Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Front Pharmacol ; 12: 627503, 2021.
Article in English | MEDLINE | ID: mdl-33833679

ABSTRACT

Nitric oxide (NO) is produced by a family of isoenzymes, nitric oxide synthases (NOSs), which all utilize L-arginine as substrate. The production of NO in the lung and airways can play a number of roles during lung development, regulates airway and vascular smooth muscle tone, and is involved in inflammatory processes and host defense. Altered L-arginine/NO homeostasis, due to the accumulation of endogenous NOS inhibitors and competition for substrate with the arginase enzymes, has been found to play a role in various conditions affecting the lung and in pulmonary diseases, such as asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), pulmonary hypertension, and bronchopulmonary dysplasia. Different therapeutic strategies to increase L-arginine levels or bioavailability are currently being explored in pre-clinical and clinical studies. These include supplementation of L-arginine or L-citrulline and inhibition of arginase.

2.
PLoS One ; 12(1): e0163614, 2017.
Article in English | MEDLINE | ID: mdl-28107345

ABSTRACT

BACKGROUND: Asthma is a chronic inflammatory disease characterized by airways hyper-responsiveness (AHR), reversible airway obstruction, and airway inflammation and remodeling. We previously showed that Syk modulates methacholine-induced airways contractility in naïve mice and in mice with allergic airways inflammation. We hypothesize that Syk plays a role in the pathogenesis of AHR; this was evaluated in a chronic 8-week mouse model of house dust mite (HDM)-induced allergic airways inflammation. METHODS: We used the Sykflox/flox//rosa26CreERT2 conditional Syk knock-out mice to assess the role of Syk prior to HDM exposure, and treated HDM-sensitized mice with the Syk inhibitor, GSK143, to evaluate its role in established allergic airways inflammation. Respiratory mechanics and methacholine (MCh)-responsiveness were assessed using the flexiVent® system. Lungs underwent bronchoalveolar lavage to isolate inflammatory cells or were frozen for determination of gene expression in tissues. RESULTS: MCh-induced AHR was observed following HDM sensitization in the Syk-intact (Sykflox/flox) and vehicle-treated BALB/c mice. MCh responsiveness was reduced to control levels in HDM-sensitized Sykdel/del mice and in BALB/c and Sykflox/flox mice treated with GSK143. Both Sykdel/del and GSK143-treated mice mounted appropriate immune responses to HDM, with HDM-specific IgE levels that were comparable to Sykflox/flox and vehicle-treated BALB/c mice. HDM-induced increases in bronchoalveolar lavage cell counts were attenuated in both Sykdel/del and GSK143-treated mice, due primarily to decreased neutrophil recruitment. Gene expression analysis of lung tissues revealed that HDM-induced expression of IL-17 and CXCL-1 was significantly attenuated in both Sykdel/del and GSK143-treated mice. CONCLUSION: Syk inhibitors may play a role in the management of neutrophilic asthma.


Subject(s)
Bronchial Hyperreactivity/enzymology , Disease Models, Animal , Syk Kinase/metabolism , Animals , Mice , Mice, Inbred BALB C , Mice, Transgenic , Pyroglyphidae/immunology , Syk Kinase/genetics
3.
J Appl Physiol (1985) ; 118(10): 1229-33, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25979935

ABSTRACT

L-arginine metabolism is important in the maintenance of airway tone. Shift of metabolism from the nitric oxide synthase to arginase pathways contributes to the increased airway responsiveness in asthma. We tested the hypothesis that systemic levels of L-arginine metabolites are biomarkers reflective of airway dysfunction. We used a mouse model of acute allergic airway inflammation to OVA that manifests with significant airway hyperresponsiveness to methacholine. To determine tissue arginase activity in vivo, the isotopic enrichment of an infused L-arginine stable isotope and its product amino acid L-ornithine were measured in lung and airway homogenates using liquid chromatography-tandem mass spectrometry. Tissue and plasma concentrations of other L-arginine metabolites, including L-citrulline and symmetric and asymmetric dimethylarginine, were measured and correlated with lung arginase activity and methacholine responsiveness of the airways. The effectiveness of intratracheal instillation of an arginase inhibitor (boronoethylcysteine) on pulmonary arginase activity and circulating concentrations of L-arginine metabolites was also studied. We demonstrate that 1) plasma indexes of L-arginine bioavailability and impairment of nitric oxide synthase function correlate with airway responsiveness to methacholine; 2) plasma levels of L-ornithine predict in vivo pulmonary arginase activity and airway function; and 3) acute arginase inhibition reduces in vivo pulmonary arginase activity to control levels and normalizes plasma L-ornithine, but not L-arginine, bioavailability in this model. We conclude that plasma L-ornithine may be useful as a systemic biomarker to predict responses to therapeutic interventions targeting airway arginase in asthma.


Subject(s)
Arginine/blood , Asthma/blood , Asthma/physiopathology , Respiratory System/physiopathology , Animals , Arginase/antagonists & inhibitors , Arginase/metabolism , Arginine/analogs & derivatives , Bronchoconstrictor Agents/pharmacology , Citrulline/blood , Enzyme Inhibitors/pharmacology , Female , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase/antagonists & inhibitors , Ornithine/blood , Ovalbumin
4.
Am J Physiol Lung Cell Mol Physiol ; 308(6): L503-10, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25595650

ABSTRACT

Arginase is an enzyme that limits substrate L-arginine bioavailability for the production of nitric oxide by the nitric oxide synthases and produces L-ornithine, which is a precursor for collagen formation and tissue remodeling. We studied the pulmonary vascular effects of arginase inhibition in an established model of repeated systemic bleomycin sulfate administration in neonatal rats that results in pulmonary hypertension and lung injury mimicking the characteristics typical of bronchopulmonary dysplasia. We report that arginase expression is increased in the lungs of bleomycin-exposed neonatal rats and that treatment with the arginase inhibitor amino-2-borono-6-hexanoic acid prevented the bleomycin-induced development of pulmonary hypertension and deposition of collagen. Arginase inhibition resulted in increased L-arginine and L-arginine bioavailability and increased pulmonary nitric oxide production. Arginase inhibition also normalized the expression of inducible nitric oxide synthase, and reduced bleomycin-induced nitrative stress while having no effect on bleomycin-induced inflammation. Our data suggest that arginase is a promising target for therapeutic interventions in neonates aimed at preventing lung vascular remodeling and pulmonary hypertension.


Subject(s)
Aminocaproates/pharmacology , Antibiotics, Antineoplastic/adverse effects , Arginase/antagonists & inhibitors , Bleomycin/adverse effects , Boron Compounds/pharmacology , Collagen/metabolism , Hypertension, Pulmonary , Lung/enzymology , Vascular Remodeling/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Arginase/metabolism , Arginine/metabolism , Bleomycin/pharmacology , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/enzymology , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/prevention & control , Disease Models, Animal , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/prevention & control , Lung/pathology , Lung Injury/chemically induced , Lung Injury/enzymology , Lung Injury/pathology , Lung Injury/prevention & control , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley
5.
Immunol Allergy Clin North Am ; 34(4): 767-75, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25282289

ABSTRACT

Nitric oxide (NO) is important in the regulation of airway tone and airway responsiveness. Alterations in the L-arginine metabolism resulting in reduced availability of the substrate L-arginine for NO synthases, as well as the presence of NO synthase inhibitors such as asymmetric dimethylarginine, contribute to the reduced NO formation and airway dysfunction in asthma. Therapeutic interventions aiming to modulate the impaired L-arginine metabolism may help correct the enhanced airway tone and responsiveness in asthma.


Subject(s)
Arginine/analogs & derivatives , Asthma/metabolism , Bronchial Hyperreactivity/immunology , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Anti-Asthmatic Agents/therapeutic use , Arginine/metabolism , Asthma/drug therapy , Bronchial Hyperreactivity/drug therapy , Humans , Molecular Targeted Therapy
6.
Int J Mol Sci ; 15(4): 6062-71, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24727374

ABSTRACT

L-arginine metabolism including the nitric oxide (NO) synthase and arginase pathways is important in the maintenance of airways function. We have previously reported that accumulation of asymmetric dimethylarginine (ADMA) in airways, resulting in changes in L-arginine metabolism, contributes to airways obstruction in asthma and cystic fibrosis. Herein, we assessed L-arginine metabolism in airways of patients with chronic obstructive pulmonary disease (COPD). Lung function testing, measurement of fractional exhaled NO (FeNO) and sputum NO metabolites, as well as quantification of L-arginine metabolites (L-arginine, L-ornithine, L-citrulline, ADMA and symmetric dimethylarginine) using liquid chromatography-mass spectrometry (LC-MS) were performed. Concentrations of L-ornithine, the product of arginase activity, correlated directly with L-arginine and ADMA sputum concentrations. FeNO correlated directly with pre- and post-bronchodilator forced expiratory volume in one second (FEV1). Sputum arginase activity correlated inversely with total NO metabolite (NOx) and nitrite concentrations in sputum, and with pre- and post-bronchodilator FEV1. These findings suggest that ADMA in COPD airways results in a functionally relevant shift of L-arginine breakdown by the NO synthases towards the arginase pathway, which contributes to airway obstruction in these patients.


Subject(s)
Arginine/analogs & derivatives , Arginine/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Tandem Mass Spectrometry , Aged , Arginase/metabolism , Arginine/analysis , Chromatography, High Pressure Liquid , Female , Forced Expiratory Volume , Humans , Lung/physiology , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Ornithine/analysis , Pulmonary Disease, Chronic Obstructive/pathology , Sputum/enzymology
7.
Curr Stem Cell Res Ther ; 9(4): 319-29, 2014.
Article in English | MEDLINE | ID: mdl-24588087

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) have become the focus of intense research effort over the past 10 years, in an effort to harness their regenerative and immune-modulating capacity for a variety of clinical conditions. In Acute Respiratory Distress Syndrome (ARDS), pre-clinical studies point towards a therapy that modulates multiple aspects of a complex disease process. Almost universally, these cells have demonstrated an immune modulating phenotype, balancing protective host responses with a reduction in damaging inflammation, while enhancing bacterial killing. MSCs also lead to more efficient tissue repair, and MSC-mediated lung tissue repair and regeneration after ARDS are some of the exciting clinical prospects. Recent investigation into the role of endogenous MSCs has led to new insights into MSC physiology and its role in regulating the immune system. However, significant deficits remain in our knowledge regarding the mechanisms of action of MSCs, their efficacy in relevant pre-clinical models, and their safety in critically ill patients. These gaps need to be addressed before the enormous therapeutic potential of stem cells for ALI/ARDS can be realized.


Subject(s)
Mesenchymal Stem Cell Transplantation , Respiratory Distress Syndrome/therapy , Animals , Humans , Mesenchymal Stem Cells/physiology , Regenerative Medicine
9.
Toxicol Rep ; 1: 145-156, 2014.
Article in English | MEDLINE | ID: mdl-28962235

ABSTRACT

Epidemiological and toxicological studies have suggested that the health effects associated with exposure to particulate matter (PM) are related to the different physicochemical properties of PM. These effects occur through the initiation of differential cellular responses including: the induction of antioxidant defenses, proinflammatory responses, and ultimately cell death. The main objective of this study was to investigate the effects of size-fractionated ambient PM on epithelial cells in relation to their physicochemical properties. Concentrated ambient PM was collected on filters for three size fractions: coarse (aerodynamic diameter [AD] 2.5-10 µm), fine (0.15-2.5 µm), and quasi-ultrafine (<0.2 µm), near a busy street in Toronto, Ontario, Canada. Filters were extracted and analyzed for chemical composition and redox activity. Chemical analyses showed that the coarse, fine, and quasi-ultrafine particles were comprised primarily of metals, water-soluble species, and organic compounds, respectively. The highest redox activity was observed for fine PM. After exposure of A549 cells to PM (10-100 µg/ml) for 4 h, activation of antioxidant, proinflammatory and cytotoxic responses were assessed by determining the expression of heme oxygenase (HMOX-1, mRNA), interleukin-8 (IL-8, mRNA), and metabolic activity of the cells, respectively. All three size fractions induced mass-dependent antioxidant, proinflammatory, and cytotoxic responses to different degrees. Quasi-ultrafine PM caused significant induction of HMOX-1 at the lowest exposure dose. Correlation analyses with chemical components suggested that the biological responses correlated mainly with transition metals and organic compounds for coarse and fine PM and with organic compounds for quasi-ultrafine PM. Overall, the observed biological responses appeared to be related to the combined effects of size and chemical composition and thus both of these physicochemical properties should be considered when explaining PM toxicity.

11.
J Am Heart Assoc ; 2(3): e000212, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23782920

ABSTRACT

BACKGROUND: Short-term exposures to fine (<2.5 µm aerodynamic diameter) ambient particulate-matter (PM) have been related with increased blood pressure (BP) in controlled-human exposure and community-based studies. However, whether coarse (2.5 to 10 µm) PM exposure increases BP is uncertain. Recent observational studies have linked PM exposures with blood DNA hypomethylation, an epigenetic alteration that activates inflammatory and vascular responses. No experimental evidence is available to confirm those observational data and demonstrate the relations between PM, hypomethylation, and BP. METHODS AND RESULTS: We conducted a cross-over trial of controlled-human exposure to concentrated ambient particles (CAPs). Fifteen healthy adult participants were exposed for 130 minutes to fine CAPs, coarse CAPs, or HEPA-filtered medical air (control) in randomized order with ≥2-week washout. Repetitive-element (Alu, long interspersed nuclear element-1 [LINE-1]) and candidate-gene (TLR4, IL-12, IL-6, iNOS) blood methylation, systolic and diastolic BP were measured pre- and postexposure. After adjustment for multiple comparisons, fine CAPs exposure lowered Alu methylation (ß-standardized=-0.74, adjusted-P=0.03); coarse CAPs exposure lowered TLR4 methylation (ß-standardized=-0.27, adjusted-P=0.04). Both fine and coarse CAPs determined significantly increased systolic BP (ß=2.53 mm Hg, P=0.001; ß=1.56 mm Hg, P=0.03, respectively) and nonsignificantly increased diastolic BP (ß=0.98 mm Hg, P=0.12; ß=0.82 mm Hg, P=0.11, respectively). Decreased Alu and TLR4 methylation was associated with higher postexposure DBP (ß-standardized=0.41, P=0.04; and ß-standardized=0.84, P=0.02; respectively). Decreased TLR4 methylation was associated with higher postexposure SBP (ß-standardized=1.45, P=0.01). CONCLUSIONS: Our findings provide novel evidence of effects of coarse PM on BP and confirm effects of fine PM. Our results provide the first experimental evidence of PM-induced DNA hypomethylation and its correlation to BP.


Subject(s)
Blood Pressure , DNA Methylation , Environmental Exposure , Particulate Matter/adverse effects , Adolescent , Adult , Blood Pressure/drug effects , Blood Pressure/genetics , Cross-Over Studies , Double-Blind Method , Female , Humans , Hypertension/etiology , Male , Middle Aged , Young Adult
12.
Am J Respir Cell Mol Biol ; 48(6): 694-702, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23470627

ABSTRACT

Up-regulation of arginase contributes to airways hyperresponsiveness (AHR) in asthma by reducing L-arginine bioavailability for the nitric oxide (NO) synthase isozymes. The product of arginase activity, L-ornithine, can be metabolized into polyamines by ornithine decarboxylase. We tested the hypothesis that increases in L-ornithine-derived polyamines contribute to AHR in mouse models of allergic airways inflammation. After measuring significantly increased polyamine levels in sputum samples from human subjects with asthma after allergen challenge, we used acute and subacute ovalbumin sensitization and challenge mouse models of allergic airways inflammation and naive mice to investigate the relationship of AHR to methacholine and polyamines in the lung. We found that spermine levels were elevated significantly in lungs from the acute model, which exhibits robust AHR, but not in the subacute murine model of asthma, which does not develop AHR. Intratracheal administration of spermine significantly augmented airways responsiveness to methacholine in both naive mice and mice with subacute airways inflammation, and reduced nitrite/nitrate levels in lung homogenates, suggesting that the AHR developed as a consequence of inhibition of constitutive NO production in the airways. Chronic inhibition of polyamine synthesis using an ornithine decarboxylase inhibitor significantly reduced polyamine levels, restored nitrite/nitrate levels to normal, and abrogated the AHR to methacholine in the acute model of allergic airways inflammation. We demonstrate that spermine increases airways responsiveness to methacholine, likely through inhibition of constitutive NO synthesis. Thus, inhibition of polyamine production may represent a new therapeutic target to treat airway obstruction in allergic asthma.


Subject(s)
Asthma/pathology , Hypersensitivity/pathology , Ornithine/metabolism , Polyamines/metabolism , Adolescent , Adult , Animals , Asthma/drug therapy , Asthma/immunology , Asthma/metabolism , Disease Models, Animal , Eflornithine/pharmacology , Female , Humans , Hypersensitivity/drug therapy , Hypersensitivity/immunology , Hypersensitivity/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lung/drug effects , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Methacholine Chloride/metabolism , Methacholine Chloride/pharmacology , Mice , Middle Aged , Nitrates/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase/metabolism , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase Inhibitors , Ovalbumin/adverse effects , Ovalbumin/immunology , Polyamines/antagonists & inhibitors , Spermine/administration & dosage , Spermine/adverse effects , Spermine/pharmacology , Sputum/metabolism , Young Adult
13.
J Allergy Clin Immunol ; 131(2): 512-20.e1-10, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22981792

ABSTRACT

BACKGROUND: Asthma is a chronic inflammatory disease characterized by airways hyperresponsiveness (AHR), reversible airflow obstruction, airway remodeling, and episodic exacerbations caused by air pollutants, such as particulate matter (PM; PM <2.5 µm in diameter [PM(2.5)]) and ozone (O(3)). Spleen tyrosine kinase (Syk), an immunoregulatory kinase, has been implicated in the pathogenesis of asthma. OBJECTIVE: We sought to evaluate the effect of Syk inhibition on AHR in a chronic mouse model of allergic airways inflammation and pollutant exposure. METHODS: We used a 12-week chronic ovalbumin (OVA) sensitization and challenge mouse model of airways inflammation followed by exposure to PM(2.5) plus O(3). Respiratory mechanics and methacholine (MCh) responsiveness were assessed by using the flexiVent system. The Syk inhibitor NVP-QAB-205 was nebulized intratracheally by using a treatment-based protocol 15 minutes before assessment of MCh responsiveness. RESULTS: Syk expression increased significantly in the airway epithelia of OVA-sensitized and OVA-challenged (OVA/OVA) mice compared with OVA-sensitized but PBS-challenged (OVA/PBS) control mice. OVA/OVA mice exhibited AHR to MCh, which was attenuated by a single administration of NVP-QAB-205 (0.3 and 3 mg/kg). PM(2.5) plus O(3) significantly augmented AHR to MCh in the OVA/OVA mice, which was abrogated by NVP-QAB-205. Total inflammatory cell counts were significantly higher in the bronchoalveolar lavage fluid from OVA/OVA than OVA/PBS mice and were unaffected by PM(2.5) plus O(3) or NVP-QAB-205. CONCLUSION: NVP-QAB-205 reduced AHR and the enhanced response to PM(2.5) plus O(3) to normal levels in an established model of chronic allergic airways inflammation, suggesting that Syk inhibitors have promise as a therapy for asthma.


Subject(s)
Air Pollutants/adverse effects , Asthma/drug therapy , Bronchial Hyperreactivity/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Asthma/chemically induced , Asthma/metabolism , Asthma/pathology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Bronchoalveolar Lavage Fluid/chemistry , Cells, Cultured , Disease Models, Animal , Female , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C , Ovalbumin/pharmacology , Protein-Tyrosine Kinases/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Syk Kinase , Vascular Endothelial Growth Factor A/metabolism
14.
Inhal Toxicol ; 24(3): 161-71, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22356274

ABSTRACT

CONTEXT: Strong epidemiological evidence exists linking particulate matter (PM) exposures with hospital admissions of individuals for cardiopulmonary symptoms. The PM size is important in influencing the extent of infiltration into the respiratory tract and systemic circulation and directs the differential physiological impacts. OBJECTIVE: To investigate the differential effects of the quasi-ultrafine (PM(0.2)), fine (PM(0.15-2.5)), and coarse PM (PM(2.5-10)) size fractions on pulmonary and cardiac function. METHODS: Female BALB/c mice were exposed to HEPA-filtered laboratory air or concentrated coarse, fine, or quasi-ultrafine PM using Harvard Ambient Particle Concentrators in conjunction with our nose-only exposure system. These exposures were conducted as part of the "Health Effects of Aerosols in Toronto (HEAT)" campaign. Following a 4 h exposure, mice underwent assessment of respiratory function and recording of electrocardiograms using the flexiVent® system. RESULTS: Exposure to coarse and fine PM resulted in a significant reduction in quasistatic compliance of the lung. Baseline total respiratory resistance and maximum responsiveness to methacholine were augmented after coarse PM exposures but were not affected by quasi-ultrafine PM exposures. In contrast, quasi-ultrafine PM alone had a significant effect on heart rate and in reducing heart rate variability. CONCLUSION: These findings indicate that coarse and fine PM influence lung function and airways responsiveness, while ultrafine PM can perturb cardiac function. This study supports the hypothesis that coarse and fine PM exerts its predominant physiologic effects at the site of deposition in the airways, whereas ultrafine PM likely crosses the alveolar epithelial barrier into the systemic circulation to affect cardiovascular function.


Subject(s)
Airway Resistance/drug effects , Heart Rate/drug effects , Particulate Matter/toxicity , Respiration/drug effects , Animals , Bronchoalveolar Lavage Fluid/cytology , Electrocardiography/drug effects , Female , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C
15.
Am J Respir Cell Mol Biol ; 46(4): 532-40, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22108300

ABSTRACT

The increase of airway smooth muscle (ASM) mass in asthma results from hypertrophic and hyperplastic stimuli, and leads to an increase in cellular contractile proteins. However, little evidence correlates the relative contributions of hypertrophic and hyperplastic muscle with functional effects on airway resistance. We performed a ventilator-based assessment of respiratory mechanics and responsiveness to methacholine in a murine model of acute (3-week) ovalbumin (OVA)-induced airway inflammation, compared with a chronic (12-week) model. We correlated functional changes in airways Newtonian resistance (RN), peripheral tissue damping (G), and elastance (H) with the relative contributions of proliferation, hypertrophy, and apoptosis to increased ASM mass. Immunohistochemical analyses of treated (OVA-sensitized and OVA-challenged; OVA/OVA) and control (OVA-sensitized and saline-challenged; OVA/PBS) murine lungs showed an increase in ASM area in chronic, but not acute, OVA/OVA-treated mice that correlated positively with increased airway resistance to methacholine. Acute OVA/OVA-treated ASM exhibited an increase in proliferation with diminished apoptosis, which resolved in the chronic OVA/OVA model. Chronic OVA/OVA-treated ASM exhibited hypertrophy. Distinct temporal differences exist in the response of murine airways to antigenic challenge. We report that ASM proliferation and diminished apoptosis occur during the acute phase, followed by the development of smooth muscle hypertrophy and an increased muscle mass with chronic challenge, that correlate strongly with increased airway Newtonian resistance. The identification of a functionally relevant hypertrophic bronchial muscle mass highlights the possibility of regulating airway muscle hypertrophy as a novel therapeutic target in asthma.


Subject(s)
Asthma/physiopathology , Muscle, Smooth/pathology , Respiratory Hypersensitivity/physiopathology , Airway Resistance , Animals , Apoptosis/immunology , Asthma/immunology , Cell Proliferation , Disease Models, Animal , Female , Hypertrophy/physiopathology , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C , Muscle, Smooth/drug effects , Muscle, Smooth/immunology , Ovalbumin/adverse effects , Ovalbumin/immunology , Respiratory Hypersensitivity/immunology
16.
J Pharmacol Exp Ther ; 340(2): 277-85, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22031919

ABSTRACT

Spleen tyrosine kinase (Syk) is an immunoregulatory tyrosine kinase that was identified originally in leukocytes. It is a key regulator of innate immunity as well as hematopoietic cell differentiation and proliferation. A role for Syk in regulating normal cellular functions in nonhematopoietic cells is increasingly recognized. We have shown previously robust Syk expression in airway epithelium, where it regulates the early inflammatory response to human rhinovirus (HRV) infections, and HRV cell entry by clathrin-mediated endocytosis. To test the hypothesis that Syk plays a role in modulating airway epithelial cell proliferation, migration, and production of vascular endothelial growth factor and interleukin-8, we studied the BEAS-2B human bronchial epithelial cell line and primary human airway epithelia from normal and asthmatic donors using Syk-specific pharmacologic inhibitors and small interfering RNA. Using an in vitro "wounding" model, we demonstrated significant impairment of "wound" closure after treatment with the Syk inhibitors N4-(2,2-dimethyl-3-oxo-4H-pyrid[1,4]oxazin-6-yl)-5-fluoro-N2-(3,4,5-trimethoxyphenyl)-2,4-pyrimidinediamine (R406) and 2-[7-(3,4-dimethoxyphenyl)-imidazo[1,2-c]pyrimidin-5-ylamino]-nicotinamide dihydrochloride (BAY61-3606), overexpression of the kinase-inactive Syk(K396R) mutant, and Syk knockdown by small interfering RNA. HRV infection also impaired wound healing, an effect that was partly Syk-dependent because wound healing was impaired further when HRV infection occurred in the presence of Syk inhibition. Further investigation of potential regulatory mechanisms revealed that inhibition of Syk suppressed HRV-induced vascular endothelial growth factor expression while promoting the activation of caspase-3, a mediator of epithelial cell apoptosis. Together, these results indicate that Syk plays a role in promoting epithelial cell proliferation and migration, while mitigating the effects of apoptosis.


Subject(s)
Cell Movement/physiology , Cell Proliferation/drug effects , Epithelial Cells/physiology , Interleukin-8/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Protein-Tyrosine Kinases/physiology , Rhinovirus , Vascular Endothelial Growth Factor A/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Asthma/metabolism , Caspase 3/metabolism , Cell Line , Cell Movement/drug effects , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/virology , Gene Expression/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Small Interfering/genetics , Respiratory Mucosa/cytology , Syk Kinase , Wound Healing/drug effects
17.
Am J Respir Crit Care Med ; 184(7): 779-85, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21719758

ABSTRACT

RATIONALE: Asymmetric dimethylarginine (ADMA) is an endogenous nitric oxide synthase (NOS) inhibitor that competes with L-arginine for binding to NOS. It has been suggested that ADMA contributes to inflammation, collagen deposition, nitrosative stress, and lung function in murine models. OBJECTIVES: To test the hypothesis that ADMA is increased in asthma and that NOS inhibition by ADMA contributes to airways obstruction. METHODS: We assessed alterations of L-arginine, ADMA, and symmetric dimethylarginine (SDMA) levels in a murine model of allergic airways inflammation using LC-tandem mass spectrometry. Based on the levels of ADMA observed in the murine model, we further tested the direct effects of nebulized inhaled ADMA on airways responsiveness in naive control mice. We also assessed alterations of L-arginine, ADMA, and SDMA in humans in adult lung specimens and sputum samples from pediatric patients with asthma. MEASUREMENTS AND MAIN RESULTS: ADMA was increased in lungs from the murine model of allergic airways inflammation. Exogenous administration of ADMA to naive mice, at doses consistent with the levels observed in the allergically inflamed lungs, resulted in augmentation of the airways responsiveness to methacholine. ADMA levels were also increased in human asthma lungs and sputum samples. CONCLUSIONS: ADMA levels are increased in asthma and contribute to NOS-related pathophysiology.


Subject(s)
Arginine/analogs & derivatives , Arginine/metabolism , Asthma/metabolism , Adolescent , Animals , Biomarkers/metabolism , Bronchial Hyperreactivity/metabolism , Case-Control Studies , Child , Female , Humans , Male , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase/metabolism , Sputum/metabolism
18.
Respir Res ; 12: 19, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21291525

ABSTRACT

BACKGROUND: Arginase overexpression contributes to airways hyperresponsiveness (AHR) in asthma. Arginase expression is further augmented in cigarette smoking asthmatics, suggesting that it may be upregulated by environmental pollution. Thus, we hypothesize that arginase contributes to the exacerbation of respiratory symptoms following exposure to air pollution, and that pharmacologic inhibition of arginase would abrogate the pollution-induced AHR. METHODS: To investigate the role of arginase in the air pollution-induced exacerbation of airways responsiveness, we employed two murine models of allergic airways inflammation. Mice were sensitized to ovalbumin (OVA) and challenged with nebulized PBS (OVA/PBS) or OVA (OVA/OVA) for three consecutive days (sub-acute model) or 12 weeks (chronic model), which exhibit inflammatory cell influx and remodeling/AHR, respectively. Twenty-four hours after the final challenge, mice were exposed to concentrated ambient fine particles plus ozone (CAP+O3), or HEPA-filtered air (FA), for 4 hours. After the CAP+O3 exposures, mice underwent tracheal cannulation and were treated with an aerosolized arginase inhibitor (S-boronoethyl-L-cysteine; BEC) or vehicle, immediately before determination of respiratory function and methacholine-responsiveness using the flexiVent®. Lungs were then collected for comparison of arginase activity, protein expression, and immunohistochemical localization. RESULTS: Compared to FA, arginase activity was significantly augmented in the lungs of CAP+O3-exposed OVA/OVA mice in both the sub-acute and chronic models. Western blotting and immunohistochemical staining revealed that the increased activity was due to arginase 1 expression in the area surrounding the airways in both models. Arginase inhibition significantly reduced the CAP+O3-induced increase in AHR in both models. CONCLUSIONS: This study demonstrates that arginase is upregulated following environmental exposures in murine models of asthma, and contributes to the pollution-induced exacerbation of airways responsiveness. Thus arginase may be a therapeutic target to protect susceptible populations against the adverse health effects of air pollution, such as fine particles and ozone, which are two of the major contributors to smog.


Subject(s)
Arginase/metabolism , Asthma/etiology , Bronchial Hyperreactivity/etiology , Bronchoconstriction , Lung/drug effects , Ozone/toxicity , Particulate Matter/toxicity , Animals , Arginase/antagonists & inhibitors , Asthma/enzymology , Asthma/immunology , Asthma/physiopathology , Blotting, Western , Boronic Acids/pharmacology , Bronchial Hyperreactivity/enzymology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Bronchial Hyperreactivity/prevention & control , Bronchial Provocation Tests , Bronchoconstriction/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Immunohistochemistry , Inflammation Mediators/metabolism , Inhalation Exposure , Lung/enzymology , Lung/immunology , Lung/physiopathology , Mice , Mice, Inbred BALB C , Ovalbumin , Oxidative Stress/drug effects , Up-Regulation
19.
Am J Respir Crit Care Med ; 183(10): 1363-8, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21278301

ABSTRACT

RATIONALE: Airway nitric oxide is reduced in cystic fibrosis airways. Asymmetric dimethylarginine is an endogenous nitric oxide synthase inhibitor that may contribute to nitric oxide deficiency in cystic fibrosis. OBJECTIVES: To test the hypothesis that asymmetric dimethylarginine is increased in cystic fibrosis and contributes to nitric oxide deficiency and airway obstruction. METHODS: The concentrations of asymmetric dimethylarginine, symmetric dimethylarginine, and l-arginine were measured in sputum of clinically stable patients with cystic fibrosis, in patients with cystic fibrosis before and after treatment for a pulmonary exacerbation, and in healthy control subjects, using liquid chromatography-tandem mass spectrometry. MEASUREMENTS AND MAIN RESULTS: Asymmetric dimethylarginine was increased in cystic fibrosis compared with control sputum, and the l-arginine/asymmetric dimethylarginine ratio was decreased. Symmetric dimethylarginine exceeded asymmetric dimethylarginine concentrations in control sputum, but this ratio was reversed in cystic fibrosis. Treatment for pulmonary exacerbation resulted in a decrease in sputum asymmetric dimethylarginine and an improved l-arginine/asymmetric dimethylarginine ratio. The treatment-related decrease in asymmetric dimethylarginine correlated significantly with an increase in sputum nitric oxide metabolites and improvement in pulmonary function. The activity of the asymmetric dimethylarginine-metabolizing enzyme, dimethylarginine dimethylaminohydrolase, was higher in cystic fibrosis sputum before rather than after treatment, suggesting that the accumulation of asymmetric dimethylarginine is caused by increased production, not decreased degradation, of asymmetric dimethylarginine. CONCLUSIONS: Asymmetric dimethylarginine is increased in cystic fibrosis airways and may contribute to airway obstruction in patients with cystic fibrosis by reducing nitric oxide formation.


Subject(s)
Arginine/analogs & derivatives , Cystic Fibrosis/metabolism , Nitric Oxide/metabolism , Respiratory System/metabolism , Adolescent , Airway Obstruction/complications , Arginine/metabolism , Child , Chromatography, Liquid , Cystic Fibrosis/complications , Female , Humans , Lung/metabolism , Male , Sputum/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...