Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38573173

ABSTRACT

Rationale: Pulmonary ionocytes are a newly discovered airway epithelial cell type proposed to be a major contributor to cystic fibrosis (CF) lung disease based on observations they express the cystic fibrosis transmembrane conductance regulator (CFTR) anion channel at a higher level than any other cell type in the airway epithelia. Moreover, genetically manipulated experimental models that lack ionocytes develop NaCl transport abnormalities and airway surface liquid (ASL) dehydration consistent with CF. However, no direct evidence indicates ionocytes engage in NaCl transport or contribute to ASL formation, questioning the relevance of ionocytes to CF lung disease. Objectives: To determine the ion transport properties of pulmonary ionocytes and club cells in genetically intact healthy and CF airway epithelia. Methods: We measured ion transport at the single-cell level using a self-referencing ion-selective microelectrode technique in primary human bronchial epithelial cell culture. Measurements and Main Results: cAMP-stimulated non-CF ionocytes do not secrete Na+ or Cl- into the ASL, but rather modulate its pH by secreting bicarbonate via CFTR-linked Cl-/bicarbonate exchange. Non-CF club cells secrete Na+ and Cl- to the lumen side after cAMP stimulation. CF ionocytes and club cells do not transport ions in response to cAMP stimulation, but incubation with CFTR modulators elexacaftor/tezacaftor/ivacaftor restores transport properties. Conclusions: We conclude that ionocytes do not contribute to ASL formation but regulate ASL pH. Club cells secrete the bulk of airway fluid. In CF, abnormal ionocyte and club cell function results in acidic and dehydrated ASL, causing reduced antimicrobial properties and mucociliary clearance. This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License 4.0 (http://creativecommons.org/licenses/by-nc-nd/4.0/).

2.
Cell Rep ; 37(1): 109795, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34610318

ABSTRACT

A controversial hypothesis pertaining to cystic fibrosis (CF) lung disease is that the CF transmembrane conductance regulator (CFTR) channel fails to inhibit the epithelial Na+ channel (ENaC), yielding increased Na+ reabsorption and airway dehydration. We use a non-invasive self-referencing Na+-selective microelectrode technique to measure Na+ transport across individual folds of distal airway surface epithelium preparations from CFTR-/- (CF) and wild-type (WT) swine. We show that, under unstimulated control conditions, WT and CF epithelia exhibit similar, low rates of Na+ transport that are unaffected by the ENaC blocker amiloride. However, in the presence of the cyclic AMP (cAMP)-elevating agents forskolin+IBMX (isobutylmethylxanthine), folds of WT tissues secrete large amounts of Na+, while CFTR-/- tissues absorb small, but potentially important, amounts of Na+. In cAMP-stimulated conditions, amiloride inhibits Na+ absorption in CFTR-/- tissues but does not affect secretion in WT tissues. Our results are consistent with the hypothesis that ENaC-mediated Na+ absorption may contribute to dehydration of CF distal airways.


Subject(s)
Cyclic AMP/metabolism , Epithelial Sodium Channels/metabolism , Epithelium/metabolism , Sodium/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Amiloride/pharmacology , Animals , Animals, Genetically Modified/metabolism , Colforsin/pharmacology , Cystic Fibrosis , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Models, Animal , Epithelial Sodium Channel Blockers/pharmacology , Epithelial Sodium Channels/chemistry , Ion Transport/drug effects , Male , Swine
3.
Sci Rep ; 11(1): 8336, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33863932

ABSTRACT

Thoracic dorsal root ganglia (tDRG) contribute to fluid secretion in the upper airways. Inflammation potentiates DRG responses, but the mechanisms remain under investigation. The receptor for advanced glycation end-products (RAGE) underlies potentiation of DRG responses in pain pathologies; however, its role in other sensory modalities is less understood. We hypothesize that RAGE contributes to electrophysiological and biochemical changes in tDRGs during inflammation. We used tDRGs and tracheas from wild types (WT), RAGE knock-out (RAGE-KO), and with the RAGE antagonist FPS-ZM1, and exposed them to lipopolysaccharides (LPS). We studied: capsaicin (CAP)-evoked currents and action potentials (AP), tracheal submucosal gland secretion, RAGE expression and downstream pathways. In WT neurons, LPS increased CAP-evoked currents and AP generation, and it caused submucosal gland hypersecretion in tracheas from WT mice exposed to LPS. In contrast, LPS had no effect on tDRG excitability or gland secretion in RAGE-KO mice or mice treated with FPS-ZM1. LPS upregulated full-length RAGE (encoded by Tv1-RAGE) and downregulated a soluble (sRAGE) splice variant (encoded by MmusRAGEv4) in tDRG neurons. These data suggest that sensitization of tDRG neurons contributes to hypersecretion in the upper airways during inflammation. And at least two RAGE variants may be involved in these effects of LPS.


Subject(s)
Ganglia, Spinal/physiopathology , Lipopolysaccharides/adverse effects , Receptor for Advanced Glycation End Products/physiology , Respiratory Mucosa/metabolism , Trachea/metabolism , Action Potentials/drug effects , Animals , Benzamides/pharmacology , Down-Regulation/drug effects , Gene Expression , Mice, Inbred C57BL , Mice, Knockout , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Up-Regulation/drug effects
4.
J Exp Med ; 218(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33533914

ABSTRACT

The airway mucosal microenvironment is crucial for host defense against inhaled pathogens but remains poorly understood. We report here that the airway surface normally undergoes surprisingly large excursions in pH during breathing that can reach pH 9.0 during inhalation, making it the most alkaline fluid in the body. Transient alkalinization requires luminal bicarbonate and membrane-bound carbonic anhydrase 12 (CA12) and is antimicrobial. Luminal bicarbonate concentration and CA12 expression are both reduced in cystic fibrosis (CF), and mucus accumulation both buffers the pH and obstructs airflow, further suppressing the oscillations and bacterial-killing efficacy. Defective pH oscillations may compromise airway host defense in other respiratory diseases and explain CF-like airway infections in people with CA12 mutations.


Subject(s)
Cystic Fibrosis/immunology , Host Microbial Interactions/immunology , Nasal Mucosa/chemistry , Nasal Mucosa/immunology , Respiratory Tract Infections/immunology , Adult , Bicarbonates/metabolism , Bronchi/cytology , Bronchi/immunology , Bronchi/metabolism , Carbonic Anhydrases/metabolism , Case-Control Studies , Cells, Cultured , Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Female , Humans , Hydrogen-Ion Concentration , Male , Nasal Mucosa/metabolism , Respiratory Tract Infections/metabolism , Young Adult
5.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L931-L942, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32130033

ABSTRACT

The human airway is protected by an efficient innate defense mechanism that requires healthy secretion of airway surface liquid (ASL) to clear pathogens from the lungs. Most of the ASL in the upper airway is secreted by submucosal glands. In cystic fibrosis (CF), the function of airway submucosal glands is abnormal, and these abnormalities are attributed to anomalies in ion transport across the epithelia lining the different sections of the glands that function coordinately to produce the ASL. However, the ion transport properties of most of the anatomical regions of the gland have never been measured, and there is controversy regarding which segments express CFTR. This makes it difficult to determine the glandular abnormalities that may contribute to CF lung disease. Using a noninvasive, extracellular self-referencing ion-selective electrode technique, we characterized ion transport properties in all four segments of submucosal glands from wild-type and CFTR-/- swine. In wild-type airways, the serous acini, mucus tubules, and collecting ducts secrete Cl- and Na+ into the lumen in response to carbachol and forskolin stimulation. The ciliated duct also transports Cl- and Na+ but in the opposite direction, i.e., reabsorption from the ASL, which may contribute to lowering Na+ and Cl- activities in the secreted fluid. In CFTR-/- airways, the serous acini, collecting ducts, and ciliated ducts fail to transport ions after forskolin stimulation, resulting in the production of smaller volumes of ASL with normal Cl-, Na+, and K+ concentration.


Subject(s)
Acinar Cells/metabolism , Cilia/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/metabolism , Lung/metabolism , Acinar Cells/drug effects , Acinar Cells/pathology , Animals , Carbachol/pharmacology , Cations, Monovalent , Chlorides/metabolism , Cilia/drug effects , Cilia/pathology , Colforsin/pharmacology , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Disease Models, Animal , Electrochemical Techniques , Electrodes , Gene Deletion , Gene Expression , Humans , Ion Transport , Lung/drug effects , Lung/pathology , Potassium/metabolism , Sodium/metabolism , Swine
6.
Sci Rep ; 9(1): 540, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679487

ABSTRACT

Inhaled hypertonic saline (HTS) treatment is used to improve lung health in patients with cystic fibrosis (CF). The current consensus is that the treatment generates an osmotic gradient that draws water into the airways and increases airway surface liquid (ASL) volume. However, there is evidence that HTS may also stimulate active secretion of ASL by airway epithelia through the activation of sensory neurons. We tested the contribution of the nervous system and airway epithelia on HTS-stimulated ASL height increase in CF and wild-type swine airway. We used synchrotron-based imaging to investigate whether airway neurons and epithelia are involved in HTS treatment-triggered ASL secretion in CFTR-/- and wild-type swine. We showed that blocking parasympathetic and sensory neurons in airway resulted in ~50% reduction of the effect of HTS treatment on ASL volume in vivo. Incubating tracheal preparations with inhibitors of epithelial ion transport across airway decreased secretory responses to HTS treatment. CFTR-/- swine ex-vivo tracheal preparations showed substantially decreased secretory response to HTS treatment after blockage of neuronal activity. Our results indicated that HTS-triggered ASL secretion is partially mediated by the stimulation of airway neurons and the subsequent activation of active epithelia secretion; osmosis accounts for only ~50% of the effect.


Subject(s)
Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Mediastinal Cyst/drug therapy , Mediastinal Cyst/metabolism , Saline Solution, Hypertonic/therapeutic use , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Administration, Inhalation , Animals , Animals, Genetically Modified , Bodily Secretions/drug effects , Bodily Secretions/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Female , Gene Knockout Techniques , Ion Transport/drug effects , Male , Osmosis/drug effects , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Saline Solution, Hypertonic/administration & dosage , Saline Solution, Hypertonic/pharmacology , Swine
7.
Elife ; 62017 11 21.
Article in English | MEDLINE | ID: mdl-29157359

ABSTRACT

Blood-sucking insects experience thermal stress at each feeding event on endothermic vertebrates. We used thermography to examine how kissing-bugs Rhodnius prolixus actively protect themselves from overheating. During feeding, these bugs sequester and dissipate the excess heat in their heads while maintaining an abdominal temperature close to ambient. We employed a functional-morphological approach, combining histology, µCT and X-ray-synchrotron imaging to shed light on the way these insects manage the flow of heat across their bodies. The close alignment of the circulatory and ingestion systems, as well as other morphological characteristics, support the existence of a countercurrent heat exchanger in the head of R. prolixus, which decreases the temperature of the ingested blood before it reaches the abdomen. This kind of system has never been described before in the head of an insect. For the first time, we show that countercurrent heat exchange is associated to thermoregulation during blood-feeding.


Subject(s)
Rhodnius/physiology , Animals , Body Temperature Regulation , Feeding Behavior , Head/physiology , Histocytochemistry , Hot Temperature , Stress, Physiological , X-Ray Microtomography
8.
Nat Commun ; 8(1): 786, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28983075

ABSTRACT

Cystic fibrosis is caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) channel, which can result in chronic lung disease. The sequence of events leading to lung disease is not fully understood but recent data show that the critical pathogenic event is the loss of the ability to clear bacteria due to abnormal airway surface liquid secretion (ASL). However, whether the inhalation of bacteria triggers ASL secretion and whether this is abnormal in cystic fibrosis has never been tested. Here we show, using a novel synchrotron-based in vivo imaging technique, that wild-type pigs display both a basal and a Toll-like receptor-mediated ASL secretory response to the inhalation of cystic fibrosis relevant bacteria. Both mechanisms fail in CFTR-/- swine, suggesting that cystic fibrosis airways do not respond to inhaled pathogens, thus favoring infection and inflammation that may eventually lead to tissue remodeling and respiratory disease.Cystic fibrosis is caused by mutations in the CFTR chloride channel, leading to reduced airway surface liquid secretion. Here the authors show that exposure to bacteria triggers secretion in wild-type but not in pig models of cystic fibrosis, suggesting an impaired response to pathogens contributes to infection.


Subject(s)
Cystic Fibrosis/metabolism , Lung/metabolism , Pseudomonas aeruginosa , Respiratory Mucosa/metabolism , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Models, Animal , Inhalation Exposure , Lung/diagnostic imaging , Male , Radiography , Swine
9.
Proc Natl Acad Sci U S A ; 111(35): 12930-5, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136096

ABSTRACT

Cystic fibrosis (CF) is an autosomal recessive genetic disorder caused by mutations in the gene encoding for the anion channel cystic fibrosis transmembrane conductance regulator (CFTR). Several organs are affected in CF, but most of the morbidity and mortality comes from lung disease. Recent data show that the initial consequence of CFTR mutation is the failure to eradicate bacteria before the development of inflammation and airway remodeling. Bacterial clearance depends on a layer of airway surface liquid (ASL) consisting of both a mucus layer that traps, kills, and inactivates bacteria and a periciliary liquid layer that keeps the mucus at an optimum distance from the underlying epithelia, to maximize ciliary motility and clearance of bacteria. The airways in CF patients and animal models of CF demonstrate abnormal ASL secretion and reduced antimicrobial properties. Thus, it has been proposed that abnormal ASL secretion in response to bacteria may facilitate the development of the infection and inflammation that characterize CF airway disease. Whether the inhalation of bacteria triggers ASL secretion, and the role of CFTR, have never been tested, however. We developed a synchrotron-based imaging technique to visualize the ASL layer and measure the effect of bacteria on ASL secretion. We show that the introduction of Pseudomonas aeruginosa and other bacteria into the lumen of intact isolated swine tracheas triggers CFTR-dependent ASL secretion by the submucosal glands. This response requires expression of the bacterial protein flagellin. In patients with CF, the inhalation of bacteria would fail to trigger ASL secretion, leading to infection and inflammation.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/microbiology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/metabolism , Trachea/metabolism , Trachea/microbiology , Animals , Cystic Fibrosis/immunology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/immunology , Female , Haemophilus influenzae/metabolism , Immunity, Innate/physiology , Male , Mucins/metabolism , Pseudomonas Infections/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Staphylococcus aureus/metabolism , Swine , Synchrotrons , Trachea/immunology
10.
Am J Physiol Lung Cell Mol Physiol ; 303(4): L327-33, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22683572

ABSTRACT

The airway is kept sterile by an efficient innate defense mechanism. The cornerstone of airway defense is mucus containing diverse antimicrobial factors that kill or inactivate pathogens. Most of the mucus in the upper airways is secreted by airway submucosal glands. In patients with cystic fibrosis (CF), airway defense fails and the lungs are colonized by bacteria, usually Pseudomonas aeruginosa. Accumulating evidence suggests that airway submucosal glands contribute to CF pathogenesis by failing to respond appropriately to inhalation of bacteria. However, the regulation of submucosal glands by the innate immune system remains poorly understood. We studied the response of submucosal glands to the proinflammatory cytokines interleukin-1ß and tumor necrosis factor-α. These are released into the airway submucosa in response to infection with the bacterium P. aeruginosa and are elevated in CF airways. Stimulation with IL-1ß and TNF-α increased submucosal gland secretion in a concentration-dependent manner with a maximal secretion rate of 240 ± 20 and 190 ± 40 pl/min, respectively. The half maximal effective concentrations were 11 and 20 ng/ml, respectively. The cytokine effect was dependent on cAMP but was independent of cGMP, nitric oxide, Ca(2+), or p38 MAP kinase. Most importantly, IL-1ß- and TNF-α-stimulated secretion was blocked by the CF transmembrane conductance regulator (CFTR) blocker, CFTRinh172 (100 µmol/l) but was not affected by the Ca(2+)-activated Cl(-) channel blocker, niflumic acid (1 µmol/l). The data suggest, that during bacterial infections and resulting release of proinflammatory cytokines, the glands are stimulated to secrete fluid, and this response is mediated by cAMP-activated CFTR, a process that would fail in patients with CF.


Subject(s)
Body Fluids/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Interleukin-1beta/pharmacology , Mucus/metabolism , Respiratory System/anatomy & histology , Respiratory System/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Body Fluids/drug effects , Calcium/metabolism , Cyclic AMP/pharmacology , Cyclic GMP/pharmacology , Female , In Vitro Techniques , Male , Mucus/drug effects , Niflumic Acid/pharmacology , Nitric Oxide/metabolism , Respiratory System/drug effects , Respiratory System/enzymology , Second Messenger Systems/drug effects , Signal Transduction/drug effects , Swine , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...