79 results on '"David A Stoltz"'
Search Results
2. Sustained Coinfections with Staphylococcus aureus and Pseudomonas aeruginosa in Cystic Fibrosis
- Author
-
Mason M LaMarche, Dominique H. Limoli, Zoe E Kienenberger, Lucas J Maakestad, Sachinkumar B. Singh, David A. Stoltz, Tahuanty Pena, and Anthony J. Fischer
- Subjects
Pulmonary and Respiratory Medicine ,business.industry ,Pseudomonas aeruginosa ,Critical Care and Intensive Care Medicine ,medicine.disease_cause ,medicine.disease ,Cystic fibrosis ,Methicillin-resistant Staphylococcus aureus ,Persistence (computer science) ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,030228 respiratory system ,Older patients ,Staphylococcus aureus ,Coinfection ,Medicine ,Sputum ,030212 general & internal medicine ,medicine.symptom ,business - Abstract
Rationale: Staphylococcus aureus and Pseudomonas aeruginosa often infect the airways in cystic fibrosis (CF). Because registry studies show higher prevalence of P. aeruginosa versus S. aureus in older patients with CF, a common assumption is that P. aeruginosa replaces S. aureus over time. In vitro, P. aeruginosa can outgrow and kill S. aureus. However, it is unknown how rapidly P. aeruginosa replaces S. aureus in patients with CF.Methods: We studied a longitudinal cohort of children and adults with CF who had quantitative sputum cultures. We determined the abundance of P. aeruginosa and S. aureus in cfu/ml. We determined the duration and persistence of infections and measured longitudinal changes in culture positivity and abundance for each organism.Measurements and Main Results: Between 2004 and 2017, 134 patients had ≥10 quantitative cultures, with median observation time of 10.15 years. One hundred twenty-four patients had at least one positive culture for P. aeruginosa, and 123 had at least one positive culture for S. aureus. Both species had median abundance of >106 cfu/ml. Culture abundance was stable over time for both organisms. There was an increase in the prevalence of S. aureus/P. aeruginosa coinfection but no decrease in S. aureus prevalence within individuals over time.Conclusions: S. aureus and P. aeruginosa are abundant in CF sputum cultures. Contrary to common assumption, we found no pattern of replacement of S. aureus by P. aeruginosa. Many patients with CF have durable long-term coinfection with these organisms. New strategies are needed to prevent and treat these infections.
- Published
- 2021
- Full Text
- View/download PDF
3. Cystic fibrosis carriers are at increased risk for a wide range of cystic fibrosis-related conditions
- Author
-
Alicia K. Gerke, Aaron C. Miller, Joseph E. Cavanaugh, Douglas B. Hornick, Alejandro P. Comellas, David A. Stoltz, Michael J. Welsh, Joseph Zabner, and Philip M. Polgreen
- Subjects
0301 basic medicine ,education.field_of_study ,medicine.medical_specialty ,Multidisciplinary ,Bronchiectasis ,business.industry ,Population ,Odds ratio ,medicine.disease ,Cystic fibrosis ,Short stature ,Gastroenterology ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030228 respiratory system ,Diabetes mellitus ,Internal medicine ,Failure to thrive ,Cohort ,medicine ,medicine.symptom ,education ,business - Abstract
Autosomal recessive diseases, such as cystic fibrosis (CF), require inheritance of 2 mutated genes. However, some studies indicate that CF carriers are at increased risk for some conditions associated with CF. These investigations focused on single conditions and included small numbers of subjects. Our goal was to determine whether CF carriers are at increased risk for a range of CF-related conditions. Using the Truven Health MarketScan Commercial Claims database (2001–2017), we performed a population-based retrospective matched-cohort study. We identified 19,802 CF carriers and matched each carrier with 5 controls. The prevalence of 59 CF-related diagnostic conditions was evaluated in each cohort. Odds ratios for each condition were computed for CF carriers relative to controls. All 59 CF-related conditions were more prevalent among carriers compared with controls, with significantly increased risk ( P < 0.05) for 57 conditions. Risk was increased for some conditions previously linked to CF carriers (e.g., pancreatitis, male infertility, bronchiectasis), as well as some conditions not previously reported (e.g., diabetes, constipation, cholelithiasis, short stature, failure to thrive). We compared our results with 23,557 subjects with CF, who were also matched with controls; as the relative odds of a given condition increased among subjects with CF, so did the corresponding relative odds for carriers ( P < 0.001). Although individual-level risk remained low for most conditions, because there are more than 10 million carriers in the US, population-level morbidity attributable to the CF carrier state is likely substantial. Genetic testing may inform prevention, diagnosis, and treatment for a broad range of CF carrier-related conditions.
- Published
- 2019
- Full Text
- View/download PDF
4. A Novel AAV-mediated Gene Delivery System Corrects CFTR Function in Pigs
- Author
-
Patrick L. Sinn, David A. Stoltz, Brajesh K. Singh, Joseph Zabner, Viral Shah, Ian M. Thornell, Ashley L. Cooney, and Paul B. McCray
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Staphylococcus aureus ,Cystic Fibrosis ,Swine ,Virus Integration ,viruses ,Genetic enhancement ,Genetic Vectors ,Clinical Biochemistry ,Cystic Fibrosis Transmembrane Conductance Regulator ,Gene delivery ,medicine.disease_cause ,Cystic fibrosis ,Viral vector ,Gene Knockout Techniques ,03 medical and health sciences ,0302 clinical medicine ,Genes, Synthetic ,medicine ,Animals ,Humans ,Progenitor cell ,Promoter Regions, Genetic ,Molecular Biology ,Adeno-associated virus ,Transposase ,Original Research ,biology ,Chemistry ,Genetic Therapy ,Cell Biology ,Dependovirus ,respiratory system ,medicine.disease ,Molecular biology ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,Trachea ,030104 developmental biology ,Animals, Newborn ,030228 respiratory system ,DNA Transposable Elements ,biology.protein - Abstract
Cystic fibrosis is an autosomal-recessive disease that is caused by a mutant CFTR (cystic fibrosis transmembrane conductance regulator) gene and is characterized by chronic bacterial lung infections and inflammation. Complementation with functional CFTR normalizes anion transport across the airway surface. Adeno-associated virus (AAV) is a useful vector for gene therapy because of its low immunogenicity and ability to persist for months to years. However, because its episomal expression may decrease after cell division, readministration of the AAV vector may be required. To overcome this, we designed an integrating AAV-based CFTR-expressing vector, termed piggyBac (PB)/AAV, carrying CFTR flanked by the terminal repeats of the piggyBac transposon. With codelivery of the piggyBac transposase, PB/AAV can integrate into the host genome. Because of the packaging constraints of AAV, careful consideration was required to ensure that the vector would package and express its CFTR cDNA cargo. In this short-term study, PB/AAV-CFTR was aerosolized to the airways of CF pigs in the absence of the transposase. Two weeks later, transepithelial Cl(−) current was restored in freshly excised tracheal and bronchial tissue. Additionally, we observed an increase in tracheal airway surface liquid pH and bacterial killing in comparison with untreated CF pigs. Airway surface liquid from primary airway cells cultured from treated CF pigs exhibited increased pH correlating with decreased viscosity. Together, these results show that complementing CFTR in CF pigs with PB/AAV rescues the anion transport defect in a large-animal CF model. Delivery of this integrating viral vector system to airway progenitor cells could lead to persistent, life-long expression in vivo.
- Published
- 2019
- Full Text
- View/download PDF
5. Early intrahepatic duct defects in a cystic fibrosis porcine model
- Author
-
David K. Meyerholz, Keyan Zarei, and David A. Stoltz
- Subjects
Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Physiology ,Swine ,Intrahepatic bile ducts ,Cystic Fibrosis Transmembrane Conductance Regulator ,Cystic fibrosis ,Cholangiocyte ,Animals, Genetically Modified ,Physiology (medical) ,medicine ,QP1-981 ,Animals ,CFTR ,cholangiocytes ,organoids ,business.industry ,Gallbladder ,Hepatobiliary disease ,Original Articles ,medicine.disease ,Mucus ,Small intestine ,Disease Models, Animal ,medicine.anatomical_structure ,Bile Ducts, Intrahepatic ,Animals, Newborn ,Biliary tract ,Original Article ,intrahepatic bile ducts ,business - Abstract
Hepatobiliary disease causes significant morbidity and mortality in people with cystic fibrosis (CF), yet this problem remains understudied. Previous studies in the newborn CF pig demonstrated decreased bile flow into the small intestine and a microgallbladder with increased luminal mucus and fluid secretion defects. In this study, we examined the intrahepatic bile ducts of the newborn CF pig. We assessed whether our findings from the gallbladder are present elsewhere in the porcine biliary tract and if CF pig cholangiocytes have fluid secretion defects. Immunohistochemistry demonstrated apical CFTR expression in non‐CF pig intrahepatic bile ducts of a variety of sizes; CF pig intrahepatic bile ducts lacked CFTR expression. Assessment of serum markers did not reveal significant signs of hepatobiliary disease except for an elevation in direct bilirubin. Quantitative histology demonstrated that CF pigs had smaller bile ducts that more frequently contained luminal mucus. CF intrahepatic cholangiocyte organoids were smaller and lacked cAMP‐mediated fluid secretion. Together these data suggest that cholangiocyte fluid secretion is decreased in the CF pig, contributing to structural changes in bile ducts and decreased biliary flow., Using a porcine CF model, we discovered that intrahepatic ducts in newborn CF pigs have structural changes and are commonly plugged with mucus. CF intrahepatic cholangiocyte organoids had impaired cAMP‐mediated fluid secretion. Together these data suggest that impaired cholangiocyte fluid secretion contributes to structural changes in bile ducts, bile duct plugging, and decreased biliary flow.
- Published
- 2021
6. Inflammatory cytokines TNF-α and IL-17 enhance the efficacy of cystic fibrosis transmembrane conductance regulator modulators
- Author
-
Ping Tan, Alejandro A. Pezzulo, Tayyab Rehman, Ian M. Thornell, Michael J. Welsh, S.L. Durfey, Andrew L Thurman, Brian J Goodell, Edward F. McKone, David A. Stoltz, Michael E. Duffey, Philip H. Karp, and Pradeep K. Singh
- Subjects
medicine.medical_specialty ,Indoles ,Cystic Fibrosis ,Pyridines ,Cystic Fibrosis Transmembrane Conductance Regulator ,Inflammation ,Respiratory Mucosa ,Aminophenols ,Cystic fibrosis ,Proinflammatory cytokine ,Internal medicine ,medicine ,otorhinolaryngologic diseases ,Humans ,Secretion ,Benzodioxoles ,Respiratory system ,Cells, Cultured ,Ion Transport ,biology ,Chemistry ,Tumor Necrosis Factor-alpha ,Interleukin-17 ,Infant, Newborn ,Infant ,General Medicine ,Pendrin ,respiratory system ,Hydrogen-Ion Concentration ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,Bicarbonates ,Drug Combinations ,Endocrinology ,Sulfate Transporters ,Mutation ,biology.protein ,Quinolines ,Pyrazoles ,Interleukin 17 ,medicine.symptom ,Research Article - Abstract
Without cystic fibrosis transmembrane conductance regulator–mediated (CFTR-mediated) HCO(3)(–) secretion, airway epithelia of newborns with cystic fibrosis (CF) produce an abnormally acidic airway surface liquid (ASL), and the decreased pH impairs respiratory host defenses. However, within a few months of birth, ASL pH increases to match that in non-CF airways. Although the physiological basis for the increase is unknown, this time course matches the development of inflammation in CF airways. To learn whether inflammation alters CF ASL pH, we treated CF epithelia with TNF-α and IL-17 (TNF-α+IL-17), 2 inflammatory cytokines that are elevated in CF airways. TNF-α+IL-17 markedly increased ASL pH by upregulating pendrin, an apical Cl(–)/HCO(3)(–) exchanger. Moreover, when CF epithelia were exposed to TNF-α+IL-17, clinically approved CFTR modulators further alkalinized ASL pH. As predicted by these results, in vivo data revealed a positive correlation between airway inflammation and CFTR modulator–induced improvement in lung function. These findings suggest that inflammation is a key regulator of HCO(3)(–) secretion in CF airways. Thus, they explain earlier observations that ASL pH increases after birth and indicate that, for similar levels of inflammation, the pH of CF ASL is abnormally acidic. These results also suggest that a non-cell-autonomous mechanism, airway inflammation, is an important determinant of the response to CFTR modulators.
- Published
- 2021
7. Transduction of Pig Small Airway Epithelial Cells and Distal Lung Progenitor Cells by AAV4
- Author
-
Oliver G Chen, Christian M Brommel, Xiaopeng Li, Joseph Zabner, Annie Ehler, David A. Stoltz, Raul Villacreses, Reda E. Girgis, Steven E Mather, Bradley A. Hamilton, and Mahmoud H. Abou Alaiwa
- Subjects
0301 basic medicine ,Cystic Fibrosis ,Swine ,QH301-705.5 ,Genetic Vectors ,Cystic Fibrosis Transmembrane Conductance Regulator ,Respiratory Mucosa ,Biology ,Cystic fibrosis ,Article ,small airway epithelia ,Pathogenesis ,03 medical and health sciences ,Transduction (genetics) ,0302 clinical medicine ,In vivo ,medicine ,Animals ,Humans ,Progenitor cell ,CFTR ,Biology (General) ,Lung ,Tropism ,Cells, Cultured ,Stem Cells ,General Medicine ,progenitor cells ,Dependovirus ,respiratory system ,medicine.disease ,respiratory tract diseases ,030104 developmental biology ,medicine.anatomical_structure ,030228 respiratory system ,Cancer research ,AAV4 ,Respiratory epithelium ,cystic fibrosis 1 - Abstract
Cystic fibrosis (CF) is caused by genetic mutations of the CF transmembrane conductance regulator (CFTR), leading to disrupted transport of Cl− and bicarbonate and CF lung disease featuring bacterial colonization and chronic infection in conducting airways. CF pigs engineered by mutating CFTR develop lung disease that mimics human CF, and are well-suited for investigating CF lung disease therapeutics. Clinical data suggest small airways play a key role in the early pathogenesis of CF lung disease, but few preclinical studies have focused on small airways. Efficient targeted delivery of CFTR cDNA to small airway epithelium may correct the CFTR defect and prevent lung infections. Adeno-associated virus 4 (AAV4) is a natural AAV serotype and a safe vector with lower immunogenicity than other gene therapy vectors such as adenovirus. Our analysis of AAV natural serotypes using cultured primary pig airway epithelia showed that AAV4 has high tropism for airway epithelia and higher transduction efficiency for small airways compared with large airways. AAV4 mediated the delivery of CFTR, and corrected Cl− transport in cultured primary small airway epithelia from CF pigs. Moreover, AAV4 was superior to all other natural AAV serotypes in transducing ITGα6β4+ pig distal lung progenitor cells. In addition, AAV4 encoding eGFP can infect pig distal lung epithelia in vivo. This study demonstrates AAV4 tropism in small airway progenitor cells, which it efficiently transduces. AAV4 offers a novel tool for mechanistical study of the role of small airway in CF lung pathogenesis in a preclinical large animal model.
- Published
- 2021
8. Early Lung Disease Exhibits Bacteria-Dependent and -Independent Abnormalities in Cystic Fibrosis Pigs
- Author
-
Alejandro A. Pezzulo, Ryan J. Adam, Daniel P. Cook, Patrick Ten Eyck, Mahmoud H. Abou Alaiwa, David K. Meyerholz, Drake C. Bouzek, Chaorong Wu, David A. Stoltz, Maria I Aguilar Pescozo, Eric A. Hoffman, Douglas B. Hornick, Leah R. Reznikov, and Thomas J. Gross
- Subjects
Pulmonary and Respiratory Medicine ,Respiratory Mucosa ,Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Swine ,Respiratory physiology ,Disease pathogenesis ,Critical Care and Intensive Care Medicine ,Cystic fibrosis ,03 medical and health sciences ,0302 clinical medicine ,Medicine ,Animals ,Pseudomonas Infections ,030212 general & internal medicine ,biology ,business.industry ,Original Articles ,biology.organism_classification ,medicine.disease ,Mucus ,Anti-Bacterial Agents ,Young age ,030228 respiratory system ,Lung disease ,Tobramycin ,business ,Bacteria - Abstract
Rationale: Although it is clear that cystic fibrosis (CF) airway disease begins at a very young age, the early and subsequent steps in disease pathogenesis and the relative contribution of infection, mucus, and inflammation are not well understood. Objectives: As one approach to assessing the early contribution of infection, we tested the hypothesis that early and continuous antibiotics would decrease the airway bacterial burden. We believed that, if they do, this might reveal aspects of the disease that are more or less sensitive to decreasing infection. Methods: Three groups of pigs were studied from birth until ∼3 weeks of age: 1) wild-type, 2) CF, and 3) CF pigs treated continuously with broad-spectrum antibiotics from birth until study completion. Disease was assessed with chest computed tomography, histopathology, microbiology, and BAL. Measurements and Main Results: Disease was present by 3 weeks of age in CF pigs. Continuous antibiotics from birth improved chest computed tomography imaging abnormalities and airway mucus accumulation but not airway inflammation in the CF pig model. However, reducing bacterial infection did not improve two disease features already present at birth in CF pigs: air trapping and submucosal gland duct plugging. In the CF sinuses, antibiotics did not prevent the development of infection or disease or the number of bacteria but did alter the bacterial species. Conclusions: These findings suggest that CF airway disease begins immediately after birth and that early and continuous antibiotics impact some, but not all, aspects of CF lung disease development.
- Published
- 2021
9. Acidic Submucosal Gland pH and Elevated Protein Concentration Produce Abnormal Cystic Fibrosis Mucus
- Author
-
Yuliang Xie, Tony Jun Huang, David A. Stoltz, Thomas O. Moninger, Lin Lu, Michael J. Welsh, and Xiao Xiao Tang
- Subjects
medicine.medical_specialty ,Cystic Fibrosis ,Mucociliary clearance ,Swine ,Respiratory System ,Cystic Fibrosis Transmembrane Conductance Regulator ,Respiratory Mucosa ,Cystic fibrosis ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,fluids and secretions ,Internal medicine ,medicine ,Animals ,Humans ,Secretion ,Respiratory system ,Molecular Biology ,Lung ,030304 developmental biology ,Submucosal glands ,0303 health sciences ,biology ,Biological Transport ,Serum Albumin, Bovine ,Cell Biology ,respiratory system ,Hydrogen-Ion Concentration ,medicine.disease ,Mucus ,Cystic fibrosis transmembrane conductance regulator ,Trachea ,Endocrinology ,medicine.anatomical_structure ,biology.protein ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
In response to respiratory insults, airway submucosal glands secrete copious mucus strands to increase mucociliary clearance and protect the lung. However, in cystic fibrosis, stimulating submucosal glands has the opposite effect, disrupting mucociliary transport. In cystic fibrosis (CF) pigs, loss of cystic fibrosis transmembrane conductance regulator (CFTR) anion channels produced submucosal gland mucus that was abnormally acidic with an increased protein concentration. To test whether these variables alter mucus, we produced a microfluidic model of submucosal glands using mucus vesicles from banana slugs. Acidic pH and increased protein concentration decreased mucus gel volume and increased mucus strand elasticity and tensile strength. However, once mucus strands were formed, changing pH or protein concentration largely failed to alter the biophysical properties. Likewise, raising pH or apical perfusion did not improve clearance of mucus strands from CF airways. These findings reveal mechanisms responsible for impaired mucociliary transport in CF and have important implications for potential treatments.
- Published
- 2020
10. Glycogen depletion can increase the specificity of mucin detection in airway tissues
- Author
-
Amanda P. Beck, Heather A. Flaherty, Thomas R. Businga, Mariah R. Leidinger, David K. Meyerholz, David A. Stoltz, J. Adam Goeken, Hannah C. Brown, Leah R. Reznikov, and Georgina K. Ofori-Amanfo
- Subjects
Male ,0301 basic medicine ,Swine ,lcsh:Medicine ,Respiratory Mucosa ,Periodic acid–Schiff stain ,Alcian blue ,Sensitivity and Specificity ,Cystic fibrosis ,Stain ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Nonspecific staining ,medicine ,Animals ,lcsh:Science (General) ,lcsh:QH301-705.5 ,Staining ,Staining and Labeling ,Glycogen ,Chemistry ,Periodic acid Schiff (PAS) ,Mucin ,lcsh:R ,Mucins ,General Medicine ,Periodic Acid-Schiff Reaction ,medicine.disease ,Molecular biology ,Mucus ,Epithelium ,Diastase-periodic acid Schiff (dPAS) ,Trachea ,Research Note ,030104 developmental biology ,medicine.anatomical_structure ,Liver ,lcsh:Biology (General) ,Specificity ,Female ,Scoring ,lcsh:Q1-390 - Abstract
Objective Mucin is an important parameter for detection and assessment in studies of airway disease including asthma and cystic fibrosis. Histochemical techniques are often used to evaluate mucin in tissues sections. Periodic acid Schiff (PAS) is a common technique to detect neutral mucins in tissue, but this technique also detects other tissue components including cellular glycogen. We tested whether depletion of glycogen, a common cellular constituent, could impact the detection of mucin in the surface epithelium of the trachea. Results Normal tissues stained by PAS had significantly more staining than serial sections of glycogen-depleted tissue with PAS staining (i.e. dPAS technique) based on both quantitative analysis and semiquantitative scores. Most of the excess stain by the PAS technique was detected in ciliated cells adjacent to goblet cells. We also compared normal tissues using the Alcian blue technique, which does not have reported glycogen staining, with the dPAS technique. These groups had similar amounts of staining consistent with a high degree of mucin specificity. Our results suggest that when using PAS techniques to stain airways, the dPAS approach is preferred as it enhances the specificity for airway mucin.
- Published
- 2018
- Full Text
- View/download PDF
11. Widespread airway distribution and short-term phenotypic correction of cystic fibrosis pigs following aerosol delivery of piggyBac/adenovirus
- Author
-
Laura I. Marquez Loza, Brajesh K. Singh, Linda S. Powers, Ian M. Thornell, Ashley L. Cooney, Camilla E Hippee, Chris Wohlford-Lenane, David A. Stoltz, Lynda S. Ostedgaard, Paul B. McCray, Patrick L. Sinn, and David K. Meyerholz
- Subjects
0301 basic medicine ,Cell type ,Cystic Fibrosis ,Swine ,Genetic Vectors ,Cystic Fibrosis Transmembrane Conductance Regulator ,Respiratory Mucosa ,Cystic fibrosis ,Viral vector ,Adenoviridae ,03 medical and health sciences ,Transduction (genetics) ,Genetics ,medicine ,Animals ,Tissue Distribution ,Gene ,Molecular Biology ,Lung ,Aerosols ,biology ,Gene Transfer Techniques ,Epithelial Cells ,Genetic Therapy ,Anion channel activity ,respiratory system ,medicine.disease ,Molecular biology ,Cystic fibrosis transmembrane conductance regulator ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Phenotype ,Treatment Outcome ,biology.protein ,DNA Transposable Elements - 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.
- Published
- 2018
12. Lack of cystic fibrosis transmembrane conductance regulator disrupts fetal airway development in pigs
- Author
-
Erica N. LeClair, David A. Stoltz, Nicholas D. Gansemer, Katherine N. Gibson-Corley, Paul B. McCray, Michael J. Welsh, Mariah R. Leidinger, Sarah E. Ernst, Carrie K. Barker, Matthew D. Strub, Ryan J. Adam, Daniel P. Cook, Philip H. Karp, and David K. Meyerholz
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Swine ,Cystic Fibrosis Transmembrane Conductance Regulator ,Fibroblast growth factor ,Cystic fibrosis ,Article ,Pathology and Forensic Medicine ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Cyclic AMP ,Morphogenesis ,medicine ,Animals ,Humans ,Lung ,Molecular Biology ,Cells, Cultured ,FGF10 ,biology ,business.industry ,Cell Biology ,respiratory system ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,Epithelium ,respiratory tract diseases ,Trachea ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Female ,business ,Airway ,Fibroblast Growth Factor 10 ,030217 neurology & neurosurgery - Abstract
Loss of cystic fibrosis transmembrane conductance regulator (CFTR) function causes cystic fibrosis (CF), predisposing the lungs to chronic infection and inflammation. In young infants with CF, structural airway defects are increasingly recognized before the onset of significant lung disease, which suggests a developmental origin and a possible role in lung disease pathogenesis. The role(s) of CFTR in lung development is unclear and developmental studies in humans with CF are not feasible. Young CF pigs have structural airway changes and develop spontaneous postnatal lung disease similar to humans, therefore, we studied lung development in the pig model (non-CF and CF). CF trachea and proximal airways had structural lesions detectable as early as pseudoglandular development. At this early developmental stage, budding CF airways had smaller, hypo-distended lumens compared to non-CF airways. Non-CF lung explants exhibited airway lumen distension in response to forskolin/IBMX as well as to fibroblast growth factor (FGF)-10, consistent with CFTR-dependent anion transport/secretion, but this was lacking in CF airways. We studied primary pig airway epithelial cell cultures and found that that FGF10 increased cellular proliferation (non-CF and CF) and CFTR expression/function (in non-CF only). In pseudoglandular stage lung tissue, CFTR protein was exclusively localized to the leading edges of budding airways in non-CF (but not CF) lungs. This discreet microanatomic localization of CFTR is consistent with the site, during branching morphogenesis, where airway epithelia are responsive to FGF10 regulation. In summary, our results suggest that the CF proximal airway defects originate during branching morphogenesis and that the lack of CFTR-dependent anion transport/liquid secretion likely contributes to these hypo-distended airways.
- Published
- 2018
- Full Text
- View/download PDF
13. 679: Host defense defects and inflammation within the nasal airways of CFTR knockout mice
- Author
-
Ian M. Thornell, C. Cano Portillo, P. Allen, E. Stapleton, J. Ash, A. Comellas, Alejandro A. Pezzulo, Lynda S. Ostedgaard, W. Yu, Michael J. Welsh, Steven E Mather, David A. Stoltz, Joseph Zabner, and David K. Meyerholz
- Subjects
Pulmonary and Respiratory Medicine ,business.industry ,Host (biology) ,Pediatrics, Perinatology and Child Health ,Immunology ,Knockout mouse ,medicine ,Inflammation ,medicine.symptom ,medicine.disease ,business ,Cystic fibrosis - Published
- 2021
- Full Text
- View/download PDF
14. 362: Multicolor flow cytometry approach to study airway injury and reepithelialization in primary human cell culture
- Author
-
G. Romano Ibarra, N. Gansamer, David A. Stoltz, Ian M. Thornell, and Alejandro A. Pezzulo
- Subjects
Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Primary (chemistry) ,medicine.diagnostic_test ,business.industry ,Human cell ,medicine.disease ,Cystic fibrosis ,Flow cytometry ,Pediatrics, Perinatology and Child Health ,medicine ,business ,Airway - Published
- 2021
- Full Text
- View/download PDF
15. 217: Pancreatic enzyme treatment of obstructive meconium from CF pigs
- Author
-
I. Thornell, David A. Stoltz, G. Gangadharan Nambiar, and A. Fischer
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Meconium ,business.industry ,Internal medicine ,Pediatrics, Perinatology and Child Health ,medicine ,medicine.disease ,business ,Cystic fibrosis ,Pancreatic enzymes ,Gastroenterology - Published
- 2021
- Full Text
- View/download PDF
16. 517: Regional evolution of Pseudomonas aeruginosa in the human host
- Author
-
S.L. Durfey, T. Pena, J. Brewington, Pradeep K. Singh, M. Stroik, David A. Stoltz, Siddhartha G. Kapnadak, Sachinkumar B. Singh, Moira L. Aitken, Anh T. Vo, M. Teresi, J. Godwin, Linda D. Boyken, Hillary S. Hayden, and Matthew C. Radey
- Subjects
Pulmonary and Respiratory Medicine ,business.industry ,Pseudomonas aeruginosa ,Host (biology) ,Pediatrics, Perinatology and Child Health ,Medicine ,business ,medicine.disease_cause ,medicine.disease ,Cystic fibrosis ,Microbiology - Published
- 2021
- Full Text
- View/download PDF
17. Cystic Fibrosis Transmembrane Conductance Regulator Potentiation as a Therapeutic Strategy for Pulmonary Edema
- Author
-
Ryan J. Adam, David A. Stoltz, Luis G. Vargas Buonfiglio, Joseph Zabner, Alejandro P. Comellas, and Xiaopeng Li
- Subjects
Pathology ,medicine.medical_specialty ,Swine ,Hydrostatic pressure ,Volume overload ,Cystic Fibrosis Transmembrane Conductance Regulator ,Pulmonary Edema ,Quinolones ,030204 cardiovascular system & hematology ,Aminophenols ,Critical Care and Intensive Care Medicine ,Article ,Ivacaftor ,03 medical and health sciences ,0302 clinical medicine ,Edema ,Administration, Inhalation ,Animals ,Medicine ,Prospective Studies ,Chloride Channel Agonists ,Lung ,biology ,business.industry ,Pulmonary edema ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,Disease Models, Animal ,medicine.anatomical_structure ,030228 respiratory system ,Alveolar Epithelial Cells ,biology.protein ,medicine.symptom ,business ,Ex vivo ,medicine.drug - Abstract
Objectives To determine the feasibility of using a cystic fibrosis transmembrane conductance regulator potentiator, ivacaftor (VX-770/Kalydeco, Vertex Pharmaceuticals, Boston, MA), as a therapeutic strategy for treating pulmonary edema. Design Prospective laboratory animal investigation. Setting Animal research laboratory. Subjects Newborn and 3 days to 1 week old pigs. Interventions Hydrostatic pulmonary edema was induced in pigs by acute volume overload. Ivacaftor was nebulized into the lung immediately after volume overload. Grams of water per grams of dry lung tissue were determined in the lungs harvested 1 hour after volume overload. Measurements and main results Ivacaftor significantly improved alveolar liquid clearance in isolated pig lung lobes ex vivo and reduced edema in a volume overload in vivo pig model of hydrostatic pulmonary edema. To model hydrostatic pressure-induced edema in vitro, we developed a method of applied pressure to the basolateral surface of alveolar epithelia. Elevated hydrostatic pressure resulted in decreased cystic fibrosis transmembrane conductance regulator activity and liquid absorption, an effect which was partially reversed by cystic fibrosis transmembrane conductance regulator potentiation with ivacaftor. Conclusions Cystic fibrosis transmembrane conductance regulator potentiation by ivacaftor is a novel therapeutic approach for pulmonary edema.
- Published
- 2017
- Full Text
- View/download PDF
18. Postnatal airway growth in cystic fibrosis piglets
- Author
-
Eric A. Hoffman, Daniel P. Cook, Ryan J. Adam, Linda S. Powers, Mallory R. Stroik, Peter J. Taft, Nicholas D. Gansemer, David A. Stoltz, David K. Meyerholz, Joseph Zabner, Mahmoud H. Abou Alaiwa, Mark J. Hoegger, James D. McMenimen, Michael J. Welsh, and Drake C. Bouzek
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Swine ,Physiology ,Population ,Cystic Fibrosis Transmembrane Conductance Regulator ,Bronchi ,Computed tomography ,Cystic fibrosis ,Animals, Genetically Modified ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,medicine ,Animals ,Lung volumes ,education ,Lung ,Cause of death ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,respiratory system ,medicine.disease ,respiratory tract diseases ,Trachea ,030104 developmental biology ,Animals, Newborn ,030228 respiratory system ,In utero ,Airway ,business ,Research Article - Abstract
Mutations in the gene encoding the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) anion channel cause CF. The leading cause of death in the CF population is lung disease. Increasing evidence suggests that in utero airway development is CFTR-dependent and that developmental abnormalities may contribute to CF lung disease. However, relatively little is known about postnatal CF airway growth, largely because such studies are limited in humans. Therefore, we examined airway growth and lung volume in a porcine model of CF. We hypothesized that CF pigs would have abnormal postnatal airway growth. To test this hypothesis, we performed CT-based airway and lung volume measurements in 3-wk-old non-CF and CF pigs. We found that 3-wk-old CF pigs had tracheas of reduced caliber and irregular shape. Their bronchial lumens were reduced in size proximally but not distally, were irregularly shaped, and had reduced distensibility. Our data suggest that lack of CFTR results in aberrant postnatal airway growth and development, which could contribute to CF lung disease pathogenesis. NEW & NOTEWORTHY This CT scan-based study of airway morphometry in the cystic fibrosis (CF) postnatal period is unique, as analogous studies in humans are greatly limited for ethical and technical reasons. Findings such as reduced airway lumen area and irregular caliber suggest that airway growth and development are CF transmembrane conductance regulator-dependent and that airway growth defects may contribute to CF lung disease pathogenesis.
- Published
- 2017
- Full Text
- View/download PDF
19. Restoring Cystic Fibrosis Transmembrane Conductance Regulator Function Reduces Airway Bacteria and Inflammation in People with Cystic Fibrosis and Chronic Lung Infections
- Author
-
Sonya L. Heltshe, S. Carter, James E. Bruce, Rachael M. Edwards, Matthew C. Radey, Michael J. Welsh, Gordon Cooke, Christopher E. Pope, Xia Wu, Pradeep K. Singh, Charles G. Gallagher, Peter Jorth, Katherine B. Hisert, Daniel J. Wolter, Frank J. Accurso, B. Grogan, Ryan J. Adam, Edward F. McKone, Seamas C. Donnelly, David A. Stoltz, Lucas R. Hoffman, and Janice L. Launspach
- Subjects
Adult ,Male ,0301 basic medicine ,Pulmonary and Respiratory Medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Cystic Fibrosis ,Cystic Fibrosis Transmembrane Conductance Regulator ,Inflammation ,Quinolones ,Aminophenols ,Critical Care and Intensive Care Medicine ,medicine.disease_cause ,Cystic fibrosis ,Ivacaftor ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Chloride Channel Agonists ,Lung ,Respiratory Tract Infections ,biology ,business.industry ,Pseudomonas aeruginosa ,Sputum ,respiratory system ,medicine.disease ,biology.organism_classification ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,030104 developmental biology ,030228 respiratory system ,Immunology ,biology.protein ,Female ,medicine.symptom ,Tomography, X-Ray Computed ,Airway ,business ,Bacteria ,medicine.drug - Abstract
Rationale: Previous work indicates that ivacaftor improves cystic fibrosis transmembrane conductance regulator (CFTR) activity and lung function in people with cystic fibrosis and G551D-CFTR mutations but does not reduce density of bacteria or markers of inflammation in the airway. These findings raise the possibility that infection and inflammation may progress independently of CFTR activity once cystic fibrosis lung disease is established. Objectives: To better understand the relationship between CFTR activity, airway microbiology and inflammation, and lung function in subjects with cystic fibrosis and chronic airway infections. Methods: We studied 12 subjects with G551D-CFTR mutations and chronic airway infections before and after ivacaftor. We measured lung function, sputum bacterial content, and inflammation, and obtained chest computed tomography scans. Measurements and Main Results: Ivacaftor produced rapid decreases in sputum Pseudomonas aeruginosa density that began within 48 hours and continued in the first year of treatment. However, no subject eradicated their infecting P. aeruginosa strain, and after the first year P. aeruginosa densities rebounded. Sputum total bacterial concentrations also decreased, but less than P. aeruginosa. Sputum inflammatory measures decreased significantly in the first week of treatment and continued to decline over 2 years. Computed tomography scans obtained before and 1 year after ivacaftor treatment revealed that ivacaftor decreased airway mucous plugging. Conclusions: Ivacaftor caused marked reductions in sputum P. aeruginosa density and airway inflammation and produced modest improvements in radiographic lung disease in subjects with G551D-CFTR mutations. However, P. aeruginosa airway infection persisted. Thus, measures that control infection may be required to realize the full benefits of CFTR-targeting treatments.
- Published
- 2017
- Full Text
- View/download PDF
20. Gel-forming mucins form distinct morphologic structures in airways
- Author
-
Drake C. Bouzek, Mahmoud H. Abou Alaiwa, Daniel P. Cook, Connor P. Parker, David K. Meyerholz, Linda S. Powers, Nicholas M. Sawin, Nicholas D. Gansemer, David A. Stoltz, Ian M. Thornell, Thomas O. Moninger, Michael J. Welsh, Lynda S. Ostedgaard, and James D. McMenimen
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Inflammation ,Mucin 5AC ,Biology ,Cystic fibrosis ,Gel forming ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Secretion ,Mice, Knockout ,Submucosal glands ,Multidisciplinary ,Lung ,Mucin ,Biological Sciences ,respiratory system ,medicine.disease ,Mucin-5B ,Mucus ,030104 developmental biology ,medicine.anatomical_structure ,030228 respiratory system ,Airway Remodeling ,Goblet Cells ,medicine.symptom - 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.
- Published
- 2017
- Full Text
- View/download PDF
21. Mounier–Kuhn syndrome: a case of tracheal smooth muscle remodeling
- Author
-
Julia Klesney-Tait, Mahmoud H. Abou Alaiwa, David A. Stoltz, Daniel P. Cook, Kalpaj R. Parekh, David K. Meyerholz, Michael Eberlein, and Ryan J. Adam
- Subjects
Pathology ,medicine.medical_specialty ,Case Report ,Disease ,Case Reports ,03 medical and health sciences ,0302 clinical medicine ,Smooth muscle ,Medicine ,030212 general & internal medicine ,tracheobronchomalacia ,business.industry ,General Medicine ,Airway smooth muscle ,Airway remodeling ,medicine.disease ,Recurrent lower respiratory tract infection ,airway smooth muscle ,3. Good health ,030228 respiratory system ,Tracheobronchomalacia ,Mounier-Kuhn syndrome ,Mounier–Kuhn syndrome ,Etiology ,Dilation (morphology) ,business - Abstract
Key Clinical Message Mounier–Kuhn syndrome is a rare clinical disorder characterized by tracheobronchial dilation and recurrent lower respiratory tract infections. While the etiology of the disease remains unknown, histopathological analysis of Mounier–Kuhn airways demonstrates that the disease is, in part, characterized by cellular changes in airway smooth muscle.
- Published
- 2016
22. Newborn Cystic Fibrosis Pigs Have a Blunted Early Response to an Inflammatory Stimulus
- Author
-
Michael J. Welsh, Carrie K. Barker, David K. Meyerholz, David A. Stoltz, Alejandro A. Pezzulo, Joseph Zabner, Jennifer A. Bartlett, Christine L. Wohlford-Lenane, Shyam Ramachandran, and Paul B. McCray
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Staphylococcus aureus ,Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Genotype ,Swine ,Apoptosis ,Inflammation ,Respiratory Mucosa ,In Vitro Techniques ,Stimulus (physiology) ,Critical Care and Intensive Care Medicine ,medicine.disease_cause ,Cystic fibrosis ,Epithelium ,Proinflammatory cytokine ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Animals ,Medicine ,Lung ,Cell Proliferation ,medicine.diagnostic_test ,business.industry ,respiratory system ,medicine.disease ,030104 developmental biology ,Bronchoalveolar lavage ,Animals, Newborn ,030228 respiratory system ,Models, Animal ,Immunology ,Disease Progression ,Original Article ,medicine.symptom ,business ,Signal Transduction - Abstract
Studies suggest that inappropriate responses to proinflammatory stimuli might contribute to inflammation in cystic fibrosis (CF) lungs. However, technical challenges have made it difficult to distinguish whether altered responses in CF airways are an intrinsic defect or a secondary effect of chronic disease in their tissue of origin. The CF pig model provides an opportunity to study the inflammatory responses of CF airways at birth, before the onset of infection and inflammation.To test the hypothesis that acute inflammatory responses are perturbed in porcine CF airways.We investigated the inflammatory responses of newborn CF and non-CF pig airways following a 4-hour exposure to heat-killed Staphylococcus aureus, in vivo and in vitro.Following an in vivo S. aureus challenge, markers of inflammation were similar between CF and littermate control animals through evaluation of bronchoalveolar lavage and tissues. However, transcriptome analysis revealed genotype-dependent differences as CF pigs showed a diminished host defense response compared with their non-CF counterparts. Furthermore, CF pig airways exhibited an increase in apoptotic pathways and a suppression of ciliary and flagellar biosynthetic pathways. Similar differences were observed in cultured airway epithelia from CF and non-CF pigs exposed to the stimulus.Transcriptome profiling suggests that acute inflammatory responses are dysregulated in the airways of newborn CF pigs.
- Published
- 2016
- Full Text
- View/download PDF
23. CFTR Heterozygotes Are at Increased Risk of Respiratory Infections: A Population-Based Study
- Author
-
Douglas B. Hornick, Ferhaan Ahmad, David A. Stoltz, Grant D. Brown, Philip M. Polgreen, Alejandro P. Comellas, and Barry London
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Genetic counseling ,Population ,Cystic fibrosis ,Major Articles ,03 medical and health sciences ,0302 clinical medicine ,respiratory infection ,Internal medicine ,medicine ,education ,education.field_of_study ,Respiratory tract infections ,biology ,business.industry ,Respiratory infection ,Odds ratio ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,3. Good health ,030104 developmental biology ,Infectious Diseases ,030228 respiratory system ,Oncology ,Cohort ,biology.protein ,antimicrobial ,CFTR heterozygote ,business - Abstract
Background Patients heterozygous for mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene may be more susceptible to respiratory infections than the general population. Methods. We conducted a retrospective case–control study using health insurance claims. We identified patients as either highly likely to be CFTR heterozygotes (CF carriers diagnosed during genetic counseling, parents of children with a diagnosis of CF, and children of mothers diagnosed with CF) or likely CFTR heterozygotes (children of CF carriers diagnosed during genetic counseling and parents of CF carriers diagnosed during genetic counseling). Next, we examined the rates of respiratory infections and antimicrobial prescriptions between both groups of CFTR patients and only the highly likely subcohort, compared with age/sex-matched controls. We examined the presence of any claim using McNemar’s test and the number of claims using the sign test. Results CFTR heterozygotes (the pooled highly likely and likely heterozygotes) were more prone to have at least 1 claim for a respiratory infection (odds ratio [OR], 1.28; P = .020) and to have a greater number of claims for respiratory infections (53.5%; P = .043) than controls. Patients in the highly likely cohort were also more prone to have at least 1 claim for a respiratory infection (OR, 1.30; P = .028) and more claims (54.3%; P = .039) than controls. In addition, the highly likely CFTR heterozygotes were more prone to be prescribed an antibiotic used to treat respiratory infections (OR, 1.34; P = .018) and to have more of these prescriptions (54.3%; P = .035) than controls. Conclusions Patients heterozygous for CFTR mutations are at higher risk for respiratory infections. Future work to describe clinical outcomes for CFTR heterozygotes is needed.
- Published
- 2018
- Full Text
- View/download PDF
24. Ivacaftor-induced sweat chloride reductions correlate with increases in airway surface liquid pH in cystic fibrosis
- Author
-
Edward F. McKone, Joseph Zabner, Jan L. Launspach, B. Grogan, Pradeep K. Singh, Michael J. Welsh, Mahmoud H. Abou Alaiwa, S. Carter, and David A. Stoltz
- Subjects
Male ,0301 basic medicine ,Cystic Fibrosis ,Cystic Fibrosis Transmembrane Conductance Regulator ,Quinolones ,Aminophenols ,Cystic fibrosis ,Pulmonary function testing ,SWEAT ,Ivacaftor ,0302 clinical medicine ,Longitudinal Studies ,Sweat ,Lung ,biology ,Chemistry ,General Medicine ,Hydrogen-Ion Concentration ,Middle Aged ,respiratory system ,Cystic fibrosis transmembrane conductance regulator ,Female ,Bronchoalveolar Lavage Fluid ,medicine.drug ,Adult ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Sweat chloride ,Biological Transport, Active ,Respiratory Mucosa ,Young Adult ,03 medical and health sciences ,Chlorides ,In vivo ,Internal medicine ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Ion Transport ,Infant, Newborn ,Infant ,medicine.disease ,respiratory tract diseases ,Bicarbonates ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,030228 respiratory system ,Mutation ,biology.protein ,Clinical Medicine ,Airway - Abstract
Background Disruption of cystic fibrosis transmembrane conductance regulator (CFTR) anion channel function causes cystic fibrosis (CF), and lung disease produces most of the mortality. Loss of CFTR-mediated HCO3- secretion reduces the pH of airway surface liquid (ASL) in vitro and in neonatal humans and pigs in vivo. However, we previously found that, in older children and adults, ASL pH does not differ between CF and non-CF. Here, we tested whether the pH of CF ASL increases with time after birth. Finding that it did suggested that adaptations by CF airways increase ASL pH. This conjecture predicted that increasing CFTR activity in CF airways would further increase ASL pH and also that increasing CFTR activity would correlate with increases in ASL pH. Methods To test for longitudinal changes, we measured ASL pH in newborns and then at 3-month intervals. We also studied people with CF (bearing G551D or R117H mutations), in whom we could acutely stimulate CFTR activity with ivacaftor. To gauge changes in CFTR activity, we measured changes in sweat Cl- concentration immediately before and 48 hours after starting ivacaftor. Results Compared with that in the newborn period, ASL pH increased by 6 months of age. In people with CF bearing G551D or R117H mutations, ivacaftor did not change the average ASL pH; however reductions in sweat Cl- concentration correlated with elevations of ASL pH. Reductions in sweat Cl- concentration also correlated with improvements in pulmonary function. Conclusions Our results suggest that CFTR-independent mechanisms increase ASL pH in people with CF. We speculate that CF airway disease, which begins soon after birth, is responsible for the adaptation. Funding Vertex Inc., the NIH (P30DK089507, 1K08HL135433, HL091842, HL136813, K24HL102246), the Cystic Fibrosis Foundation (SINGH17A0 and SINGH15R0), and the Burroughs Wellcome Fund.
- Published
- 2018
- Full Text
- View/download PDF
25. Airway acidification initiates host defense abnormalities in cystic fibrosis mice
- Author
-
Leah R. Reznikov, Patrick D Allen, Lynda S. Ostedgaard, David A. Stoltz, Xiao Xiao Tang, Christoph O. Randak, Sarah E. Ernst, Mariah R. Leidinger, Viral Shah, Mahmoud H. Abou Alaiwa, Christine L. Wohlford-Lenane, Michael J. Welsh, Kristopher P. Heilmann, Paul B. McCray, Philip H. Karp, David K. Meyerholz, and Joseph Zabner
- Subjects
0301 basic medicine ,Cystic Fibrosis ,Swine ,Transgene ,Mice, Transgenic ,Biology ,H(+)-K(+)-Exchanging ATPase ,Cystic fibrosis ,Article ,Microbiology ,Mice ,03 medical and health sciences ,medicine ,Animals ,Humans ,Mice, Inbred CFTR ,Secretion ,Respiratory system ,Lung ,Multidisciplinary ,respiratory system ,Hydrogen-Ion Concentration ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,Bicarbonates ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,biology.protein ,Airway ,Acids - Abstract
Airway infections put to an acid test Most people with cystic fibrosis suffer from chronic respiratory infections. The mechanistic link between this symptom and the genetic cause of the disease (mutations that compromise the function of the cystic fibrosis transmembrane conductance regulator, CFTR) is not fully understood. Studying animal models, Shah et al. find that in the absence of functional CFTR, the surface liquid in the airways becomes acidic, which impairs host defenses against infection. This acidification occurs through the action of a proton pump called ATP12A. Molecules inhibiting ATP12A could potentially be developed into useful drugs. Science , this issue p. 503
- Published
- 2016
- Full Text
- View/download PDF
26. Sonographic evidence of abnormal tracheal cartilage ring structure in cystic fibrosis
- Author
-
Simon C. Kao, David A. Stoltz, Amit Diwakar, Kathryn Chaloner, Andrew S. Michalski, Rebecca A. Horan, Monelle M. Tamegnon, Ryan J. Adam, Anthony J. Fischer, David K. Meyerholz, and Jan L. Launspach
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Adolescent ,Cystic Fibrosis ,Swine ,Cystic fibrosis ,Article ,Young Adult ,Animals ,Humans ,Medicine ,business.industry ,Cartilage ,Ultrasound ,Anatomy ,respiratory system ,medicine.disease ,Sagittal plane ,Trachea ,medicine.anatomical_structure ,Otorhinolaryngology ,Female ,Ultrasonography ,business ,Airway - Abstract
Objectives/Hypothesis Tracheal cartilage ring structural abnormalities have been reported in cystic fibrosis (CF) mice and pigs. Whether similar findings are present in humans with CF is unknown. We hypothesized that tracheal cartilage ring shape and size would be different in people with CF. Study Design Tracheal cartilage ring size and shape were measured in adults with (n = 21) and without CF (n = 18). Methods Ultrasonography was used in human subjects to noninvasively assess tracheal cartilage ring structure in both the sagittal and the transverse planes. Tracheal cartilage ring thickness was also determined from histological sections obtained from newborn non-CF and CF pigs. These values were compared with human data. Results Human CF tracheas had a greater width and were less circular in shape compared to non-CF subjects. CF tracheal cartilage rings had a greater midline cross-sectional area and were thicker compared to non-CF rings. Maximal tracheal cartilage ring thickness was also greater in both newborn CF pigs and human adults with CF, compared to non-CF controls. Conclusions Our findings demonstrate that structural differences exist in tracheal cartilage rings in adults with CF. Comparison with newborn CF pig data suggests that some of these changes may be congenital in nature. Level of Evidence 3b Laryngoscope, 125:2398–2404, 2015
- Published
- 2015
- Full Text
- View/download PDF
27. Origins of Cystic Fibrosis Lung Disease
- Author
-
David K. Meyerholz, Michael J. Welsh, and David A. Stoltz
- Subjects
Mutation ,Lung ,biology ,business.industry ,General Medicine ,respiratory system ,medicine.disease ,medicine.disease_cause ,Lung pathology ,Cystic fibrosis ,Article ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,medicine.anatomical_structure ,Lung disease ,Immunology ,medicine ,biology.protein ,business - Abstract
Mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator undermine many host defense systems by inhibiting the function of airway-surface liquid, causing flaws in mucociliary transport, and compromising other lung-protection mechanisms.
- Published
- 2015
- Full Text
- View/download PDF
28. CF airway smooth muscle transcriptome reveals a role for PYK2
- Author
-
Mallory R. Stroik, Kin Fai Au, Nicholas D. Gansemer, David K. Meyerholz, Ryan J. Adam, Benjamin Deonovic, David A. Stoltz, Keyan Zarei, and Daniel P. Cook
- Subjects
0301 basic medicine ,Myosin Light Chains ,Contraction (grammar) ,Cystic Fibrosis ,Swine ,Cystic Fibrosis Transmembrane Conductance Regulator ,Gene Expression ,Bronchi ,Inflammation ,Cystic fibrosis ,Transcriptome ,03 medical and health sciences ,Respiratory Hypersensitivity ,Animals ,Medicine ,RNA, Messenger ,Phosphorylation ,business.industry ,Muscle, Smooth ,General Medicine ,Smooth muscle contraction ,respiratory system ,medicine.disease ,respiratory tract diseases ,3. Good health ,Cell biology ,Focal Adhesion Kinase 2 ,030104 developmental biology ,Animals, Newborn ,Tyrosine kinase 2 ,medicine.symptom ,business ,Airway ,Research Article - Abstract
Abnormal airway smooth muscle function can contribute to cystic fibrosis (CF) airway disease. We previously found that airway smooth muscle from newborn CF pigs had increased basal tone, an increased bronchodilator response, and abnormal calcium handling. Since CF pigs lack airway infection and inflammation at birth, these findings suggest intrinsic airway smooth muscle dysfunction in CF. In this study, we tested the hypothesis that CFTR loss in airway smooth muscle would produce a distinct set of changes in the airway smooth muscle transcriptome that we could use to develop novel therapeutic targets. Total RNA sequencing of newborn wild-type and CF airway smooth muscle revealed changes in muscle contraction-related genes, ontologies, and pathways. Using connectivity mapping, we identified several small molecules that elicit transcriptional signatures opposite of CF airway smooth muscle, including NVP-TAE684, an inhibitor of proline-rich tyrosine kinase 2 (PYK2). In CF airway smooth muscle tissue, PYK2 phosphorylation was increased and PYK2 inhibition decreased smooth muscle contraction. In vivo NVP-TAE684 treatment of wild-type mice reduced methacholine-induced airway smooth muscle contraction. These findings suggest that studies in the newborn CF pig may provide an important approach to enhance our understanding of airway smooth muscle biology and for discovery of novel airway smooth muscle therapeutics for CF and other diseases of airway hyperreactivity.
- Published
- 2017
- Full Text
- View/download PDF
29. Medical reversal of chronic sinusitis in a cystic fibrosis patient with ivacaftor
- Author
-
Mahmoud H. Abou Alaiwa, Xiao Xiao Tang, David A. Stoltz, Sarah E. Ernst, Janice L. Launspach, Viral Shah, Brieanna M Hilkin, Douglas B. Hornick, Philip H. Karp, Michael J. Welsh, Joseph Zabner, Scott M. Graham, and Eugene H. Chang
- Subjects
medicine.medical_specialty ,Pathology ,biology ,business.industry ,Treatment outcome ,Chronic sinusitis ,medicine.disease ,Gastroenterology ,Cystic fibrosis ,Cystic fibrosis transmembrane conductance regulator ,Ivacaftor ,Chronic disease ,Otorhinolaryngology ,Internal medicine ,otorhinolaryngologic diseases ,biology.protein ,Immunology and Allergy ,Medicine ,business ,Sinusitis ,medicine.drug - Abstract
Conclusion: This report documents the reversal of CF si- nus disease. Based on our in vivo and in vitro results, we speculate that ivacaor may reverse CF sinusitis by in- creasing ASL pH and decreasing ASL viscosity. These stud- ies suggest that CFTR modulation may be effective in treat- ing CF and perhaps non-CF sinusitis. C � 2014 ARS-AAOA, LLC.
- Published
- 2014
- Full Text
- View/download PDF
30. Assessing mucociliary transport of single particles in vivo shows variable speed and preference for the ventral trachea in newborn pigs
- Author
-
Ryan J. Adam, Geoffrey McLennan, Anthony J. Fischer, Michael J. Welsh, Omar A. Itani, Mark J. Hoegger, Alexander J. Tucker, David A. Stoltz, Eman Namati, Maged Awadalla, and Eric A. Hoffman
- Subjects
Chronic bronchitis ,Pathology ,medicine.medical_specialty ,Multidisciplinary ,Lung ,Swine ,Mucociliary clearance ,Cilium ,Biological Sciences ,Biology ,medicine.disease ,Cystic fibrosis ,Mucus ,Trachea ,Pathogenesis ,medicine.anatomical_structure ,Animals, Newborn ,Mucociliary Clearance ,In vivo ,medicine ,Animals - Abstract
Mucociliary transport (MCT) is an innate defense mechanism that removes particulates, noxious material, and microorganisms from the lung. Several airway diseases exhibit abnormal MCT, including asthma, chronic bronchitis, and cystic fibrosis. However, it remains uncertain whether MCT abnormalities contribute to the genesis of disease or whether they are secondary manifestations that may fuel disease progression. Limitations of current MCT assays and of current animal models of human disease have hindered progress in addressing these questions. Therefore, we developed an in vivo assay of MCT, and here we describe its use in newborn wild-type pigs. We studied pigs because they share many physiological, biochemical, and anatomical features with humans and can model several human diseases. We used X-ray multidetector-row-computed tomography to track movement of individual particles in the large airways of newborn pigs. Multidetector-row-computed tomography imaging provided high spatial and temporal resolution and registration of particle position to airway anatomy. We discovered that cilia orientation directs particles to the ventral tracheal surface. We also observed substantial heterogeneity in the rate of individual particle movement, and we speculate that variations in mucus properties may be responsible. The increased granularity of MCT data provided by this assay may provide an opportunity to better understand host defense mechanisms and the pathogenesis of airway disease.
- Published
- 2014
- Full Text
- View/download PDF
31. Air Trapping and Airflow Obstruction in Newborn Cystic Fibrosis Piglets
- Author
-
Amit Diwakar, David A. Stoltz, Eric A. Hoffman, Drake C. Bouzek, David K. Meyerholz, Mark J. Hoegger, Thomas J. Gross, Peter J. Taft, Maged Awadalla, Ryan J. Adam, Christian Bauer, Nicholas D. Gansemer, Mahmoud H. Abou Alaiwa, Reinhard Beichel, Andrew S. Michalski, Matthias Ochs, and Joseph M. Reinhardt
- Subjects
Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Swine ,Bronchi ,Critical Care and Intensive Care Medicine ,Air trapping ,Cystic fibrosis ,Airway resistance ,Multidetector Computed Tomography ,medicine ,Animals ,Lung volumes ,Bronchography ,business.industry ,Airway Resistance ,respiratory system ,Airway obstruction ,medicine.disease ,Mucus ,respiratory tract diseases ,Airway Obstruction ,Pulmonary Alveoli ,Trachea ,medicine.symptom ,Lung Volume Measurements ,business ,Airway - Abstract
Air trapping and airflow obstruction are being increasingly identified in infants with cystic fibrosis. These findings are commonly attributed to airway infection, inflammation, and mucus buildup.To learn if air trapping and airflow obstruction are present before the onset of airway infection and inflammation in cystic fibrosis.On the day they are born, piglets with cystic fibrosis lack airway infection and inflammation. Therefore, we used newborn wild-type piglets and piglets with cystic fibrosis to assess air trapping, airway size, and lung volume with inspiratory and expiratory X-ray computed tomography scans. Micro-computed tomography scanning was used to assess more distal airway sizes. Airway resistance was determined with a mechanical ventilator. Mean linear intercept and alveolar surface area were determined using stereologic methods.On the day they were born, piglets with cystic fibrosis exhibited air trapping more frequently than wild-type piglets (75% vs. 12.5%, respectively). Moreover, newborn piglets with cystic fibrosis had increased airway resistance that was accompanied by luminal size reduction in the trachea, mainstem bronchi, and proximal airways. In contrast, mean linear intercept length, alveolar surface area, and lung volume were similar between both genotypes.The presence of air trapping, airflow obstruction, and airway size reduction in newborn piglets with cystic fibrosis before the onset of airway infection, inflammation, and mucus accumulation indicates that cystic fibrosis impacts airway development. Our findings suggest that early airflow obstruction and air trapping in infants with cystic fibrosis might, in part, be caused by congenital airway abnormalities.
- Published
- 2013
- Full Text
- View/download PDF
32. Early Airway Structural Changes in Cystic Fibrosis Pigs as a Determinant of Particle Distribution and Deposition
- Author
-
Ryan J. Adam, Andrew S. Michalski, Maged Awadalla, Ching-Long Lin, David A. Stoltz, Karen J. Reynolds, Drake C. Bouzek, Eric A. Hoffman, Matthew K. Fuld, Shinjiro Miyawaki, and Mahmoud H. Abou Alaiwa
- Subjects
Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Swine ,Biomedical Engineering ,Cystic Fibrosis Transmembrane Conductance Regulator ,Biology ,Cystic fibrosis ,Article ,Animals, Genetically Modified ,Pathogenesis ,medicine ,Animals ,Distribution (pharmacology) ,Lung ,A determinant ,Anatomy ,respiratory system ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,medicine.anatomical_structure ,Animals, Newborn ,Hydrodynamics ,Breathing ,biology.protein ,Pulmonary Ventilation ,Tomography, X-Ray Computed ,Airway - Abstract
The pathogenesis of cystic fibrosis (CF) airway disease is not well understood. A porcine CF model was recently generated, and these animals develop lung disease similar to humans with CF. At birth, before infection and inflammation, CF pigs have airways that are irregularly shaped and have a reduced caliber compared to non-CF pigs. We hypothesized that these airway structural abnormalities affect airflow patterns and particle distribution. To test this hypothesis we used computational fluid dynamics (CFD) on airway geometries obtained by computed tomography of newborn non-CF and CF pigs. For the same flow rate, newborn CF pig airways exhibited higher air velocity and resistance compared to non-CF. Moreover we found that, at the carina bifurcation, particles greater than 5-μm preferably distributed to the right CF lung despite almost equal airflow ventilation in non-CF and CF. CFD modeling also predicted that deposition efficiency was greater in CF compared to non-CF for 5- and 10-μm particles. These differences were most significant in the airways included in the geometry supplying the right caudal, right accessory, left caudal, and left cranial lobes. The irregular particle distribution and increased deposition in newborn CF pig airways suggest that early airway structural abnormalities might contribute to CF disease pathogenesis.
- Published
- 2013
- Full Text
- View/download PDF
33. Tracheomalacia is associated with lower FEV1 and Pseudomonas acquisition in children with CF
- Author
-
Ryan J. Adam, Christopher F. Baranano, David A. Stoltz, Anthony J. Fischer, Paul B. McCray, Miles Weinberger, Simon C. Kao, Timothy D. Starner, and Sachinkumar B. Singh
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,education.field_of_study ,Bronchiectasis ,medicine.diagnostic_test ,business.industry ,Population ,Meconium Ileus ,Airway obstruction ,medicine.disease ,Gastroenterology ,Cystic fibrosis ,Obstructive lung disease ,Surgery ,Tracheomalacia ,Bronchoscopy ,Internal medicine ,Pediatrics, Perinatology and Child Health ,medicine ,business ,education - Abstract
Summary Background Tracheomalacia (TM) occurs in approximately 1 in 2,100 children. Because the trachea develops abnormally in animal models of cystic fibrosis (CF), we hypothesized this may also occur in children with CF, increasing their risk of TM. Purpose To examine the prevalence and clinical consequences of TM in children with CF. Methods We studied children with CF born between 1995 and 2012. TM was defined as dynamic collapse of the trachea, and the severity was recorded as described in the chart. The effect of TM on patient outcomes, including FEV1, CT changes, and acquisition of CF pathogens, was assessed using a longitudinal patient dataset. Results Eighty-nine percent of children with CF had at least one bronchoscopy (n = 97/109). Fifteen percent of these children had TM described in any bronchoscopy report (n = 15/97). Of the patients with TM, eight had meconium ileus (P = 0.003) and all were pancreatic insufficient. Pseudomonas aeruginosa infection occurred 1.3 years earlier among children with TM (P = 0.01). Starting FEV1 values by age 8 were diminished by over 18% of predicted for patients with TM. Life-threatening episodes of airway obstruction occurred in 3 of 15 patients with CF and TM, including one leading to death. Gender, prematurity, and hepatic disease were not associated with TM. No difference was observed in the frequency of bronchiectasis. Conclusions TM is significantly more common in infants and children with CF than in the general population and is associated with airway obstruction and earlier Pseudomonas acquisition. Pediatr Pulmonol. 2014; 49:960–970. © 2013 Wiley Periodicals, Inc.
- Published
- 2013
- Full Text
- View/download PDF
34. Intestinal CFTR expression alleviates meconium ileus in cystic fibrosis pigs
- Author
-
David K. Meyerholz, Joseph Zabner, Paula S. Ludwig, Jeng-Haur Chen, Leah R. Reznikov, Peter J. Taft, Tanner J. Wallen, Philip H. Karp, Paul B. McCray, Sarah E. Ernst, Nicholas D. Gansemer, Michael V. Rector, George A. Nelson, Mahmoud M.Abou Alaiwa, Emma E. Hornick, Randall S. Prather, Eugene H. Chang, Ryan J. Adam, Alejandro A. Pezzulo, David A. Stoltz, Drake C. Bouzek, Lynda S. Ostedgaard, Michael J. Welsh, Tatiana Rokhlina, Melissa Samuel, James D. McMenimen, Katrina L. Bogan, Mark J. Hoegger, Christine L. Wohlford-Lenane, and Eric A. Hoffman
- Subjects
Male ,Meconium ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Cystic Fibrosis ,Biliary cirrhosis ,Sus scrofa ,Cystic Fibrosis Transmembrane Conductance Regulator ,Gene Expression ,Meconium Ileus ,Fatty Acid-Binding Proteins ,Cystic fibrosis ,Gastroenterology ,Animals, Genetically Modified ,Liver disease ,Ileus ,Ileum ,Internal medicine ,Gene expression ,medicine ,Animals ,Humans ,Promoter Regions, Genetic ,Lung ,Pancreas ,business.industry ,Infant, Newborn ,Vas deferens ,General Medicine ,respiratory system ,medicine.disease ,Penetrance ,digestive system diseases ,Pathophysiology ,Rats ,Radiography ,Trachea ,Disease Models, Animal ,Phenotype ,medicine.anatomical_structure ,Animals, Newborn ,Technical Advance ,Female ,business - 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.
- Published
- 2013
- Full Text
- View/download PDF
35. Delayed neutrophil apoptosis enhances NET formation in cystic fibrosis
- Author
-
Brian N McCullagh, Donald J. Davidson, David A. Stoltz, Jennifer M Felton, Robert D. Gray, David A. Dorward, Christopher D. Lucas, Paul B. McCray, Edward F. McKone, Pradeep K Singh, Christopher Haslett, Seamas C. Donnelly, Rodger Duffin, Lily Paemka, Maeve P. Smith, Gordon Cooke, Moira K. B. Whyte, Adriano G. Rossi, Annie Mackellar, Calum T. Robb, Gareth Hardisty, and Kate H. Regan
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Adult ,Time Factors ,Cystic Fibrosis ,Cell Survival ,Neutrophils ,Swine ,medicine.medical_treatment ,Inflammation ,Apoptosis ,Cystic fibrosis ,Extracellular Traps ,Proinflammatory cytokine ,Ivacaftor ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,biology ,business.industry ,Neutrophil extracellular traps ,medicine.disease ,Neutrophil Biology ,Cystic fibrosis transmembrane conductance regulator ,3. Good health ,030104 developmental biology ,Cytokine ,030220 oncology & carcinogenesis ,Case-Control Studies ,Immunology ,biology.protein ,medicine.symptom ,business ,medicine.drug - Abstract
BackgroundCystic fibrosis (CF) lung disease is defined by large numbers of neutrophils and associated damaging products in the airway. Delayed neutrophil apoptosis is described in CF although it is unclear whether this is a primary neutrophil defect or a response to chronic inflammation. Increased levels of neutrophil extracellular traps (NETs) have been measured in CF and we aimed to investigate the causal relationship between these phenomena and their potential to serve as a driver of inflammation. We hypothesised that the delay in apoptosis in CF is a primary defect and preferentially allows CF neutrophils to form NETs, contributing to inflammation.MethodsBlood neutrophils were isolated from patients with CF, CF pigs and appropriate controls. Neutrophils were also obtained from patients with CF before and after commencing ivacaftor. Apoptosis was assessed by morphology and flow cytometry. NET formation was determined by fluorescent microscopy and DNA release assays. NET interaction with macrophages was examined by measuring cytokine generation with ELISA and qRT-PCR.ResultsCF neutrophils live longer due to decreased apoptosis. This was observed in both cystic fibrosis transmembrane conductance regulator (CFTR) null piglets and patients with CF, and furthermore was reversed by ivacaftor (CFTR potentiator) in patients with gating (G551D) mutations. CF neutrophils formed more NETs and this was reversed by cyclin-dependent kinase inhibitor exposure. NETs provided a proinflammatory stimulus to macrophages, which was enhanced in CF.ConclusionsCF neutrophils have a prosurvival phenotype that is associated with an absence of CFTR function and allows increased NET production, which can in turn induce inflammation. Augmenting neutrophil apoptosis in CF may allow more appropriate neutrophil disposal, decreasing NET formation and thus inflammation.
- Published
- 2017
- Full Text
- View/download PDF
36. Lentiviral-mediated phenotypic correction of cystic fibrosis pigs
- Author
-
Viral Shah, Ashley L. Cooney, Mallory R. Stroik, Mahmoud H. Abou Alaiwa, Patrick L. Sinn, Michael J. Welsh, David K. Meyerholz, Linda S. Powers, David A. Stoltz, Nicholas D. Gansemer, Drake C. Bouzek, and Paul B. McCray
- Subjects
0301 basic medicine ,Feline immunodeficiency virus ,Cystic Fibrosis ,Swine ,Genetic enhancement ,Genetic Vectors ,Cystic Fibrosis Transmembrane Conductance Regulator ,Cystic fibrosis ,Viral vector ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,medicine ,Animals ,Bronchus ,Ion Transport ,Lung ,biology ,Lentivirus ,Genetic Therapy ,General Medicine ,respiratory system ,medicine.disease ,biology.organism_classification ,Molecular biology ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Research Article - Abstract
Cystic Fibrosis (CF) is an autosomal recessive disease caused by mutations in CF transmembrane conductance regulator (CFTR), resulting in defective anion transport. Regardless of the disease-causing mutation, gene therapy is a strategy to restore anion transport to airway epithelia. Indeed, viral vector–delivered CFTR can complement the anion channel defect. In this proof-of-principle study, functional in vivo CFTR channel activity was restored in the airways of CF pigs using a feline immunodeficiency virus–based (FIV-based) lentiviral vector pseudotyped with the GP64 envelope. Three newborn CF pigs received aerosolized FIV-CFTR to the nose and lung. Two weeks after viral vector delivery, epithelial tissues were analyzed for functional correction. In freshly excised tracheal and bronchus tissues and cultured ethmoid sinus cells, we observed a significant increase in transepithelial cAMP-stimulated current, evidence of functional CFTR. In addition, we observed increases in tracheal airway surface liquid pH and bacterial killing in CFTR vector–treated animals. Together, these data provide the first evidence to our knowledge that lentiviral delivery of CFTR can partially correct the anion channel defect in a large-animal CF model and validate a translational strategy to treat or prevent CF lung disease.
- Published
- 2016
- Full Text
- View/download PDF
37. CFTR gene transfer with AAV improves early cystic fibrosis pig phenotypes
- Author
-
David A. Stoltz, David D. Dickey, Linda S. Powers, Michael J. Welsh, John R. Weinstein, John F. Engelhardt, Ziying Yan, Mahmoud H. Abou Alaiwa, Lynda S. Ostedgaard, Jamie M. Bergen, Xiaopeng Li, Katherine J. D. A. Excoffon, Drake C. Bouzek, Viral Shah, Nicholas D. Gansemer, Benjamin R. Steines, Patrick L. Sinn, David V. Schaffer, and Joseph Zabner
- Subjects
0301 basic medicine ,viruses ,Genetic enhancement ,Point mutation ,General Medicine ,respiratory system ,Biology ,Vectors in gene therapy ,medicine.disease ,Cystic fibrosis ,Virology ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,3. Good health ,Cell biology ,03 medical and health sciences ,Transduction (genetics) ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Gene ,Tropism ,Research Article - 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.
- Published
- 2016
- Full Text
- View/download PDF
38. Repurposing tromethamine as inhaled therapy to treat CF airway disease
- Author
-
Peter S. Thorne, Kelsey A. Sheets, Mahmoud H. Abou Alaiwa, Michael J. Welsh, Joseph Zabner, Peter J. Taft, David A. Stoltz, Janice L. Launspach, Jade A. Rivera, and Nicholas D. Gansemer
- Subjects
0301 basic medicine ,Chemistry ,Tromethamine ,Metabolic acidosis ,General Medicine ,respiratory system ,Anion channel activity ,Pharmacology ,medicine.disease ,Cystic fibrosis ,Hypertonic saline ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030228 respiratory system ,In vivo ,Anesthesia ,medicine ,Sputum ,medicine.symptom ,Airway ,Research Article - Abstract
In cystic fibrosis (CF), loss of CF transmembrane conductance regulator (CFTR) anion channel activity causes airway surface liquid (ASL) pH to become acidic, which impairs airway host defenses. One potential therapeutic approach is to correct the acidic pH in CF airways by aerosolizing HCO3– and/or nonbicarbonate pH buffers. Here, we show that raising ASL pH with inhaled HCO3– increased pH. However, the effect was transient, and pH returned to baseline values within 30 minutes. Tromethamine (Tham) is a buffer with a long serum half-life used as an i.v. formulation to treat metabolic acidosis. We found that Tham aerosols increased ASL pH in vivo for at least 2 hours and enhanced bacterial killing. Inhaled hypertonic saline (7% NaCl) is delivered to people with CF in an attempt to promote mucus clearance. Because an increased ionic strength inhibits ASL antimicrobial factors, we added Tham to hypertonic saline and applied it to CF sputum. We found that Tham alone and in combination with hypertonic saline increased pH and enhanced bacterial killing. These findings suggest that aerosolizing the HCO3–-independent buffer Tham, either alone or in combination with hypertonic saline, might be of therapeutic benefit in CF airway disease.
- Published
- 2016
- Full Text
- View/download PDF
39. Acute administration of ivacaftor to people with cystic fibrosis and a G551D-CFTR mutation reveals smooth muscle abnormalities
- Author
-
Joseph E. Cavanaugh, Pradeep K. Singh, Michael J. Welsh, Jered Sieren, B. Grogan, Eric A. Hoffman, David A. Stoltz, Edward F. McKone, Katherine B. Hisert, Thomas J. Gross, Ryan J. Adam, Jonathan D. Dodd, Janel K. Barnes, Janice L. Launspach, Charles G. Gallagher, and Anthony J. Fischer
- Subjects
Spirometry ,medicine.medical_specialty ,Pathology ,congenital, hereditary, and neonatal diseases and abnormalities ,030204 cardiovascular system & hematology ,Air trapping ,Cystic fibrosis ,Ivacaftor ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Pulse wave velocity ,Lung ,medicine.diagnostic_test ,business.industry ,General Medicine ,respiratory system ,medicine.disease ,respiratory tract diseases ,medicine.anatomical_structure ,030228 respiratory system ,Cardiology ,medicine.symptom ,Clinical Medicine ,Airway ,business ,medicine.drug - Abstract
BACKGROUND. Airflow obstruction is common in cystic fibrosis (CF), yet the underlying pathogenesis remains incompletely understood. People with CF often exhibit airway hyperresponsiveness, CF transmembrane conductance regulator (CFTR) is present in airway smooth muscle (ASM), and ASM from newborn CF pigs has increased contractile tone, suggesting that loss of CFTR causes a primary defect in ASM function. We hypothesized that restoring CFTR activity would decrease smooth muscle tone in people with CF. METHODS. To increase or potentiate CFTR function, we administered ivacaftor to 12 adults with CF with the G551D-CFTR mutation; ivacaftor stimulates G551D-CFTR function. We studied people before and immediately after initiation of ivacaftor (48 hours) to minimize secondary consequences of CFTR restoration. We tested smooth muscle function by investigating spirometry, airway distensibility, and vascular tone. RESULTS. Ivacaftor rapidly restored CFTR function, indicated by reduced sweat chloride concentration. Airflow obstruction and air trapping also improved. Airway distensibility increased in airways less than 4.5 mm but not in larger-sized airways. To assess smooth muscle function in a tissue outside the lung, we measured vascular pulse wave velocity (PWV) and augmentation index, which both decreased following CFTR potentiation. Finally, change in distensibility of
- Published
- 2016
40. Computer-aided analysis of airway trees in micro-CT scans of ex vivo porcine lung tissue
- Author
-
David A. Stoltz, Christian Bauer, Ryan J. Adam, and Reinhard Beichel
- Subjects
Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Swine ,Bronchi ,Health Informatics ,In Vitro Techniques ,Sensitivity and Specificity ,Cystic fibrosis ,Article ,Pattern Recognition, Automated ,Porcine lung ,Artificial Intelligence ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Micro ct ,Lung ,Radiological and Ultrasound Technology ,Bronchography ,business.industry ,Reproducibility of Results ,Anatomy ,respiratory system ,medicine.disease ,Computer Graphics and Computer-Aided Design ,respiratory tract diseases ,Radiographic Image Enhancement ,medicine.anatomical_structure ,Lung disease ,Radiographic Image Interpretation, Computer-Assisted ,Computer Vision and Pattern Recognition ,Tomography, X-Ray Computed ,business ,Airway ,Algorithms ,Ex vivo - Abstract
We present a highly automated approach to obtain detailed structural models of airway trees from ex vivo porcine lung tissue imaged with a high resolution micro-CT scanner. Such information is an important prerequisite to systematically study models of lung disease that affect airway morphology. The method initially identifies all tubular airway-like structures in the lung. In a second processing step, these structures are grouped into a connected airway tree by utilizing prior knowledge about the airway trees branching pattern. The method was evaluated on 12 micro-CT scans from four tracheal lobes of piglets imaged at three different inflation levels. For this study, two control piglets and two cystic fibrosis piglets were used. For systematic validation of our approach, an airway nomenclature was developed for the pig airway tree. Out of more than 3500 airway tree segments assessed during evaluation, 88.45% were correctly identified by the method. No false positive airway branches were found. A detailed performance analysis for different airway tree hierarchy levels, lung inflation levels and piglets with/without cystic fibrosis is presented in the paper.
- Published
- 2012
- Full Text
- View/download PDF
41. Pancreatic and biliary secretion are both altered in cystic fibrosis pigs
- Author
-
Maisam Abu-El-Haija, Peter J. Taft, Aliye Uc, Alejandro A. Pezzulo, Michelle Griffin, Marwa Abu-El-Haija, David K. Meyerholz, Lynda S. Ostedgaard, Michael J. Welsh, Paula S. Ludwig, Radhamma L. Giriyappa, Xiaoxiao Tang, and David A. Stoltz
- Subjects
medicine.medical_specialty ,Cystic Fibrosis ,Swine ,Physiology ,Cystic Fibrosis Transmembrane Conductance Regulator ,Cystic fibrosis ,Gastroenterology ,Secretin ,Pancreatic Juice ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Trypsin ,Amylase ,Pancreas ,Hepatology ,biology ,Gallbladder ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,Disease Models, Animal ,Endocrinology ,medicine.anatomical_structure ,Amylases ,Pancreatic juice ,biology.protein ,Bile Ducts ,medicine.drug - 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.
- Published
- 2012
- Full Text
- View/download PDF
42. Pancreatic Damage in Fetal and Newborn Cystic Fibrosis Pigs Involves the Activation of Inflammatory and Remodeling Pathways
- Author
-
Michael J. Welsh, David K. Meyerholz, Maisam Abu-El-Haija, Marwa Abu-El-Haija, David A. Stoltz, Shyam Ramachandran, Aliye Uc, Radhamma L. Giriyappa, Michelle Griffin, and Paul B. McCray
- Subjects
Pathology ,medicine.medical_specialty ,Pancreatic disease ,Cystic Fibrosis ,Sus scrofa ,Cystic Fibrosis Transmembrane Conductance Regulator ,Apoptosis ,Inflammation ,Cystic fibrosis ,Pathology and Forensic Medicine ,Proinflammatory cytokine ,Pathogenesis ,03 medical and health sciences ,Fetus ,0302 clinical medicine ,Pregnancy ,Internal medicine ,medicine ,Animals ,Pancreas ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,biology ,Gene Expression Profiling ,Regular Article ,Complement System Proteins ,medicine.disease ,Actins ,Cystic fibrosis transmembrane conductance regulator ,3. Good health ,medicine.anatomical_structure ,Endocrinology ,Animals, Newborn ,030220 oncology & carcinogenesis ,Disease Progression ,biology.protein ,Female ,medicine.symptom ,Transcriptome ,Signal Transduction - Abstract
Pancreatic disease has onset in utero in humans with cystic fibrosis (CF), and progresses over time to complete destruction of the organ. The exact mechanisms leading to pancreatic damage in CF are incompletely understood. Inflammatory cells are present in the pancreas of newborn pigs with CF (CF pigs) and humans, which suggests that inflammation may have a role in the destructive process. We wondered whether tissue inflammation and genes associated with inflammatory pathways were increased in the pancreas of fetal CF pigs [83 to 90 days gestation (normal pig gestation is ∼114 days)] and newborn pigs. Compared with fetal pigs without CF (non-CF pigs), in fetal CF pigs, the pancreas exhibited patchy inflammation and acinar atrophy, with progression in distribution and severity in neonatal CF pigs. Large-scale transcript profiling revealed that the pancreas in fetal and newborn CF pigs exhibited significantly increased expression of proinflammatory, complement cascade, and profibrotic genes when compared with fetal and newborn non-CF pigs. Acinar cells exhibited increased apoptosis in the pancreas of fetal and newborn CF pigs. α-Smooth muscle actin and transforming growth factor β1 were increased in both fetal and newborn CF pig pancreas, suggesting activation of profibrotic pathways. Cell proliferation and mucous cell metaplasia were detected in newborn, but not fetal, CF pigs, indicating that they were not an initiator of pathogenesis but a response. Proinflammatory, complement cascade, proapoptotic, and profibrotic pathways are activated in CF pig pancreas, and likely contribute to the destructive process.
- Published
- 2012
- Full Text
- View/download PDF
43. CFTR is required for maximal transepithelial liquid transport in pig alveolar epithelia
- Author
-
Peter J. Taft, Michael V. Rector, Xiaopeng Li, Alejandro A. Pezzulo, Thomas O. Moninger, Nathan D. Rossen, Michael J. Welsh, David A. Stoltz, Alejandro P. Comellas, Philip H. Karp, Paul B. McCray, Nicholas D. Gansemer, Sarah E. Ernst, and Joseph Zabner
- Subjects
Male ,Pulmonary and Respiratory Medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Physiology ,Cystic Fibrosis Transmembrane Conductance Regulator ,Stimulation ,Biology ,Cystic fibrosis ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Secretion ,Ion transporter ,Lung ,Tight junction ,Articles ,Cell Biology ,respiratory system ,medicine.disease ,digestive system diseases ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,Cell biology ,medicine.anatomical_structure ,Endocrinology ,Alveolar Epithelial Cells ,Extravascular Lung Water ,biology.protein ,Female ,Ex vivo - Abstract
A balance between alveolar liquid absorption and secretion is critical for maintaining optimal alveolar subphase liquid height and facilitating gas exchange in the alveolar space. However, the role of cystic fibrosis transmembrane regulator protein (CFTR) in this homeostatic process has remained elusive. Using a newly developed porcine model of cystic fibrosis, in which CFTR is absent, we investigated ion transport properties and alveolar liquid transport in isolated type II alveolar epithelial cells (T2AECs) cultured at the air-liquid interface. CFTR was distributed exclusively to the apical surface of cultured T2AECs. Alveolar epithelia from CFTR−/− pigs failed to increase liquid absorption in response to agents that increase cAMP, whereas cAMP-stimulated liquid absorption in CFTR+/− epithelia was similar to that in CFTR+/+ epithelia. Expression of recombinant CFTR restored stimulated liquid absorption in CFTR−/− T2AECs but had no effect on CFTR+/+ epithelia. In ex vivo studies of nonperfused lungs, stimulated liquid absorption was defective in CFTR−/− alveolar epithelia but similar between CFTR+/+ and CFTR+/− epithelia. When epithelia were studied at the air-liquid interface, elevating cAMP levels increased subphase liquid height in CFTR+/+ but not in CFTR−/− T2AECs. Our findings demonstrate that CFTR is required for maximal liquid absorption under cAMP stimulation, but it is not the rate-limiting factor. Furthermore, our data define a role for CFTR in liquid secretion by T2AECs. These insights may help to develop new treatment strategies for pulmonary edema and respiratory distress syndrome, diseases in which lung liquid transport is disrupted.
- Published
- 2012
- Full Text
- View/download PDF
44. Sinus hypoplasia precedes sinus infection in a porcine model of cystic fibrosis
- Author
-
Andrea E. Potash, Thomas O. Moninger, Michael J. Welsh, Sarah E. Ernst, David A. Stoltz, David K. Meyerholz, Jessica C. Sieren, Paul B. McCray, Philip H. Karp, Leah R. Reznikov, Nicholas D. Gansemer, Joseph Zabner, Alejandro A. Pezzulo, Eugene H. Chang, and Tanner J. Wallen
- Subjects
Male ,Aging ,Pathology ,Cystic Fibrosis ,Swine ,Organogenesis ,Cystic Fibrosis Transmembrane Conductance Regulator ,Cystic fibrosis ,Pathogenesis ,Random Allocation ,0302 clinical medicine ,Reference Values ,Paranasal Sinuses ,Medicine ,030223 otorhinolaryngology ,Sinusitis ,Sinus (anatomy) ,0303 health sciences ,biology ,Incidence ,Biopsy, Needle ,Gene Expression Regulation, Developmental ,Immunohistochemistry ,Hypoplasia ,Cystic fibrosis transmembrane conductance regulator ,3. Good health ,medicine.anatomical_structure ,Female ,medicine.medical_specialty ,Risk Assessment ,03 medical and health sciences ,Genetic model ,otorhinolaryngologic diseases ,Animals ,Genetic Predisposition to Disease ,Allergy/Rhinology ,030304 developmental biology ,business.industry ,animal model ,medicine.disease ,Sinus development ,Disease Models, Animal ,Nasal Mucosa ,Paranasal sinuses ,Animals, Newborn ,Otorhinolaryngology ,Chronic Disease ,Immunology ,biology.protein ,sinus disease ,business - Abstract
Objectives/Hypothesis: Chronic sinusitis is nearly universal in humans with cystic fibrosis (CF) and is accompanied by sinus hypoplasia (small sinuses). However, whether impaired sinus development is a primary feature of loss of the cystic fibrosis transmembrane conductance regulator (CFTR) or a secondary consequence of chronic infection remains unknown. Our objective was to study the early pathogenesis of sinus disease in CF. Study Design: Animal/basic science research. Methods: Sinus development was studied in a porcine CF model. Results: Porcine sinus epithelia expressed CFTR and exhibited transepithelial anion transport. Disruption of the CFTR gene eliminated both. Sinuses of newborn CF pigs were not infected and showed no evidence of inflammation, yet were hypoplastic at birth. Older CF pigs spontaneously developed sinus disease similar to that seen in humans with CF. Conclusions: These results define a role for CFTR in sinus development and suggest the potential of the CF pig as a genetic model of CF-sinus disease in which to test therapeutic strategies to minimize sinus-related CF morbidity.
- Published
- 2012
- Full Text
- View/download PDF
45. Concentration of the antibacterial precursor thiocyanate in cystic fibrosis airway secretions
- Author
-
Botond Banfi, David A. Stoltz, Joseph Zabner, Lakshmi Durairaj, Gideon K. D. Zamba, Alejandro A. Pezzulo, Michael J. Welsh, Yoko Nakano, Janice L. Launspach, Daniel Lorentzen, William M. Nauseef, and Paul B. McCray
- Subjects
Bodily Secretions ,Staphylococcus aureus ,endocrine system ,medicine.medical_specialty ,Cystic Fibrosis ,Swine ,Colony Count, Microbial ,Cystic Fibrosis Transmembrane Conductance Regulator ,Gene Expression ,Bronchi ,Microbial Sensitivity Tests ,Biochemistry ,Cystic fibrosis ,Article ,Animals, Genetically Modified ,Pathogenesis ,chemistry.chemical_compound ,Physiology (medical) ,Internal medicine ,Gene expression ,medicine ,Animals ,Humans ,Secretion ,Cells, Cultured ,Thiocyanate ,Homozygote ,Lactoperoxidase ,Epithelial Cells ,Hypothiocyanite ,respiratory system ,medicine.disease ,Anti-Bacterial Agents ,respiratory tract diseases ,Trachea ,Endocrinology ,Animals, Newborn ,nervous system ,chemistry ,sense organs ,Nasal Cavity ,Airway ,Oxidation-Reduction ,Thiocyanates - Abstract
A recently discovered enzyme system produces antibacterial hypothiocyanite (OSCN(-)) in the airway lumen by oxidizing the secreted precursor thiocyanate (SCN(-)). Airway epithelial cultures have been shown to secrete SCN(-) in a CFTR-dependent manner. Thus, reduced SCN(-) availability in the airway might contribute to the pathogenesis of cystic fibrosis (CF), a disease caused by mutations in the CFTR gene and characterized by an airway host defense defect. We tested this hypothesis by analyzing the SCN(-) concentration in the nasal airway surface liquid (ASL) of CF patients and non-CF subjects and in the tracheobronchial ASL of CFTR-ΔF508 homozygous pigs and control littermates. In the nasal ASL, the SCN(-) concentration was ~30-fold higher than in serum independent of the CFTR mutation status of the human subject. In the tracheobronchial ASL of CF pigs, the SCN(-) concentration was somewhat reduced. Among human subjects, SCN(-) concentrations in the ASL varied from person to person independent of CFTR expression, and CF patients with high SCN(-) levels had better lung function than those with low SCN(-) levels. Thus, although CFTR can contribute to SCN(-) transport, it is not indispensable for the high SCN(-) concentration in ASL. The correlation between lung function and SCN(-) concentration in CF patients may reflect a beneficial role for SCN(-).
- Published
- 2011
- Full Text
- View/download PDF
46. Loss of Anion Transport without Increased Sodium Absorption Characterizes Newborn Porcine Cystic Fibrosis Airway Epithelia
- Author
-
David A. Stoltz, Michael V. Rector, Kathryn Chaloner, Philip H. Karp, Leah R. Reznikov, Jeng-Haur Chen, Alejandro A. Pezzulo, Sarah E. Ernst, Janice L. Launspach, Thomas O. Moninger, Michael J. Welsh, and Joseph Zabner
- Subjects
Anions ,medicine.medical_specialty ,Cystic Fibrosis ,Respiratory System ,Sus scrofa ,HUMDISEASE ,Cystic Fibrosis Transmembrane Conductance Regulator ,Absorption (skin) ,Biology ,Cystic fibrosis ,General Biochemistry, Genetics and Molecular Biology ,Epithelium ,Article ,Internal medicine ,medicine ,Animals ,Humans ,Respiratory system ,Transcellular ,Ion transporter ,Ion Transport ,Biochemistry, Genetics and Molecular Biology(all) ,respiratory system ,medicine.disease ,Molecular biology ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,Endocrinology ,medicine.anatomical_structure ,Animals, Newborn ,Permeability (electromagnetism) ,biology.protein - Abstract
Defective transepithelial electrolyte transport is thought to initiate cystic fibrosis (CF) lung disease. Yet, how loss of CFTR affects electrolyte transport remains uncertain. CFTR -/- pigs spontaneously develop lung disease resembling human CF. At birth, their airways exhibit a bacterial host defense defect, but are not inflamed. Therefore, we studied ion transport in newborn nasal and tracheal/bronchial epithelia in tissues, cultures, and in vivo. CFTR -/- epithelia showed markedly reduced Cl - and HCO 3 - transport. However, in contrast to a widely held view, lack of CFTR did not increase transepithelial Na + or liquid absorption or reduce periciliary liquid depth. Like human CF, CFTR -/- pigs showed increased amiloride-sensitive voltage and current, but lack of apical Cl - conductance caused the change, not increased Na + transport. These results indicate that CFTR provides the predominant transcellular pathway for Cl - and HCO 3 - in porcine airway epithelia, and reduced anion permeability may initiate CF airway disease. © 2010 Elsevier Inc., published_or_final_version
- Published
- 2010
- Full Text
- View/download PDF
47. Loss of Cystic Fibrosis Transmembrane Conductance Regulator Function Produces Abnormalities in Tracheal Development in Neonatal Pigs and Young Children
- Author
-
Amanda R. Smith, Melissa J. Suter, Shyam Ramachandran, Eman Namati, David A. Stoltz, Guillermo J. Tearney, Alejandro A. Pezzulo, Geoffrey McLennan, Paul B. McCray, Simon C. Kao, Michael V. Rector, Michael J. Welsh, David K. Meyerholz, and Joseph Zabner
- Subjects
Pulmonary and Respiratory Medicine ,Respiratory Mucosa ,Pathology ,medicine.medical_specialty ,Pancreatic disease ,Cystic Fibrosis ,Swine ,Cystic Fibrosis Transmembrane Conductance Regulator ,Inflammation ,Critical Care and Intensive Care Medicine ,Cystic fibrosis ,Intensive care ,medicine ,Animals ,Humans ,C. Cystic Fibrosis ,Retrospective Studies ,biology ,business.industry ,Respiratory disease ,Infant ,Muscle, Smooth ,Anatomy ,respiratory system ,Microarray Analysis ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,Trachea ,Animals, Newborn ,Child, Preschool ,biology.protein ,Trachealis muscle ,medicine.symptom ,Tomography, X-Ray Computed ,business - Abstract
Although airway abnormalities are common in patients with cystic fibrosis (CF), it is unknown whether they are all secondary to postnatal infection and inflammation, which characterize the disease.To learn whether loss of the cystic fibrosis transmembrane conductance regulator (CFTR) might affect major airways early in life, before the onset of inflammation and infection.We studied newborn CFTR⁻(/)⁻ pig trachea, using computed tomography (CT) scans, pathology, and morphometry. We retrospectively analyzed trachea CT scans in young children with CF and also previously published data of infants with CF.We discovered three abnormalities in the porcine CF trachea. First, the trachea and mainstem bronchi had a uniformly small caliber and cross-sections of trachea were less circular than in controls. Second, trachealis smooth muscle had an altered bundle orientation and increased transcripts in a smooth muscle gene set. Third, submucosal gland units occurred with similar frequency in the mucosa of CF and control airways, but CF submucosal glands were hypoplastic and had global reductions in tissue-specific transcripts. To learn whether any of these changes occurred in young patients with CF, we examined CT scans from children 2 years of age and younger, and found that CF tracheas were less circular in cross-section, but lacked differences in lumen area. However, analysis of previously published morphometric data showed reduced tracheal lumen area in neonates with CF.Our findings in newborn CF pigs and young patients with CF suggest that airway changes begin during fetal life and may contribute to CF pathogenesis and clinical disease during postnatal life.
- Published
- 2010
- Full Text
- View/download PDF
48. Pigs and humans with cystic fibrosis have reduced insulin-like growth factor 1 (IGF1) levels at birth
- Author
-
Alejandro A. Pezzulo, Paul B. McCray, David A. Stoltz, Nicholas D. Gansemer, Michael J. Welsh, Joseph Zabner, Mark P. Rogan, Douglas C. Fredericks, Melissa Samuel, Randall S. Prather, and Leah R. Reznikov
- Subjects
endocrine system ,medicine.medical_specialty ,Cystic Fibrosis ,Bone density ,medicine.medical_treatment ,Sus scrofa ,Cystic Fibrosis Transmembrane Conductance Regulator ,Inflammation ,Biology ,Cystic fibrosis ,Gene Knockout Techniques ,Insulin-like growth factor ,Bone Density ,Internal medicine ,medicine ,Animals ,Humans ,Body Weights and Measures ,Insulin-Like Growth Factor I ,DNA Primers ,Analysis of Variance ,Multidisciplinary ,Reverse Transcriptase Polymerase Chain Reaction ,Growth factor ,Infant, Newborn ,Biological Sciences ,Humerus ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,Endocrinology ,Animals, Newborn ,In utero ,Growth Hormone ,Pituitary Gland ,biology.protein ,Biomarker (medicine) ,medicine.symptom ,Biomarkers ,hormones, hormone substitutes, and hormone antagonists - Abstract
People with cystic fibrosis (CF) exhibit growth defects. That observation has been attributed, in part, to decreased insulin-like growth factor 1 (IGF1) levels, and the reduction has been blamed on malnutrition and pulmonary inflammation. However, patients with CF already have a reduced weight at birth, a manifestation not likely secondary to poor nutrition or inflammation. We found that, like humans, CF pigs were smaller than non-CF littermates and had lower IGF1 levels. To better understand the basis of IGF1 reduction, we studied newborn pigs and found low IGF1 levels within 12 h of birth. Moreover, humerus length and bone mineral content were decreased, consistent with less IGF1 activity in utero. These findings led us to test newborn humans with CF, and we found that they also had reduced IGF1 levels. Discovering lower IGF1 levels in newborn pigs and humans indicates that the decrease is not solely a consequence of malnutrition or pulmonary inflammation and that loss of cystic fibrosis transmembrane conductance regulator function has a more direct effect. Consistent with this hypothesis, we discovered reduced growth hormone release in organotypic pituitary slice cultures of newborn CF pigs. These findings may explain the long-standing observation that CF newborns are smaller than non-CF babies and why some patients with good clinical status fail to reach their growth potential. The results also suggest that measuring IGF1 levels might be of value as a biomarker to predict disease severity or the response to therapeutics. Finally, they raise the possibility that IGF1 supplementation beginning in infancy might be beneficial in CF.
- Published
- 2010
- Full Text
- View/download PDF
49. Disruption of the CFTR Gene Produces a Model of Cystic Fibrosis in Newborn Pigs
- Author
-
David K. Meyerholz, Peter J. Taft, Christine L. Wohlford-Lenane, Joseph Zabner, Aliye Uc, Melissa Samuel, David Wax, Kim A. Brogden, Omar A. Itani, Lynda S. Ostedgaard, Mark P. Rogan, Clifton N. Murphy, Randall S. Prather, Paul B. McCray, Amanda C. Kabel, Michael J. Welsh, August Rieke, Timothy D. Starner, Philip H. Karp, Kristin M. Whitworth, Robert A. Hanfland, Alejandro A. Pezzulo, David A. Stoltz, Tony L. Smith, Greg J. Davis, Tatiana Rokhlina, Joel Shilyansky, and Christopher S. Rogers
- Subjects
Male ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Pancreatic disease ,Cystic Fibrosis ,Swine ,Biliary cirrhosis ,Cystic Fibrosis Transmembrane Conductance Regulator ,Meconium Ileus ,Biology ,Cystic fibrosis ,Article ,Pathogenesis ,Ileus ,Chlorides ,Internal medicine ,medicine ,Animals ,Allele ,Lung ,Recombination, Genetic ,Ion Transport ,Multidisciplinary ,Liver Cirrhosis, Biliary ,Gallbladder ,respiratory system ,medicine.disease ,Pancreas, Exocrine ,Intestines ,Disease Models, Animal ,Endocrinology ,medicine.anatomical_structure ,Animals, Newborn ,Liver ,Immunology ,Female ,Pancreas - Abstract
Almost two decades after CFTR was identified as the gene responsible for cystic fibrosis (CF), we still lack answers to many questions about the pathogenesis of the disease, and it remains incurable. Mice with a disrupted CFTR gene have greatly facilitated CF studies, but the mutant mice do not develop the characteristic manifestations of human CF, including abnormalities of the pancreas, lung, intestine, liver, and other organs. Because pigs share many anatomical and physiological features with humans, we generated pigs with a targeted disruption of both CFTR alleles. Newborn pigs lacking CFTR exhibited defective chloride transport and developed meconium ileus, exocrine pancreatic destruction, and focal biliary cirrhosis, replicating abnormalities seen in newborn humans with CF. The pig model may provide opportunities to address persistent questions about CF pathogenesis and accelerate discovery of strategies for prevention and treatment.
- Published
- 2008
- Full Text
- View/download PDF
50. Production of CFTR-null and CFTR-ΔF508 heterozygous pigs by adeno-associated virus–mediated gene targeting and somatic cell nuclear transfer
- Author
-
Melissa Samuel, August Rieke, David Wax, Yanhong Hao, Yuhong Li, Daniel W. Vermeer, Scott W. Korte, Ziying Yan, Lee D. Spate, Kristin M. Whitworth, Michael J. Welsh, David A. Stoltz, John F. Engelhardt, Michael L. Linville, Tatiana Rokhlina, Clifton N. Murphy, Amanda C. Kabel, Randall S. Prather, Christopher S. Rogers, and Elena Petroff
- Subjects
Heterozygote ,Nuclear Transfer Techniques ,Swine ,Phenylalanine ,Genetic Vectors ,Cystic Fibrosis Transmembrane Conductance Regulator ,medicine.disease_cause ,Cystic fibrosis ,Animals, Genetically Modified ,medicine ,Animals ,RNA, Messenger ,Allele ,ΔF508 ,Adeno-associated virus ,Alleles ,Mutation ,Genome ,biology ,Gene targeting ,Heterozygote advantage ,General Medicine ,Dependovirus ,Fibroblasts ,medicine.disease ,Molecular biology ,Cystic fibrosis transmembrane conductance regulator ,Cell biology ,Technical Advance ,Gene Expression Regulation ,Gene Targeting ,biology.protein - Abstract
Progress toward understanding the pathogenesis of cystic fibrosis (CF) and developing effective therapies has been hampered by lack of a relevant animal model. CF mice fail to develop the lung and pancreatic disease that cause most of the morbidity and mortality in patients with CF. Pigs may be better animals than mice in which to model human genetic diseases because their anatomy, biochemistry, physiology, size, and genetics are more similar to those of humans. However, to date, gene-targeted mammalian models of human genetic disease have not been reported for any species other than mice. Here we describe the first steps toward the generation of a pig model of CF. We used recombinant adeno-associated virus (rAAV) vectors to deliver genetic constructs targeting the CF transmembrane conductance receptor (CFTR) gene to pig fetal fibroblasts. We generated cells with the CFTR gene either disrupted or containing the most common CF-associated mutation (DeltaF508). These cells were used as nuclear donors for somatic cell nuclear transfer to porcine oocytes. We thereby generated heterozygote male piglets with each mutation. These pigs should be of value in producing new models of CF. In addition, because gene-modified mice often fail to replicate human diseases, this approach could be used to generate models of other human genetic diseases in species other than mice.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.