eosinophils in bronchoalveolar lavage fluid, airway tissue eosinophilia, goblet cell hyperplasia, mucus secretion, and peribronchial edema and also inhibited the release of IL-4 and IL-13 in bronchoalveolar lavage fluid. However, tryptase inhibition did not alter airway hyper-reactivity to methacholine in vivo. These results support tryptase as a therapeutic target in asthma and indicate that selective tryptase inhibitors can reduce allergic airway inflammation.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Bronchial Diseases/drug therapy , Piperidines/therapeutic use , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/therapeutic use , Animals , Asthma/drug therapy , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bronchial Diseases/immunology , Bronchial Diseases/pathology , Bronchial Hyperreactivity/drug therapy , Bronchoalveolar Lavage Fluid/immunology , Cell Movement , Cytokines/biosynthesis , Eosinophils/immunology , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Models, Molecular , Mucus/metabolism , Ovalbumin/immunology , Piperidines/chemistry , Piperidines/pharmacology , Pulmonary Edema/drug therapy , Pulmonary Edema/pathology , Pulmonary Eosinophilia/drug therapy , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Tryptases , Vascular Cell Adhesion Molecule-1/metabolism
19.
Am J Respir Crit Care Med ; 165(1): 108-16, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11779739

ABSTRACT

Airway inflammation and remodeling in chronic asthma are characterized by airway eosinophilia, hyperplasia of goblet cells and smooth muscle, and subepithelial fibrosis. We examined the role of leukotrienes in a mouse model of allergen-induced chronic lung inflammation and fibrosis. BALB/c mice, after intraperitoneal ovalbumin (OVA) sensitization on Days 0 and 14, received intranasal OVA periodically Days 14-75. The OVA-treated mice developed an extensive eosinophil and mononuclear cell inflammatory response, goblet cell hyperplasia, and mucus occlusion of the airways. A striking feature of this inflammatory response was the widespread deposition of collagen beneath the airway epithelial cell layer and also in the lung interstitium in the sites of leukocytic infiltration that was not observed in the saline-treated controls. The cysteinyl leukotriene(1) (CysLT(1)) receptor antagonist montelukast significantly reduced the airway eosinophil infiltration, mucus plugging, smooth muscle hyperplasia, and subepithelial fibrosis in the OVA-sensitized/challenged mice. The presence of Charcot-Leyden-like crystals in airway macrophages and the increased interleukin (IL)-4 and IL-13 mRNA expression in lung tissue and protein in BAL fluid seen in OVA-treated mice were also inhibited by CysLT(1) receptor blockade. These data suggest an important role for cysteinyl leukotrienes in the pathogenesis of chronic allergic airway inflammation with fibrosis.


Subject(s)
Asthma/immunology , Asthma/pathology , Disease Models, Animal , Leukotrienes/physiology , Acetates/pharmacology , Acetates/therapeutic use , Acute Disease , Allergens , Analysis of Variance , Animals , Asthma/chemically induced , Asthma/drug therapy , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chronic Disease , Cyclopropanes , Drug Evaluation, Preclinical , Eosinophils/immunology , Eosinophils/pathology , Fibrosis , Glycoproteins/analysis , Glycoproteins/immunology , Goblet Cells/immunology , Goblet Cells/pathology , Hyperplasia , Inflammation , Leukotriene Antagonists/pharmacology , Leukotriene Antagonists/therapeutic use , Lysophospholipase , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Mice , Ovalbumin , Quinolines/pharmacology , Quinolines/therapeutic use , Respiratory Mechanics/drug effects , Sulfides
SELECTION OF CITATIONS
SEARCH DETAIL
...