Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Am J Respir Crit Care Med ; 201(3): 313-324, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31613648

ABSTRACT

Rationale: Despite therapeutic progress in treating cystic fibrosis (CF) airway disease, airway inflammation with associated mucociliary dysfunction remains largely unaddressed. Inflammation reduces the activity of apically expressed large-conductance Ca2+-activated and voltage-dependent K+ (BK) channels, critical for mucociliary function in the absence of CFTR (CF transmembrane conductance regulator).Objectives: To test losartan as an antiinflammatory therapy in CF using CF human bronchial epithelial cells and an ovine model of CF-like airway disease.Methods: Losartan's antiinflammatory effectiveness to rescue BK activity and thus mucociliary function was tested in vitro using primary, fully redifferentiated human airway epithelial cells homozygous for F508del and in vivo using a previously validated, now expanded pharmacologic sheep model of CF-like, inflammation-associated mucociliary dysfunction.Measurements and Main Results: Nasal scrapings from patients with CF showed that neutrophilic inflammation correlated with reduced expression of LRRC26 (leucine rich repeat containing 26), the γ subunit mandatory for BK function in the airways. TGF-ß1 (transforming growth factor ß1), downstream of neutrophil elastase, decreased mucociliary parameters in vitro. These were rescued by losartan at concentrations achieved by nebulization in the airway and oral application in the bloodstream: BK dysfunction recovered acutely and over time (the latter via an increase in LRRC26 expression), ciliary beat frequency and airway surface liquid volume improved, and mucus hyperconcentration and cellular inflammation decreased. These effects did not depend on angiotensin receptor blockade. Expanding on a validated and published nongenetic, CF-like sheep model, ewes inhaled CFTRinh172 and neutrophil elastase for 3 days, which resulted in prolonged tracheal mucus velocity reduction, mucus hyperconcentration, and increased TGF-ß1. Nebulized losartan rescued both mucus transport and mucus hyperconcentration and reduced TGF-ß1.Conclusions: Losartan effectively reversed CF- and inflammation-associated mucociliary dysfunction, independent of its angiotensin receptor blockade.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Cystic Fibrosis/physiopathology , Losartan/pharmacology , Mucociliary Clearance/drug effects , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Bronchi/cytology , Cells, Cultured , Cystic Fibrosis/drug therapy , Disease Models, Animal , Epithelial Cells , Female , Humans , Inflammation/physiopathology , Losartan/therapeutic use , Sheep
2.
PLoS One ; 14(11): e0224764, 2019.
Article in English | MEDLINE | ID: mdl-31697733

ABSTRACT

The purpose of this investigation was to ascertain whether nitric oxide (NO) released into the circulation by a noninvasive technology called whole body periodic acceleration (WBPA) could increase mucociliary clearance (MCC). It was based on observations by others that nitric oxide donor drugs increase ciliary beat frequency of nasal epithelium without increasing mucociliary clearance. Tracheal mucous velocity (TMV), a reflection of MCC, was measured in sheep after 1-hour treatment of WBPA and repeated after pretreatment with the NO synthase inhibitor, L-NAME to demonstrated action of NO. Aerosolized human neutrophil elastase (HNE) was administered to sheep to suppress TMV as might occur in cystic fibrosis and other inflammatory lung diseases. WBPA increased TMV to a peak of 136% of baseline 1h after intervention, an effect blocked by L-NAME. HNE reduced TMV to 55% of baseline but slowing was reversed by WBPA, protection lost in the presence of L-NAME. NO released into the circulation from eNOS by WBPA can acutely access airway epithelium for improving MCC slowed in cystic fibrosis and other inflammatory lung diseases as a means of enhancing host defense against pathogens.


Subject(s)
Acceleration , Consciousness , Mucociliary Clearance , Sheep/physiology , Aerosols , Animals , Humans , Leukocyte Elastase/metabolism , Mucus/metabolism , Nitric Oxide/metabolism , Trachea/physiology
3.
Sci Transl Med ; 11(486)2019 04 03.
Article in English | MEDLINE | ID: mdl-30944166

ABSTRACT

Although destructive airway disease is evident in young children with cystic fibrosis (CF), little is known about the nature of the early CF lung environment triggering the disease. To elucidate early CF pulmonary pathophysiology, we performed mucus, inflammation, metabolomic, and microbiome analyses on bronchoalveolar lavage fluid (BALF) from 46 preschool children with CF enrolled in the Australian Respiratory Early Surveillance Team for Cystic Fibrosis (AREST CF) program and 16 non-CF disease controls. Total airway mucins were elevated in CF compared to non-CF BALF irrespective of infection, and higher densities of mucus flakes containing mucin 5B and mucin 5AC were observed in samples from CF patients. Total mucins and mucus flakes correlated with inflammation, hypoxia, and oxidative stress. Many CF BALFs appeared sterile by culture and molecular analyses, whereas other samples exhibiting bacterial taxa associated with the oral cavity. Children without computed tomography-defined structural lung disease exhibited elevated BALF mucus flakes and neutrophils, but little/no bacterial infection. Although CF mucus flakes appeared "permanent" because they did not dissolve in dilute BALF matrix, they could be solubilized by a previously unidentified reducing agent (P2062), but not N-acetylcysteine or deoxyribonuclease. These findings indicate that early CF lung disease is characterized by an increased mucus burden and inflammatory markers without infection or structural lung disease and suggest that mucolytic and anti-inflammatory agents should be explored as preventive therapy.


Subject(s)
Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Lung/metabolism , Lung/pathology , Mucus/metabolism , Animals , Biomarkers/metabolism , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Inflammation/pathology , Lung/microbiology , Male , Microbiota , Sheep
4.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L192-L205, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28982737

ABSTRACT

In cystic fibrosis (CF) lungs, epithelial Na+ channel (ENaC) hyperactivity causes a reduction in airway surface liquid volume, leading to decreased mucocilliary clearance, chronic bacterial infection, and lung damage. Inhibition of ENaC is an attractive therapeutic option. However, ENaC antagonists have failed clinically because of off-target effects in the kidney. The S18 peptide is a naturally occurring short palate lung and nasal epithelial clone 1 (SPLUNC1)-derived ENaC antagonist that restores airway surface liquid height for up to 24 h in CF human bronchial epithelial cultures. However, its efficacy and safety in vivo are unknown. To interrogate the potential clinical efficacy of S18, we assessed its safety and efficacy using human airway cultures and animal models. S18-mucus interactions were tested using superresolution microscopy, quartz crystal microbalance with dissipation, and confocal microscopy. Human and murine airway cultures were used to measure airway surface liquid height. Off-target effects were assessed in conscious mice and anesthetized rats. Morbidity and mortality were assessed in the ß-ENaC-transgenic (Tg) mouse model. Restoration of normal mucus clearance was measured in cystic fibrosis transmembrane conductance regulator inhibitor 172 [CFTR(inh)-172]-challenged sheep. We found that S18 does not interact with mucus and rapidly penetrated dehydrated CF mucus. Compared with amiloride, an early generation ENaC antagonist, S18 displayed a superior ability to slow airway surface liquid absorption, reverse CFTR(inh)-172-induced reduction of mucus transport, and reduce morbidity and mortality in the ß-ENaC-Tg mouse, all without inducing any detectable signs of renal toxicity. These data suggest that S18 is the first naturally occurring ENaC antagonist to show improved preclinical efficacy in animal models of CF with no signs of renal toxicity.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Glycoproteins/metabolism , Lung Diseases/drug therapy , Peptides/pharmacology , Phosphoproteins/metabolism , Respiratory Mucosa/drug effects , Animals , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Humans , Ion Transport , Lung Diseases/metabolism , Lung Diseases/pathology , Male , Mice , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/metabolism
5.
Am J Respir Crit Care Med ; 196(6): 734-744, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28481660

ABSTRACT

RATIONALE: Cystic fibrosis (CF) lung disease is caused by the loss of function of the cystic fibrosis transmembrane conductance regulator (CFTR) combined with hyperactivation of the epithelial sodium channel (ENaC). In the lung, ENaC is responsible for movement of sodium. Hyperactivation of ENaC, which creates an osmotic gradient that pulls fluid out of the airway, contributes to reduced airway hydration, causing mucus dehydration, decreased mucociliary clearance, and recurrent acute bacterial infections. ENaC represents a therapeutic target to treat all patients with CF independent of their underlying CFTR mutation. OBJECTIVES: To investigate the in vitro and in vivo efficacy of SPX-101, a peptide mimetic of the natural regulation of ENaC activity by short palate, lung, and nasal epithelial clone 1, known as SPLUNC1. METHODS: ENaC internalization by SPX-101 in primary human bronchial epithelial cells from healthy and CF donors was assessed by surface biotinylation and subsequent Western blot analysis. SPX-101's in vivo therapeutic effect was assessed by survival of ß-ENaC-transgenic mice, mucus transport in these mice, and mucus transport in a sheep model of CF. MEASUREMENTS AND MAIN RESULTS: SPX-101 binds selectively to ENaC and promotes internalization of the α-, ß-, and γ-subunits. Removing ENaC from the membrane with SPX-101 causes a significant decrease in amiloride-sensitive current. The peptide increases survival of ß-ENaC-transgenic mice to greater than 90% with once-daily dosing by inhalation. SPX-101 increased mucus transport in the ß-ENaC mouse model as well as the sheep model of CF. CONCLUSIONS: These data demonstrate that SPX-101 promotes durable reduction of ENaC membrane concentration, leading to significant improvements in mucus transport.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/therapeutic use , Cystic Fibrosis/drug therapy , Epithelial Sodium Channel Blockers/therapeutic use , Epithelial Sodium Channels/therapeutic use , Mucociliary Clearance/drug effects , Respiratory Mucosa/drug effects , Humans
6.
Am J Physiol Lung Cell Mol Physiol ; 304(11): L746-56, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23542952

ABSTRACT

Mucus clearance is an important component of the lung's innate defense system. A failure of this system brought on by mucus dehydration is common to both cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD). Mucus clearance rates are regulated by the volume of airway surface liquid (ASL) and by ciliary beat frequency (CBF). Chronic treatment with macrolide antibiotics is known to be beneficial to both CF and COPD patients. However, chronic macrolide usage may induce bacterial resistance. We have developed a novel macrolide, 2'-desoxy-9-(S)-erythromycylamine (GS-459755), that has significantly diminished antibiotic activity against Staphylococcus aureus, Streptococcus pneumonia, Moraxella catarrhalis, and Haemophilus influenzae. Since neutrophilia frequently occurs in chronic lung disease and human neutrophil elastase (HNE) induces mucus stasis by activating the epithelial sodium channel (ENaC), we tested the ability of GS-459755 to protect against HNE-induced mucus stasis. GS-459755 had no effect on HNE activity. However, GS-459755 pretreatment protected against HNE-induced ASL volume depletion in human bronchial epithelial cells (HBECs). The effect of GS-459755 on ASL volume was dose dependent (IC50 ~3.9 µM) and comparable to the antibacterial macrolide azithromycin (IC50 ~2.4 µM). Macrolides had no significant effect on CBF or on transepithelial water permeability. However, the amiloride-sensitive transepithelial voltage, a marker of ENaC activity, was diminished by macrolide pretreatment. We conclude that GS-459755 may limit HNE-induced activation of ENaC and may be useful for the treatment of mucus dehydration in CF and COPD without inducing bacterial resistance.


Subject(s)
Epithelial Sodium Channels/drug effects , Erythromycin/analogs & derivatives , Leukocyte Elastase/antagonists & inhibitors , Macrolides/pharmacology , Mucus/physiology , Azithromycin/pharmacology , Erythromycin/pharmacology , Humans , Leukocyte Elastase/metabolism , Mucus/drug effects , Respiratory Mucosa/drug effects , Respiratory System/metabolism
7.
Am J Physiol Lung Cell Mol Physiol ; 295(2): L240-63, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18487356

ABSTRACT

Airway disease currently causes most of the morbidity and mortality in patients with cystic fibrosis (CF). However, understanding the pathogenesis of CF lung disease and developing novel therapeutic strategies have been hampered by the limitations of current models. Although the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) has been targeted in mice, CF mice fail to develop lung or pancreatic disease like that in humans. In many respects, the anatomy, biochemistry, physiology, size, and genetics of pigs resemble those of humans. Thus pigs with a targeted CFTR gene might provide a good model for CF. Here, we review aspects of porcine airways and lung that are relevant to CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/mortality , Disease Models, Animal , Swine/genetics , Swine/metabolism , Animals , Cystic Fibrosis/complications , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Cystic Fibrosis/therapy , Humans , Mice , Pancreatic Diseases/complications , Pancreatic Diseases/genetics , Pancreatic Diseases/metabolism , Pancreatic Diseases/pathology , Pancreatic Diseases/therapy
8.
J Pharmacol Exp Ther ; 325(1): 77-88, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18218832

ABSTRACT

Amiloride improves mucociliary clearance (MC) by blocking airway epithelial sodium channels (ENaC) and expanding airway surface liquid (ASL). However, the low potency and rapid absorption of amiloride by airway epithelia translated into a short duration of efficacy as an aerosolized therapy for cystic fibrosis (CF) patients. To improve ENaC blocker CF pharmacotherapy, a more potent and durable ENaC blocker tailored for aerosol delivery was synthesized. Parion compound N-(3,5-diamino-6-chloropyrazine-2-carbonyl)-N'-4-[4-(2,3-dihydroxypropoxy)phenyl]butyl-guanidine methanesulfonate (552-02) was tested for potency and reversibility of ENaC block, epithelial absorption and biotransformation, selectivity, durability of ASL expansion under isotonic and hypertonic conditions in canine and human CF bronchial epithelial cells, and drug dissociation on ENaC in Xenopus oocytes. Short-circuit current assessed compound potency and reversibility, patch-clamp recordings of ENaC current assessed drug off-rate (k(off)), a gravimetric method and confocal microscopy measured mucosal water retention and ASL height, and drug absorption and biotransformation were assessed using liquid chromatography-mass spectrometry. Amiloride and 552-02 were tested in vivo for MC activity in sheep immediately and 4 to 6 h after aerosol dosing. Compared with amiloride, compound 552-02 was 60 to 100-fold more potent, it was 2 to 5-fold less reversible, it was slower at crossing the epithelium, and it exhibited a 170-fold slower k(off) value. 552-02 exhibited greater ASL expansion over 8 h in vitro, and it was more effective than amiloride at increasing MC immediately and 4 to 6 h after dosing. When combining hypertonic saline and 552-02, a synergistic effect on ASL expansion was measured in canine or CF bronchial epithelia. In summary, the preclinical data support the clinical use of 552-02 +/- hypertonic saline for CF lung disease.


Subject(s)
Cystic Fibrosis/drug therapy , Epithelial Sodium Channel Blockers , Mesylates/pharmacokinetics , Sodium Channel Blockers/pharmacokinetics , Absorption , Animals , Biotransformation , Dogs , Humans , Lung Diseases , Mesylates/pharmacology , Mesylates/therapeutic use , Respiratory Mucosa/metabolism , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology
9.
J Appl Physiol (1985) ; 102(6): 2324-31, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17395761

ABSTRACT

Neutrophil elastase is a mediator common to asthma, chronic obstructive pulmonary disease, and cystic fibrosis and thought to contribute to the pathophysiology of these diseases. Previously, we found that inhaled hyaluronan blocked elastase-induced bronchoconstriction in allergic sheep through its control of tissue kallikrein. Here, we extend those studies by determining if inhaled hyaluronan can protect against the elastase-induced depression in tracheal mucus velocity, a surrogate marker of whole lung mucociliary clearance. We measured tracheal mucus velocity in allergic sheep before, and sequentially for 6 h after, aerosol challenge with porcine pancreatic elastase alone and after pretreatment with 1.5 or 6 mg aerosolized hyaluronan. Elastase (2.55 U) decreased tracheal mucus velocity. Pretreatment with 6 mg, but not 1.5 mg, hyaluronan inhibited the elastase-induced decrease in tracheal mucus velocity. Hyaluronan (6 mg) given 1 h after elastase challenge was ineffective, suggesting the involvement of secondary mediators. The elastase-induced depression in mucus transport appeared to be mediated, in part, by reactive oxygen species and bradykinin because pretreatment with either aerosolized catalase (38 mg/3 ml) or the bradykinin B2-receptor antagonist HOE140 (400 nM/kg) was also effective in blocking the response. These latter two findings are consistent with oxygen radical-induced degradation of hyaluronan with concomitant loss of its regulatory effect on tissue kallikrein, resulting in kinin generation. This hypothesis is supported by the demonstration that hyaluronan failed to block the oxygen radical-induced fall in tracheal mucus velocity resulting from xanthine-xanthine oxidase challenge and that inhaled bradykinin itself can slow mucociliary transport.


Subject(s)
Asthma/chemically induced , Asthma/prevention & control , Hyaluronic Acid/administration & dosage , Mucociliary Clearance/drug effects , Pancreatic Elastase , Respiratory Mucosa/drug effects , Respiratory Mucosa/physiopathology , Administration, Inhalation , Animals , Asthma/pathology , Asthma/physiopathology , Dose-Response Relationship, Drug , Female , Hypersensitivity/etiology , Hypersensitivity/pathology , Hypersensitivity/physiopathology , Hypersensitivity/prevention & control , Pancreas/enzymology , Sheep , Swine
10.
Chest ; 128(5): 3743-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16304342

ABSTRACT

STUDY OBJECTIVE: We measured tracheal mucus velocity (TMV), a marker of mucociliary clearance (MCC), in sheep before and for 12 h after treatment with salmeterol, albuterol, ipratropium, or vehicle to determine the effects on normal MCC. We also determined if these agents could reverse the depression in TMV caused by inhaled human neutrophil elastase (HNE), a model of abnormal MCC. METHODS: Study 1: TMV was measured initially and then for 6 h after metered-dose inhaler treatment with salmeterol (42 microg), albuterol (180 microg), ipratropium bromide (36 microg), or vehicle. After 6 h, the sheep in the albuterol and ipratropium treatment arms were administered a second dose of drug, whereas the salmeterol and vehicle treatment arms received vehicle. TMV was measured for another 6 h. Study 2: Six sheep inhaled HNE aerosol, which significantly reduced TMV by 2 h. At this point, the sheep were treated with either salmeterol, albuterol, ipratropium, or vehicle, and the effects on TMV were measured for another 6 h. This experiment was repeated in four sheep using only salmeterol and albuterol, but the posttreatment measurements were extended to 12 h. RESULTS: Study 1: Only salmeterol and albuterol increased TMV (p < 0.05) during the initial 6-h period. From 6 to 12 h only, the salmeterol-treated sheep had TMV that remained at or above the initial TMV for the entire time, although both albuterol and ipratropium showed enhancement of TMV compared to vehicle. Study 2: Salmeterol and albuterol reversed the HNE-induced depression in TMV to a similar degree over the 6-h time course. However, the protection afforded by salmeterol was more prolonged than that seen with albuterol if the posttreatment interval was extended to 12 h. Ipratropium and vehicle had no effect. CONCLUSION: We conclude that salmeterol and albuterol can stimulate normal MCC and reverse HNE-induced mucociliary dysfunction and that salmeterol has a longer duration of action in these models of normal and abnormal MCC.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Albuterol/analogs & derivatives , Albuterol/pharmacology , Cholinergic Antagonists/pharmacology , Ipratropium/pharmacology , Mucociliary Clearance/drug effects , Animals , Area Under Curve , Salmeterol Xinafoate , Sheep , Trachea/physiology
11.
Am J Respir Crit Care Med ; 171(1): 26-34, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15447946

ABSTRACT

Florida red tide brevetoxins are sodium channel neurotoxins produced by the dinoflagellate Karenia brevis. When aerosolized, the toxin causes airway symptoms in normal individuals and patients with airway disease, but systematic exposures to define the pulmonary consequences and putative mechanisms are lacking. Here we report the effects of airway challenges with lysed cultures of Karenia brevis (crude brevetoxin), pure brevetoxin-2, brevetoxin-3, and brevetoxin-tbm (brevetoxin-2 minus the side chain) on pulmonary resistance and tracheal mucus velocity, a marker of mucociliary clearance, in allergic and nonallergic sheep. Picogram concentrations of toxin caused bronchoconstriction in both groups of sheep. Brevetoxin-tbm was the least potent, indicating the importance of the side chain for maximum effect. Both histamine H(1)- and cholinergic-mediated pathways contributed to the bronchoconstriction. A synthetic antagonist, beta-naphthoyl-brevetoxin-3, and brevenal, a natural antagonist, inhibited the bronchoconstriction. Only crude brevetoxin and brevetoxin-3 decreased tracheal mucus velocity; both antagonists prevented this. More importantly, picomolar concentrations of the antagonists alone improved tracheal mucus velocity to the degree seen with mM concentrations of the sodium channel blocker amiloride. Thus, Karenia brevis, in addition to producing toxins that adversely affect the airways, may be a source of agents for treating mucociliary dysfunction.


Subject(s)
Asthma/physiopathology , Dinoflagellida , Marine Toxins/toxicity , Neurotoxins/toxicity , Oxocins/toxicity , Administration, Inhalation , Aerosols , Airway Resistance/drug effects , Animals , Bronchial Provocation Tests , Bronchoconstriction/drug effects , Dose-Response Relationship, Drug , Female , Marine Toxins/antagonists & inhibitors , Marine Toxins/chemistry , Mucociliary Clearance/drug effects , Neurotoxins/antagonists & inhibitors , Neurotoxins/chemistry , Oxocins/antagonists & inhibitors , Oxocins/chemistry , Sheep
12.
J Pharmacol Exp Ther ; 311(3): 929-38, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15273255

ABSTRACT

Epithelial sodium channel (ENaC) blockers have been proposed as a therapy to restore mucus clearance (MC) in cystic fibrosis (CF) airways. The therapeutic effects of the first generation ENaC blocker, amiloride, in CF patients, however, were minimal. Because the failure of amiloride reflected both its low potency and short duration of action on airway surfaces, we investigated whether the increased potency of benzamil and phenamil would produce more favorable pharmacodynamic properties. In vitro potency, maximal efficacy, rate of recovery from maximal block of ENaC, and rate of drug absorption were compared for amiloride, benzamil, and phenamil in cultured human and ovine bronchial epithelial cells. In both human and ovine bronchial epithelia, the rank order of potency was benzamil > phenamil >> amiloride, the maximal efficacy was benzamil = phenamil = amiloride, the recovery to baseline sodium transport was phenamil < benzamil << amiloride, and the rate of drug absorption was phenamil > benzamil >> amiloride. Based on greater potency, benzamil was compared with amiloride in in vivo pharmacodynamic studies in sheep, including tracheal mucus velocity (TMV) and MC. Benzamil enhanced MC and TMV, but acute potency or duration of effect did not exceed that of amiloride. In conclusion, our data support the hypothesis that ENaC blocker aerosol therapy increases MC. However, rapid absorption of benzamil from the mucosal surface offset its greater potency, making it equieffective with amiloride in vivo. More potent, less absorbable, third generation ENaC blockers will be required for an effective aerosol CF pharmacotherapy.


Subject(s)
Amiloride/analogs & derivatives , Amiloride/therapeutic use , Cystic Fibrosis/drug therapy , Lung Diseases/drug therapy , Sodium Channel Blockers/therapeutic use , Absorption , Amiloride/pharmacokinetics , Animals , Bronchi/drug effects , Bronchi/metabolism , Cystic Fibrosis/complications , Electrophysiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Sodium Channels , Epithelium/drug effects , Epithelium/metabolism , Female , Humans , In Vitro Techniques , Lung Diseases/etiology , Mannitol/metabolism , Microscopy, Confocal , Mucus/physiology , Muscle Contraction/drug effects , Sheep , Sodium/metabolism , Sodium Channels/metabolism , Viscosity
13.
Am J Respir Crit Care Med ; 166(11): 1457-60, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12406819

ABSTRACT

The cysteinyl leukotrienes are potent proinflammatory mediators that, in addition to their bronchospastic actions, can also contribute to mucociliary dysfunction, a central component of the pathophysiology of asthma. In this study, we determined whether montelukast, a cysteinyl leukotriene 1 receptor antagonist, could prevent and/or reverse antigen-induced mucociliary dysfunction in allergic sheep. We measured tracheal mucus velocity, a marker of mucociliary clearance, before and for 8 hours after antigen challenge in six animals treated with montelukast (0.15 mg/kg, intravenously) 30 minutes before, 1 hour after, or 4 hours after antigen challenge. In the control trial, the sheep received 0.9% saline intravenously at each of the previously mentioned time points. The maximum decrease in tracheal mucus velocity seen in the control trial was 56 +/- 4% (mean +/- SE) of baseline at 8 hours. Pretreatment with montelukast significantly protected against this reduction. However, treatment at 1 and 4 hours neither protected against nor reversed the allergen-induced fall in tracheal mucus velocity. We conclude that the early release of cysteinyl leukotrienes may contribute to the fall in tracheal mucus velocity that follows acute antigen challenge and that pretreatment with montelukast reduces this impairment.


Subject(s)
Acetates/pharmacology , Antigens, Helminth/immunology , Antigens, Helminth/pharmacology , Leukotriene Antagonists/pharmacology , Mucociliary Clearance/drug effects , Quinolines/pharmacology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/physiopathology , Animals , Ascaris suum/immunology , Cyclopropanes , Mucus/metabolism , Sheep , Sulfides , Time Factors , Trachea/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...