Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Cyst Fibros ; 22 Suppl 1: S23-S26, 2023 03.
Article in English | MEDLINE | ID: mdl-36117114

ABSTRACT

Cystic fibrosis (CF), an autosomal genetic disorder caused by the dysfunction of the cystic fibrosis transmembrane conductance regulator (CFTR) protein, is characterized by mucus accumulation in the lungs, the intestinal tract, and the pancreatic ducts. Mucins are high-molecular-weight glycoproteins that govern the biochemical and biophysical properties of mucus. In the CF lung, increased mucus viscoelasticity is associated with decreased mucociliary clearance and defects in host defense mechanisms. The link between defective ion channel and abnormal mucus properties has been investigated in studies involving cell and animal models. In this review article, we discuss recent progress toward understanding the different regions and cells that express CFTR in the airways and how mucus is produced and cleared from the lungs. In addition, we reflect on animal models that provided insights into the organization and the role of the mucin network and how mucus and antimicrobial activities act in concert to protect the lungs from invading pathogens.


Subject(s)
Cystic Fibrosis , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Mucus/metabolism , Mucins/metabolism , Lung , Models, Animal
2.
Elife ; 92020 10 07.
Article in English | MEDLINE | ID: mdl-33026343

ABSTRACT

Submucosal glands (SMGs) are a prominent structure that lines human cartilaginous airways. Although it has been assumed that SMGs contribute to respiratory defense, that hypothesis has gone without a direct test. Therefore, we studied pigs, which have lungs like humans, and disrupted the gene for ectodysplasin (EDA-KO), which initiates SMG development. EDA-KO pigs lacked SMGs throughout the airways. Their airway surface liquid had a reduced ability to kill bacteria, consistent with SMG production of antimicrobials. In wild-type pigs, SMGs secrete mucus that emerges onto the airway surface as strands. Lack of SMGs and mucus strands disrupted mucociliary transport in EDA-KO pigs. Consequently, EDA-KO pigs failed to eradicate a bacterial challenge in lung regions normally populated by SMGs. These in vivo and ex vivo results indicate that SMGs are required for normal antimicrobial activity and mucociliary transport, two key host defenses that protect the lung.


Subject(s)
Ectodysplasins/genetics , Exocrine Glands/immunology , Respiratory Mucosa/immunology , Staphylococcus aureus/physiology , Sus scrofa/immunology , Animals , Ectodysplasins/immunology , Female , Gene Knockout Techniques , Male , Sus scrofa/genetics
3.
Lab Invest ; 100(11): 1388-1399, 2020 11.
Article in English | MEDLINE | ID: mdl-32719544

ABSTRACT

Hepatobiliary disease causes significant morbidity in people with cystic fibrosis (CF), yet this problem remains understudied. We previously found that newborn CF pigs have microgallbladders with significant luminal obstruction in the absence of infection and consistent inflammation. In this study, we sought to better understand the early pathogenesis of CF pig gallbladder disease. We hypothesized that loss of CFTR would impair gallbladder epithelium anion/liquid secretion and increase mucin production. CFTR was expressed apically in non-CF pig gallbladder epithelium but was absent in CF. CF pig gallbladders lacked cAMP-stimulated anion transport. Using a novel gallbladder epithelial organoid model, we found that Cl- or HCO3- was sufficient for non-CF organoid swelling. This response was absent for non-CF organoids in Cl-/HCO3--free conditions and in CF. Single-cell RNA-sequencing revealed a single epithelial cell type in non-CF gallbladders that coexpressed CFTR, MUC5AC, and MUC5B. Despite CF gallbladders having increased luminal MUC5AC and MUC5B accumulation, there was no significant difference in the epithelial expression of gel-forming mucins between non-CF and CF pig gallbladders. In conclusion, these data suggest that loss of CFTR-mediated anion transport and fluid secretion contribute to microgallbladder development and luminal mucus accumulation in CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cystic Fibrosis/complications , Gallbladder Diseases/etiology , Gallbladder/metabolism , Animals , Animals, Newborn , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Disease Models, Animal , Epithelial Cells/metabolism , Gallbladder/physiopathology , Gallbladder Diseases/metabolism , Mucin 5AC/metabolism , Mucin-5B/metabolism , Swine , Transcriptome
4.
JCI Insight ; 4(1)2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30626743

ABSTRACT

Mucus produced by submucosal glands is a key component of respiratory mucociliary transport (MCT). When it emerges from submucosal gland ducts, mucus forms long strands on the airway surface. However, the function of those strands is uncertain. To test the hypothesis that mucus strands facilitate transport of large particles, we studied newborn pigs. In ex vivo experiments, interconnected mucus strands moved over the airway surface, attached to immobile spheres, and initiated their movement by pulling them. Stimulating submucosal gland secretion with methacholine increased the percentage of spheres that moved and shortened the delay until mucus strands began moving spheres. To disrupt mucus strands, we applied reducing agents tris-(2-carboxyethyl)phosphine and dithiothreitol. They decreased the fraction of moving spheres and delayed initiation of movement for spheres that did move. We obtained similar in vivo results with CT-based tracking of microdisks in spontaneously breathing pigs. Methacholine increased the percentage of microdisks moving and reduced the delay until they were propelled up airways. Aerosolized tris-(2-carboxyethyl)phosphine prevented those effects. Once particles started moving, reducing agents did not alter their speed either ex vivo or in vivo. These findings indicate that submucosal glands produce mucus in the form of strands and that the strands initiate movement of large particles, facilitating their removal from airways.

5.
Ann Am Thorac Soc ; 15(Suppl 3): S171-S176, 2018 11.
Article in English | MEDLINE | ID: mdl-30431346

ABSTRACT

Cystic fibrosis (CF) lung disease is the major cause of morbidity and mortality in people with CF. Abnormal mucociliary transport has been the leading hypothesis for the underlying pathogenesis of CF airway disease. However, this has been difficult to investigate at very early time points. A porcine CF model, which recapitulates many features of CF disease in humans, enables studies to be performed in non-CF and CF pigs on the day that they are born. In newborn CF pigs, we found that under basal conditions, mucociliary transport rates in non-CF and CF pigs are similar. However, after cholinergic stimulation, which stimulates submucosal gland secretion, particles become stuck in the CF airways owing to a failure of mucus strands to release from submucosal glands. In this review, we summarize these recent discoveries and also discuss the morphology, composition, and function of mucins in the porcine lung.


Subject(s)
Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Mucociliary Clearance/physiology , Respiratory Mucosa/physiology , Animals , Animals, Newborn , Cystic Fibrosis/etiology , Disease Models, Animal , Mucus/metabolism , Swine
6.
Nucleic Acids Res ; 46(18): 9591-9600, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30165523

ABSTRACT

Cystic fibrosis (CF) is a common genetic disease caused by mutations in the gene coding for cystic fibrosis transmembrane conductance regulator (CFTR). Although CF affects multiple organ systems, chronic bacterial infections and inflammation in the lung are the leading causes of morbidity and mortality in people with CF. Complementation with a functional CFTR gene repairs this defect, regardless of the disease-causing mutation. In this study, we used a gene delivery system termed piggyBac/adenovirus (Ad), which combines the delivery efficiency of an adenoviral-based vector with the persistent expression of a DNA transposon-based vector. We aerosolized piggyBac/Ad to the airways of pigs and observed widespread pulmonary distribution of vector. We quantified the regional distribution in the airways and observed transduction of large and small airway epithelial cells of non-CF pigs, with ∼30-50% of surface epithelial cells positive for GFP. We transduced multiple cell types including ciliated, non-ciliated, basal, and submucosal gland cells. In addition, we phenotypically corrected CF pigs following delivery of piggyBac/Ad expressing CFTR as measured by anion channel activity, airway surface liquid pH, and bacterial killing ability. Combining an integrating DNA transposon with adenoviral vector delivery is an efficient method for achieving functional CFTR correction from a single vector administration.


Subject(s)
Adenoviridae/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/therapy , DNA Transposable Elements/genetics , Genetic Therapy/methods , Lung/metabolism , Aerosols/administration & dosage , Aerosols/pharmacokinetics , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/administration & dosage , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/pharmacokinetics , Phenotype , Respiratory Mucosa/metabolism , Swine , Tissue Distribution , Treatment Outcome
7.
Proc Natl Acad Sci U S A ; 115(6): 1370-1375, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29358407

ABSTRACT

Differentiated airway epithelia produce sonic hedgehog (SHH), which is found in the thin layer of liquid covering the airway surface. Although previous studies showed that vertebrate HH signaling requires primary cilia, as airway epithelia mature, the cells lose primary cilia and produce hundreds of motile cilia. Thus, whether airway epithelia have apical receptors for SHH has remained unknown. We discovered that motile cilia on airway epithelial cells have HH signaling proteins, including patched and smoothened. These cilia also have proteins affecting cAMP-dependent signaling, including Gαi and adenylyl cyclase 5/6. Apical SHH decreases intracellular levels of cAMP, which reduces ciliary beat frequency and pH in airway surface liquid. These results suggest that apical SHH may mediate noncanonical HH signaling through motile cilia to dampen respiratory defenses at the contact point between the environment and the lung, perhaps counterbalancing processes that stimulate airway defenses.


Subject(s)
Bronchi/cytology , Epithelial Cells/metabolism , Hedgehog Proteins/metabolism , Trachea/cytology , Cells, Cultured , Cilia/metabolism , Cilia/physiology , Cyclic AMP/metabolism , Epithelial Cells/cytology , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
8.
Proc Natl Acad Sci U S A ; 114(26): 6842-6847, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28607090

ABSTRACT

Gel-forming mucins, the primary macromolecular components of airway mucus, facilitate airway clearance by mucociliary transport. In cystic fibrosis (CF) altered mucus properties impair mucociliary transport. Airways primarily secrete two closely related gel-forming mucins, MUC5B and MUC5AC. However, their morphologic structures and associations in airways that contain abundant submucosal glands and goblet cells are uncertain. Moreover, there is limited knowledge about mucins in airways not affected by inflammation, infection, or remodeling or in CF airways. Therefore, we examined airways freshly excised from newborn non-CF pigs and CF pigs before secondary manifestations develop. We found that porcine submucosal glands produce MUC5B, whereas goblet cells produce predominantly MUC5AC plus some MUC5B. We found that MUC5B emerged from submucosal gland ducts in the form of strands composed of multiple MUC5B filaments. In contrast, MUC5AC emerged from goblet cells as wispy threads and sometimes formed mucin sheets. In addition, MUC5AC often partially coated the MUC5B strands. Compared with non-CF, MUC5B more often filled CF submucosal gland ducts. MUC5AC sheets also accumulated in CF airways overlying MUC5B strands. These results reveal distinct morphology and interactions for MUC5B and MUC5AC and suggest that the two mucins make distinct contributions to mucociliary transport. Thus, they provide a framework for understanding abnormalities in disease.


Subject(s)
Airway Remodeling , Cystic Fibrosis/metabolism , Goblet Cells/metabolism , Mucin 5AC/metabolism , Mucin-5B/metabolism , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Goblet Cells/pathology , Mice , Mice, Knockout , Mucin 5AC/genetics , Mucin-5B/genetics
9.
PLoS Biol ; 15(5): e2000779, 2017 05.
Article in English | MEDLINE | ID: mdl-28510592

ABSTRACT

Synonymous single nucleotide polymorphisms (sSNPs) are considered neutral for protein function, as by definition they exchange only codons, not amino acids. We identified an sSNP that modifies the local translation speed of the cystic fibrosis transmembrane conductance regulator (CFTR), leading to detrimental changes to protein stability and function. This sSNP introduces a codon pairing to a low-abundance tRNA that is particularly rare in human bronchial epithelia, but not in other human tissues, suggesting tissue-specific effects of this sSNP. Up-regulation of the tRNA cognate to the mutated codon counteracts the effects of the sSNP and rescues protein conformation and function. Our results highlight the wide-ranging impact of sSNPs, which invert the programmed local speed of mRNA translation and provide direct evidence for the central role of cellular tRNA levels in mediating the actions of sSNPs in a tissue-specific manner.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , RNA, Transfer/metabolism , Silent Mutation , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , HEK293 Cells , HeLa Cells , Humans , Polymorphism, Single Nucleotide , Protein Stability , Structure-Activity Relationship
10.
JCI Insight ; 1(14): e88728, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27699238

ABSTRACT

The physiological components that contribute to cystic fibrosis (CF) lung disease are steadily being elucidated. Gene therapy could potentially correct these defects. CFTR-null pigs provide a relevant model to test gene therapy vectors. Using an in vivo selection strategy that amplifies successful capsids by replicating their genomes with helper adenovirus coinfection, we selected an adeno-associated virus (AAV) with tropism for pig airway epithelia. The evolved capsid, termed AAV2H22, is based on AAV2 with 5 point mutations that result in a 240-fold increased infection efficiency. In contrast to AAV2, AAV2H22 binds specifically to pig airway epithelia and is less reliant on heparan sulfate for transduction. We administer AAV2H22-CFTR expressing the CF transmembrane conductance regulator (CFTR) cDNA to the airways of CF pigs. The transduced airways expressed CFTR on ciliated and nonciliated cells, induced anion transport, and improved the airway surface liquid pH and bacterial killing. Most gene therapy studies to date focus solely on Cl- transport as the primary metric of phenotypic correction. Here, we describe a gene therapy experiment where we not only correct defective anion transport, but also restore bacterial killing in CFTR-null pig airways.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/therapeutic use , Cystic Fibrosis/therapy , Genetic Vectors , Animals , Dependovirus , Phenotype , Swine
11.
Proc Natl Acad Sci U S A ; 113(19): 5382-7, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27114540

ABSTRACT

Cystic fibrosis (CF) is caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) anion channel. Airway disease is the major source of morbidity and mortality. Successful implementation of gene- and cell-based therapies for CF airway disease requires knowledge of relationships among percentages of targeted cells, levels of CFTR expression, correction of electrolyte transport, and rescue of host defense defects. Previous studies suggested that, when ∼10-50% of airway epithelial cells expressed CFTR, they generated nearly wild-type levels of Cl(-) secretion; overexpressing CFTR offered no advantage compared with endogenous expression levels. However, recent discoveries focused attention on CFTR-mediated HCO3 (-) secretion and airway surface liquid (ASL) pH as critical for host defense and CF pathogenesis. Therefore, we generated porcine airway epithelia with varying ratios of CF and wild-type cells. Epithelia with a 50:50 mix secreted HCO3 (-) at half the rate of wild-type epithelia. Likewise, heterozygous epithelia (CFTR(+/-) or CFTR(+/∆F508)) expressed CFTR and secreted HCO3 (-) at ∼50% of wild-type values. ASL pH, antimicrobial activity, and viscosity showed similar relationships to the amount of CFTR. Overexpressing CFTR increased HCO3 (-) secretion to rates greater than wild type, but ASL pH did not exceed wild-type values. Thus, in contrast to Cl(-) secretion, the amount of CFTR is rate-limiting for HCO3 (-) secretion and for correcting host defense abnormalities. In addition, overexpressing CFTR might produce a greater benefit than expressing CFTR at wild-type levels when targeting small fractions of cells. These findings may also explain the risk of airway disease in CF carriers.


Subject(s)
Bicarbonates/immunology , Cystic Fibrosis Transmembrane Conductance Regulator/immunology , Cystic Fibrosis/immunology , Immunity, Innate/immunology , Respiratory Mucosa/immunology , Animals , Animals, Newborn , Cystic Fibrosis/therapy , Genetic Therapy , Stem Cell Transplantation , Swine
12.
Science ; 351(6272): 503-7, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26823428

ABSTRACT

Cystic fibrosis (CF) is caused by mutations in the gene that encodes the cystic fibrosis transmembrane conductance regulator (CFTR) anion channel. In humans and pigs, the loss of CFTR impairs respiratory host defenses, causing airway infection. But CF mice are spared. We found that in all three species, CFTR secreted bicarbonate into airway surface liquid. In humans and pigs lacking CFTR, unchecked H(+) secretion by the nongastric H(+)/K(+) adenosine triphosphatase (ATP12A) acidified airway surface liquid, which impaired airway host defenses. In contrast, mouse airways expressed little ATP12A and secreted minimal H(+); consequently, airway surface liquid in CF and non-CF mice had similar pH. Inhibiting ATP12A reversed host defense abnormalities in human and pig airways. Conversely, expressing ATP12A in CF mouse airways acidified airway surface liquid, impaired defenses, and increased airway bacteria. These findings help explain why CF mice are protected from infection and nominate ATP12A as a potential therapeutic target for CF.


Subject(s)
Cystic Fibrosis/metabolism , Cystic Fibrosis/microbiology , H(+)-K(+)-Exchanging ATPase/metabolism , Lung/metabolism , Lung/microbiology , Acids/metabolism , Animals , Bicarbonates/metabolism , H(+)-K(+)-Exchanging ATPase/genetics , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred CFTR/genetics , Mice, Inbred CFTR/metabolism , Mice, Transgenic , Swine
13.
J Clin Invest ; 126(3): 879-91, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26808501

ABSTRACT

Cystic fibrosis (CF) disrupts respiratory host defenses, allowing bacterial infection, inflammation, and mucus accumulation to progressively destroy the lungs. Our previous studies revealed that mucus with abnormal behavior impaired mucociliary transport in newborn CF piglets prior to the onset of secondary manifestations. To further investigate mucus abnormalities, here we studied airway surface liquid (ASL) collected from newborn piglets and ASL on cultured airway epithelia. Fluorescence recovery after photobleaching revealed that the viscosity of CF ASL was increased relative to that of non-CF ASL. CF ASL had a reduced pH, which was necessary and sufficient for genotype-dependent viscosity differences. The increased viscosity of CF ASL was not explained by pH-independent changes in HCO3- concentration, altered glycosylation, additional pH-induced disulfide bond formation, increased percentage of nonvolatile material, or increased sulfation. Treating acidic ASL with hypertonic saline or heparin largely reversed the increased viscosity, suggesting that acidic pH influences mucin electrostatic interactions. These findings link loss of cystic fibrosis transmembrane conductance regulator-dependent alkalinization to abnormal CF ASL. In addition, we found that increasing Ca2+ concentrations elevated ASL viscosity, in part, independently of pH. The results suggest that increasing pH, reducing Ca2+ concentration, and/or altering electrostatic interactions in ASL might benefit early CF.


Subject(s)
Cystic Fibrosis/metabolism , Mucus/metabolism , Respiratory Mucosa/metabolism , Alveolar Epithelial Cells/metabolism , Animals , Bicarbonates/metabolism , Carbohydrate Sequence , Cells, Cultured , Cystic Fibrosis/chemically induced , Female , Humans , Hydrogen-Ion Concentration , Male , Methacholine Chloride , Mucin 5AC/genetics , Mucin 5AC/metabolism , Mucin-5B/genetics , Mucin-5B/metabolism , Polysaccharides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sus scrofa , Viscosity
14.
Am J Respir Crit Care Med ; 193(4): 417-26, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26488271

ABSTRACT

RATIONALE: An asthma-like airway phenotype has been described in people with cystic fibrosis (CF). Whether these findings are directly caused by loss of CF transmembrane conductance regulator (CFTR) function or secondary to chronic airway infection and/or inflammation has been difficult to determine. OBJECTIVES: Airway contractility is primarily determined by airway smooth muscle. We tested the hypothesis that CFTR is expressed in airway smooth muscle and directly affects airway smooth muscle contractility. METHODS: Newborn pigs, both wild type and with CF (before the onset of airway infection and inflammation), were used in this study. High-resolution immunofluorescence was used to identify the subcellular localization of CFTR in airway smooth muscle. Airway smooth muscle function was determined with tissue myography, intracellular calcium measurements, and regulatory myosin light chain phosphorylation status. Precision-cut lung slices were used to investigate the therapeutic potential of CFTR modulation on airway reactivity. MEASUREMENTS AND MAIN RESULTS: We found that CFTR localizes to the sarcoplasmic reticulum compartment of airway smooth muscle and regulates airway smooth muscle tone. Loss of CFTR function led to delayed calcium reuptake following cholinergic stimulation and increased myosin light chain phosphorylation. CFTR potentiation with ivacaftor decreased airway reactivity in precision-cut lung slices following cholinergic stimulation. CONCLUSIONS: Loss of CFTR alters porcine airway smooth muscle function and may contribute to the airflow obstruction phenotype observed in human CF. Airway smooth muscle CFTR may represent a therapeutic target in CF and other diseases of airway narrowing.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiopathology , Sarcoplasmic Reticulum/physiology , Animals , Animals, Newborn , Blotting, Western , Fluorescent Antibody Technique , Lung/physiopathology , Models, Animal , Swine
15.
J Biol Chem ; 290(22): 14140-53, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25887396

ABSTRACT

The ATP-binding cassette (ABC) transporter cystic fibrosis transmembrane conductance regulator (CFTR) and two other non-membrane-bound ABC proteins, Rad50 and a structural maintenance of chromosome (SMC) protein, exhibit adenylate kinase activity in the presence of physiologic concentrations of ATP and AMP or ADP (ATP + AMP ⇆ 2 ADP). The crystal structure of the nucleotide-binding domain of an SMC protein in complex with the adenylate kinase bisubstrate inhibitor P(1),P(5)-di(adenosine-5') pentaphosphate (Ap5A) suggests that AMP binds to the conserved Q-loop glutamine during the adenylate kinase reaction. Therefore, we hypothesized that mutating the corresponding residue in CFTR, Gln-1291, selectively disrupts adenylate kinase-dependent channel gating at physiologic nucleotide concentrations. We found that substituting Gln-1291 with bulky side-chain amino acids abolished the effects of Ap5A, AMP, and adenosine 5'-monophosphoramidate on CFTR channel function. 8-Azidoadenosine 5'-monophosphate photolabeling of the AMP-binding site and adenylate kinase activity were disrupted in Q1291F CFTR. The Gln-1291 mutations did not alter the potency of ATP at stimulating current or ATP-dependent gating when ATP was the only nucleotide present. However, when physiologic concentrations of ADP and AMP were added, adenylate kinase-deficient Q1291F channels opened significantly less than wild type. Consistent with this result, we found that Q1291F CFTR displayed significantly reduced Cl(-) channel function in well differentiated primary human airway epithelia. These results indicate that a highly conserved residue of an ABC transporter plays an important role in adenylate kinase-dependent CFTR gating. Furthermore, the results suggest that adenylate kinase activity is important for normal CFTR channel function in airway epithelia.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenylate Kinase/genetics , Bronchi/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Glutamine/chemistry , Adenosine Diphosphate/chemistry , Adenosine Monophosphate/chemistry , Adenosine Triphosphate/chemistry , Adenylate Kinase/metabolism , Amino Acid Motifs , Binding Sites , Biotinylation , Chloride Channels/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Enzymologic , HeLa Cells , Humans , Immunohistochemistry , Mutagenesis, Site-Directed , Mutation , Patch-Clamp Techniques , Protein Binding
16.
Clin Sci (Lond) ; 128(2): 131-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25142104

ABSTRACT

Diabetes is a common and significant co-morbidity in cystic fibrosis (CF). The pathogenesis of cystic fibrosis related diabetes (CFRD) is incompletely understood. Because exocrine pancreatic disease is similar between humans and pigs with CF, the CF pig model has the potential to contribute significantly to the understanding of CFRD pathogenesis. We determined the structure of the endocrine pancreas in fetal, newborn and older CF and non-CF pigs and assessed endocrine pancreas function by intravenous glucose tolerance test (IV-GTT). In fetal pigs, pancreatic insulin and glucagon density was similar between CF and non-CF. In newborn and older pigs, the insulin and glucagon density was unchanged between CF and non-CF per total pancreatic area, but increased per remnant lobular tissue in CF reflecting exocrine pancreatic loss. Although fasting glucose levels were not different between CF and non-CF newborns, CF newborns demonstrated impaired glucose tolerance and increased glucose area under the curve during IV-GTT. Second phase insulin secretion responsiveness was impaired in CF newborn pigs and significantly lower than that observed in non-CF newborns. Older CF pigs had elevated random blood glucose levels compared with non-CF. In summary, glycaemic abnormalities and insulin secretion defects were present in newborn CF pigs and spontaneous hyperglycaemia developed over time. Functional changes in CF pig pancreas were not associated with a decline in islet cell mass. Our results suggest that functional islet abnormalities, independent of structural islet loss, contribute to the early pathogenesis of CFRD.


Subject(s)
Blood Glucose , Cystic Fibrosis/metabolism , Diabetes Mellitus/metabolism , Glucose Intolerance , Insulin/metabolism , Animals , Cystic Fibrosis/complications , Cystic Fibrosis/pathology , Diabetes Mellitus/pathology , Enzyme-Linked Immunosorbent Assay , Glucose Tolerance Test , Insulin/blood , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans/physiology , Pancreas/metabolism , Pancreas/pathology , Swine
17.
Science ; 345(6198): 818-22, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25124441

ABSTRACT

Lung disease in people with cystic fibrosis (CF) is initiated by defective host defense that predisposes airways to bacterial infection. Advanced CF is characterized by a deficit in mucociliary transport (MCT), a process that traps and propels bacteria out of the lungs, but whether this deficit occurs first or is secondary to airway remodeling has been unclear. To assess MCT, we tracked movement of radiodense microdisks in airways of newborn piglets with CF. Cholinergic stimulation, which elicits mucus secretion, substantially reduced microdisk movement. Impaired MCT was not due to periciliary liquid depletion; rather, CF submucosal glands secreted mucus strands that remained tethered to gland ducts. Inhibiting anion secretion in non-CF airways replicated CF abnormalities. Thus, impaired MCT is a primary defect in CF, suggesting that submucosal glands and tethered mucus may be targets for early CF treatment.


Subject(s)
Cystic Fibrosis/physiopathology , Exocrine Glands/metabolism , Mucociliary Clearance , Mucus/metabolism , Respiratory Mucosa/physiopathology , Respiratory System/physiopathology , Animals , Animals, Newborn , Anions/metabolism , Cilia/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Disease Models, Animal , Lung/physiopathology , Methacholine Chloride/pharmacology , Swine , Trachea/physiopathology
18.
J Clin Invest ; 123(6): 2685-93, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23676501

ABSTRACT

Cystic fibrosis (CF) pigs develop disease with features remarkably similar to those in people with CF, including exocrine pancreatic destruction, focal biliary cirrhosis, micro-gallbladder, vas deferens loss, airway disease, and meconium ileus. Whereas meconium ileus occurs in 15% of babies with CF, the penetrance is 100% in newborn CF pigs. We hypothesized that transgenic expression of porcine CF transmembrane conductance regulator (pCFTR) cDNA under control of the intestinal fatty acid-binding protein (iFABP) promoter would alleviate the meconium ileus. We produced 5 CFTR-/-;TgFABP>pCFTR lines. In 3 lines, intestinal expression of CFTR at least partially restored CFTR-mediated anion transport and improved the intestinal phenotype. In contrast, these pigs still had pancreatic destruction, liver disease, and reduced weight gain, and within weeks of birth, they developed sinus and lung disease, the severity of which varied over time. These data indicate that expressing CFTR in intestine without pancreatic or hepatic correction is sufficient to rescue meconium ileus. Comparing CFTR expression in different lines revealed that approximately 20% of wild-type CFTR mRNA largely prevented meconium ileus. This model may be of value for understanding CF pathophysiology and testing new preventions and therapies.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Cystic Fibrosis/metabolism , Ileus/metabolism , Animals , Animals, Genetically Modified , Animals, Newborn , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Models, Animal , Fatty Acid-Binding Proteins/genetics , Female , Gene Expression , Humans , Ileum/metabolism , Ileum/pathology , Ileus/pathology , Infant, Newborn , Lung/diagnostic imaging , Lung/metabolism , Lung/pathology , Male , Meconium/metabolism , Pancreas/metabolism , Pancreas/pathology , Phenotype , Promoter Regions, Genetic , Radiography , Rats , Sus scrofa , Trachea/metabolism , Trachea/pathology
19.
Am J Physiol Gastrointest Liver Physiol ; 303(8): G961-8, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22936270

ABSTRACT

The pancreas, liver, and gallbladder are commonly involved in cystic fibrosis (CF), and acidic, dehydrated, and protein-rich secretions are characteristic findings. Pancreatic function studies in humans have been done by sampling the jejunal fluid. However, it has been difficult to separately study the function of pancreatic and biliary systems in humans with CF, because jejunal fluid contains a mixture of bile and pancreatic fluids. In contrast, pancreatic and biliary ducts open separately into the porcine intestine; therefore, biliary and pancreatic fluid can be individually analyzed in CF pigs. We studied newborn wild-type (WT) and CF pigs and found that CFTR was localized to the pancreatic ducts. We collected bile and pancreatic fluid and analyzed pancreatic enzymes with activity assays and immunoblot. Pancreatic enzyme expression was significantly decreased in CF compared with WT pigs. The volume and pH of pancreatic fluid were significantly lower and protein concentration was >5-fold higher in CF pigs. Secretin stimulation increased pancreatic fluid volume and pH in WT, but not CF, pigs. Baseline bile volume did not differ between WT and CF pigs, but volume did not increase in response to secretin in CF pigs. Bile pH was lower and protein concentration was twofold higher in CF pigs. These results indicate that pancreatic and biliary secretions are altered in CF pigs. Abnormal pancreatic and biliary secretion in CF may have important implications in disease pathogenesis.


Subject(s)
Bile Ducts/metabolism , Cystic Fibrosis/metabolism , Pancreas/metabolism , Amylases/metabolism , Animals , Bile Ducts/physiopathology , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Models, Animal , Pancreas/physiopathology , Pancreatic Juice/metabolism , Swine , Trypsin/metabolism
20.
Proc Natl Acad Sci U S A ; 109(33): 13362-7, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22853952

ABSTRACT

Production of functional proteins requires multiple steps, including gene transcription and posttranslational processing. MicroRNAs (miRNAs) can regulate individual stages of these processes. Despite the importance of the cystic fibrosis transmembrane conductance regulator (CFTR) channel for epithelial anion transport, how its expression is regulated remains uncertain. We discovered that miRNA-138 regulates CFTR expression through its interactions with the transcriptional regulatory protein SIN3A. Treating airway epithelia with an miR-138 mimic increased CFTR mRNA and also enhanced CFTR abundance and transepithelial Cl(-) permeability independent of elevated mRNA levels. An miR-138 anti-miR had the opposite effects. Importantly, miR-138 altered the expression of many genes encoding proteins that associate with CFTR and may influence its biosynthesis. The most common CFTR mutation, ΔF508, causes protein misfolding, protein degradation, and cystic fibrosis. Remarkably, manipulating the miR-138 regulatory network also improved biosynthesis of CFTR-ΔF508 and restored Cl(-) transport to cystic fibrosis airway epithelia. This miRNA-regulated network directs gene expression from the chromosome to the cell membrane, indicating that an individual miRNA can control a cellular process more broadly than recognized previously. This discovery also provides therapeutic avenues for restoring CFTR function to cells affected by the most common cystic fibrosis mutation.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Gene Expression Regulation , Gene Regulatory Networks/genetics , MicroRNAs/metabolism , Biological Transport , Chlorides/metabolism , Epithelium/metabolism , Epithelium/pathology , Gene Expression Profiling , HeLa Cells , Humans , Lung/metabolism , Lung/pathology , MicroRNAs/genetics , Protein Processing, Post-Translational , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex
SELECTION OF CITATIONS
SEARCH DETAIL
...