Skip to main content
Annals of the American Thoracic Society logoLink to Annals of the American Thoracic Society
. 2016 Apr;13(Suppl 2):S169–S176. doi: 10.1513/AnnalsATS.201509-601KV

Therapeutic Approaches to Acquired Cystic Fibrosis Transmembrane Conductance Regulator Dysfunction in Chronic Bronchitis

George M Solomon 1,2,*, S Vamsee Raju 1,2,*, Mark T Dransfield 1,3, Steven M Rowe 1,2,3,4,5,
PMCID: PMC5015723  PMID: 27115953

Abstract

Chronic obstructive pulmonary disease is a common cause of morbidity and a rising cause of mortality worldwide. Its rising impact indicates the ongoing unmet need for novel and effective therapies. Previous work has established a pathophysiological link between the chronic bronchitis phenotype of chronic obstructive pulmonary disease and cystic fibrosis as well as phenotypic similarities between these two airways diseases. An extensive body of evidence has established that cigarette smoke and its constituents contribute to acquired dysfunction of the cystic fibrosis transmembrane conductance regulator (CFTR) protein in the airways, pointing to a mechanistic link with smoking-related and chronic bronchitis. Recent interest surrounding new drugs that target both mutant and wild-type CFTR channels has paved the way for a new treatment opportunity addressing the mucus defect in chronic bronchitis. We review the clinical and pathologic evidence for modulating CFTR to address acquired CFTR dysfunction and pragmatic issues surrounding clinical trials as well as a discussion of other ion channels that may represent alternative therapeutic targets.

Keywords: chronic obstructive pulmonary disease, cystic fibrosis transmembrane conductance regulator, chronic bronchitis


Chronic obstructive pulmonary disease (COPD) is a major international health concern and the third leading cause of death in the United States (1). Most cases are due to cigarette smoking, although other environmental exposures can also cause disease, including heavy metals and toxic fumes such as pollution or diesel particulates. Alpha-1 antitrypsin deficiency is also responsible for approximately 2 to 3% of cases and can precipitate an early presentation, especially in the context of smoking (2). COPD is associated with two predominant clinical phenotypes, emphysema and chronic bronchitis; however, they often overlap, and the distinction in most patients is imprecise (35). Chronic bronchitis is particularly troubling as it can often dominate symptoms, is associated with pulmonary exacerbations, and presently has few treatment options (4, 5). As chronic bronchitis shares many clinical and pathologic features with cystic fibrosis (CF), an archetypal obstructive lung disease caused by genetic mutations in the cystic fibrosis transmembrane conductance regulator (CFTR), it is possible that there are common mechanisms and shared therapeutic strategies. This has been bolstered by the recognition that cigarette smoking and COPD itself induce an acquired state of CFTR protein dysfunction in patients with normal CFTR genes. Furthermore, the discovery of CFTR potentiators, including the recently Food and Drug Administration–approved agent ivacaftor, which can also activate wild-type CFTR, has generated excitement that ion transport activation may represent an important therapeutic target for the treatment of COPD (6). Here we review the potential contribution of acquired CFTR dysfunction to the pathophysiology of COPD, and in particular chronic bronchitis, with a focus on potential therapies.

Acquired CFTR Dysfunction in Smoking

There is a longstanding interest in the effects of cigarette smoking on epithelial ion transport since Welsh reported that cigarette smoke extract decreased chloride (Cl) secretion across canine airway epithelium, even before CFTR had been identified (7). Years later, Kreindler and colleagues established that cigarette smoke extract decreases CFTR-dependent Cl secretion in vitro (8). Cantin and colleagues confirmed these findings in Calu-3 monolayers, an airway glandular cell line, and showed that the effects of cigarette smoke were not specific to pulmonary epithelium by demonstrating similar effects on CFTR expression and function in the intestinal epithelial cell line T84 (9). Moreover, in a retrospective analysis using nasal potential difference (NPD) involving men with and without a history of smoking and the absence of CFTR genetic mutations, healthy smokers exhibited CFTR deficiency, as observed by a blunted NPD response to Cl-free isoproterenol perfusion, indicating reduced CFTR activity in vivo. Administration of cigarette smoke to the nares of healthy smokers caused an acute blockade of CFTR activity, as measured by NPD, establishing the causality of cigarette smoke and indicating the kinetics of this reaction are relatively rapid (10).

Emerging data from multiple prospective studies have established that acquired CFTR dysfunction caused by smoking is not limited to healthy smokers; it also affects patients with COPD and correlates with disease severity and clinical symptoms. Cigarette smokers and patients with COPD exhibited reduced CFTR function by NPD, and this was accompanied by reduced mRNA transcript levels from the nares (6, 11). Furthermore, lower airway potential difference measured via bronchoscopy was also reduced in a distinct cohort; functional decrements were confirmed by Western blot of endobronchial biopsies indicating reduced protein expression (11). In each of these studies, patients with COPD who stopped smoking for at least 1 year did not exhibit CFTR dysfunction, although there was some variability between subjects, particularly in the lower airway where other factors such as hypoxia, neutrophilic inflammation, and goblet cell hyperplasia may also contribute to CFTR deficits (12, 13). Furthermore, CFTR dysfunction was also associated with symptoms of chronic bronchitis, supporting the notion that abnormal epithelial anion transport may contribute to this phenotype, which like CF is plagued by chronic mucus hypersecretion (6, 11). Supporting the overall hypothesis, the expression of CFTR was positively correlated with FEV1 in the lungs of patients with COPD, whereas expression of negative regulators of airway hydration, including ENaC, was negatively correlated to lung function (14).

Although reduced CFTR activity in the airway was supported by previous studies in the epithelial cells, surprisingly, deficient CFTR activity was not limited to the respiratory epithelium in vivo. Rather, systemic (nonrespiratory) CFTR dysfunction was established by elevated sweat chloride concentration in smokers and patients with COPD and was also observed using intestinal current measurements in a confirmatory analysis in this patient population (15, 16). β-adrenergic–stimulated sweat rate, a parameter dependent on CFTR-mediated anion secretion in the coil of the sweat gland, confirmed these findings and also replicated the abnormality in sweat chloride (16). Interestingly, both sweat chloride and sweat rate were altered in former smokers with COPD, suggesting residual CFTR dysfunction is present in this population, a finding distinct from parameters measured on the airway surface. The reason for the difference in these tissues is unknown but may be due to the relative sensitivity of the sweat assays for detecting small degrees of CFTR dysfunction or differences in the biology of the sweat gland from the respiratory epithelium. Each of these studies excluded patients who harbor one or more genetic mutations in CFTR, indicating inheritance was not a factor in these diseases. Sweat rate was also shown to partially recover after smoking cessation, an indicator of causality, although this was not evident until 2 to 3 weeks after quitting, suggesting some time is required to demonstrate improved CFTR activity (17).

There are thousands of noxious substances in cigarette smoke, but a few constituents dominate its toxic effects. Current evidence implicates acrolein and cadmium as cigarette smoke constituents that block CFTR function in vitro, and they can also reach detectable levels in humans (15, 18). These agents may be potentially amenable to therapeutic intervention. The mechanistic basis of reduced CFTR dysfunction on a molecular level continues to evolve, as more than one pathway likely contributes, including those that effect CFTR mRNA expression, protein stability, and channel gating. In vitro and in vivo studies have shown cigarette smoke affects mRNA levels (6, 10). CFTR expression was found to be altered by up-regulation of miR-101, a repressive miRNA that modulates CFTR 3′-UTR to affect its transcription (19). Epigenetic effects have also been postulated (20, 21) as well as changes in protein expression (6, 10). In addition, CFTR surface expression is altered by sequestration of channels into an aggresome-like perinuclear (but not lysosomal) compartment that was driven by smoke-induced spikes in intracellular Ca2+ mobilization (10, 22). Finally, cigarette smoke has been shown to directly reduce channel gating as measured by altered open probability (23); this was recapitulated by acrolein exposure (15). In addition, cigarette smoke induces transforming growth factor-β1, which modulates CFTR activation (via β-adrenergic stimulation) via endogenous stimulation (24). Because smoking also increases mucus expression and reduces cilia beat frequency and ciliogenesis, even small decrements in CFTR function are likely to impose marked effects on airway physiology and mucus clearance (25).

Therapeutic Challenges in Chronic Obstructive Pulmonary Disease

Although COPD is among the most common causes of death worldwide, no current pharmacologic treatments alter its natural history (2628). Although bronchodilators (frequently in combination with inhaled corticosteroids) are the mainstay of treatment and do reduce exacerbations, improve dyspnea, and increase quality of life, they do not definitively alter mortality (2931). In addition, inhaled steroids in COPD are associated with an increased risk of pneumonia and do not alter mucus obstruction or its clearance (29, 32, 33). Mucolytic therapy for COPD has yielded marginal benefits thus far (3436). This has been attributed to the lack of efficacy of currently available agents, poor bioavailability, and failure to deliver the drug to the distal airways where mucus obstruction is observed (3740). An improved approach that targets the small airways (with a potent and orally available agent) could be expected to overcome these limitations and thus improve mucus stasis, an important cause of residual airway obstruction in COPD that cannot be addressed by bronchodilators alone, and a feature closely linked to mortality and lung function decline (4143).

Like lung cancer and coronary artery disease, the need for new COPD therapies is well accepted, despite evidence that cigarette smoking is the major contributing and modifiable risk factor (44, 45). Even after cessation, COPD continues to progress, and airflow obstruction is present in a substantial subset of ex-smokers (i.e., ∼4–5 million people), of whom up to half exhibit chronic bronchitis (46).

Crossover between Therapies for Cystic Fibrosis and Chronic Obstructive Pulmonary Disease

The shared pathophysiologic features between CF and COPD have led to a number of prior investigations examining the potential role of therapeutics developed for patients with one of these airway diseases for use in the other. Although not all efforts have been successful (4750), randomized trials have demonstrated the shared benefits of intravenous corticosteroids during exacerbations (51, 52), chest physiotherapy (5355), and, most recently, the chronic administration of azithromycin to reduce acute exacerbations (56, 57). This experience suggests that some agents effective in CF may also be efficacious in COPD.

Rescue of CFTR Function: A New Paradigm in Cystic Fibrosis Care

“CFTR potentiators” are a class of agents developed to correct the underlying cause of CF and principally target surface mutations. CFTR mutations can be grouped into several categories based on the molecular defect. Class I and II mutations are generally severe and result in relatively little protein product at the cell membrane. In comparison, class III (severe gating mutants, e.g., G551D), class IV (altered conductance), and class V (reduced expression) CFTR defects (5860) have relatively preserved cell surface expression and more closely resemble the situation in COPD. The archetype agent ivacaftor (formerly VX-770) robustly activates multiple CFTR forms, including wild type, G551D (a class III mutation), and other mutations with residual expression or function (class III, IV, and V) in primary airway epithelial monolayers, but only if they are located at the cell surface in sufficient number (6163). Ivacaftor is now Food and Drug Administration approved for treatment of CF with several mutations. Ivacaftor confers robust activation of G551D to markedly improve CFTR function, mucociliary clearance, pulmonary function, and exacerbation frequency in patients with CF (64); positive effects are also observed in the class IV allele R117H (65).

Although CFTR potentiators were originally developed to restore activity to mutant CFTR, some CFTR potentiators also augment wild-type CFTR function (6, 61). For example, both ivacaftor and other CFTR potentiators activate wild-type CFTR by augmenting open channel probability (62, 66, 67), which is reduced by smoke exposure (23). Because CFTR regulates airway surface liquid (ASL) depth (68), which in turn determines mucociliary clearance (6971), it follows that potentiation of CFTR anion secretion should result in augmented mucus transport compared with resting conditions (72, 73).

CFTR Potentiators Reverse Acquired CFTR Abnormalities and Augment Mucociliary Transport In Vitro

In smoke-exposed epithelial cells, ivacaftor potentiated CFTR-dependent short-circuit current, regardless of prior smoke exposure and to levels that exceeded wild-type CFTR activity (6) Ivacaftor also potentiated CFTR-dependent currents in normal explanted human trachea. The effects of ivacaftor were similar whether the individual was an active smoker or nonsmoker, providing evidence of strong activity of ivacaftor in tissue (6). Confocal microscopy revealed ivacaftor increased ASL depth in smoke-exposed epithelia and caused a large increase in mucociliary transport rate (MCT) (6, 15). Hypertonic saline, which increases ASL depth in vitro by osmotic force, also improved MCT in vitro, indicating CFTR can be circumvented (10). Of note, all studies examining the effect of potentiators on ASL depth and MCT were performed with “chronic” (24 h) exposure, indicating concerns regarding the effect of ivacaftor on CFTR stability did not substantially diminish treatment benefit (74). In total, these data strongly indicate that CFTR potentiators and other agonists of ion transport can reverse CFTR dysfunction induced by smoking, augmenting ASL depth and increasing MCT in airway epithelia (6). Whether this is principally due to CFTR-mediated chloride secretion and its resultant effects on ASL hydration, or partial inhibition of CFTR-dependent bicarbonate transport, which can influence mucus viscosity (75) and adhesion to the airway surface (76) observed in the latest models of the CF airway, deserves exploration.

Roflumilast and Its Role in Chronic Bronchitis

Roflumilast is a phosphodiesterase-4 (PDE-4) inhibitor approved for the treatment of frequent exacerbations in patients with COPD with chronic bronchitis (77). It was advanced for testing as a putative antiinflammatory, because PDEs are highly expressed in a variety of inflammatory cells. Interestingly, roflumilast was only shown to be effective in patients with chronic bronchitis, raising questions as to whether antiinflammatory properties were the sole reason for its therapeutic benefit (78). As a PDE inhibitor, roflumilast also increased cellular cAMP levels in vitro and activated CFTR by augmenting its phosphorylation in a manner that was competitive with cAMP-dependent stimuli, a distinction from CFTR potentiators (79). Roflumilast is relatively specific for PDE4 and has minimal effects on cGMP. Roflumilast was also effective in restoring CFTR function in human bronchial epithelial cells exposed to cigarette smoke (80). Downstream of CFTR augmentation, roflumilast augmented ASL depth, including cells affected by cigarette smoking (79, 80). Although Tyrrell and colleagues did not observe an increase in cAMP or ASL depth when human bronchial epithelial (HBE) cells were treated with roflumilast alone, roflumilast did augmented cAMP levels and ASL hydration in presence of adenosine, a physiologic activator of cAMP and CFTR in the airways (80). Moreover, roflumilast was found to be superior to β-agonists such as salmeterol and isoproterenol in restoring ASL height. Currently, there are no published reports regarding the effect of roflumilast on CFTR expression.

Roflumilast-N-oxide, the more stable and potent metabolite of roflumilast, protected against cigarette smoke–induced loss of ciliated epithelium and the rate of ciliary beating (81). Taken together, the bioactivity of roflumilast on CFTR function suggests it may partially explain its therapeutic benefit in patients with COPD with chronic bronchitis and frequent exacerbations, because this phenotype is most responsive to roflumilast therapy and also is associated with an increased prevalence of CFTR dysfunction (11, 15); this also adds additional credence to the hypothesis that CFTR potentiation may be beneficial in COPD. Interestingly, CFTR activation and fluid secretion in the intestine was also shown to occur with roflumilast and could explain the adverse effect of diarrhea commonly observed in the clinic (79). Inhaled PDE inhibition could be an approach to circumvent this side effect.

Patient Selection for Trials with CFTR Potentiators and Activators

The heterogeneity of COPD pathophysiology and phenotypic expression has been a challenge to drug development and raises the importance of patient selection for the evaluation of experimental therapeutics (82). Selection criteria to target the COPD subphenotypes likely to be responsive to CFTR or other ion transport pathways will be imperative, especially in the early phases of drug development. The presence of smoking would be logical criterion for patient selection, because smoking clearly contributes to acquired CFTR dysfunction. However, we posit the presence of chronic bronchitis would be more clinically relevant and possibly more feasible, as this clinical criterion is associated with CFTR dysfunction and also with mucus hyperplasia, which together contribute to impaired mucociliary clearance in the small airways (4, 5, 83). The presence of chronic bronchitis also carries a much higher risk of exacerbation and morbidity (3, 41), representing significant unmet medical need not addressed by current therapies. The benefits of CFTR activation may be generalizable beyond patients with COPD with chronic bronchitis, however, as even patients without chronic mucus hypersecretion exhibit airways obstruction due to mucus impaction of the small airways, as detected by endobronchial biopsy (43). Of the two clinical criteria noted, the presence of chronic bronchitis may be most appropriate, because new treatments that encourage continued smoking by abating symptoms could be problematic.

Laboratory studies could also be used to select patients likely to respond to CFTR potentiators or other anion transport activators. An obvious strategy would be to populate the study with individuals with clear evidence of CFTR dysfunction. Sweat chloride would be highly feasible for this purpose, because the test is reproducible, well standardized, and is sensitive to even modest degrees of CFTR dysfunction (15, 84). Our previous studies indicate that 40 to 50% of patients with Global Initiative for Chronic Obstructive Lung Disease stage II or greater COPD demonstrate significant elevation of sweat chloride (although still below the diagnostic criteria of CF) compared with healthy nonsmokers (15). NPD could also be performed and has the potential advantage of reflecting the status of CFTR function in the airway (6). It is not clear whether CF-carrier status would impact likelihood of treatment response. So far, relatively small studies have not consistently detected an association between chronic bronchitis and CFTR carrier status (85), although it is possible an effect of CFTR mutations could be confirmed with larger study cohorts. On the other hand, the presence of an allele that does not respond to a CFTR potentiator (at least as compared with a wild-type channel) may also diminish the treatment effect.

Biomarkers of CFTR and Other Indicators of Clinical Efficacy

Experience in CF indicates that improved CFTR function measured by sweat chloride or NPD predicts improved lung function on a group-wise basis with CFTR potentiator therapy (84). However, this has not yet been proven in COPD; thus, a comprehensive approach to clinical endpoints and biomarkers downstream of CFTR seem warranted. Other measures of CFTR function besides these two endpoints that could be considered include function of the secretory coil of the sweat gland using β-adrenergic–stimulated sweat secretion. This assay is sensitive to small decrements in normal CFTR function (86) and to the COPD phenotype among smokers (16); furthermore, β-adrenergic sweat rate improves in healthy smokers on initiating a smoking cessation program, indicating the assay is dynamic within an individual (17). Other markers intermediate to clinical efficacy could also be useful. Augmentation of CFTR activity in COPD, if acquired CFTR dysfunction contributes to COPD pathogenesis, should confer improved mucociliary clearance, as observed with ivacaftor treatment in patients with the G551D CFTR mutation (64); this can be measured by clearance of Tc99-radiolabeled particles (72). Other indicators could include resolution of hyperinflation, small airway obstruction, or bronchial wall thickening as detected by changes in high-resolution computed tomography scan. These studies have also been illustrative in patients with CF treated with ivacaftor, even in relatively short-term studies (87, 88). Patient symptoms could also be informative, as there are a number of validated outcome measures in COPD that quantify mucus production (8991). Ultimately, clinical efficacy will have to be demonstrated by changes in spirometry and exacerbation frequency, but these endpoints require relatively large studies. Even in CF, CFTR biomarkers have not predicted whether a patient experiences improved lung function on an individual basis (92). Thus, there is much to be learned about whether an improvement in a short-term biomarker can be used to select patients likely to benefit from prolonged administration of a new COPD therapy.

Potential Contribution of Other Ion Channels and Their Therapeutic Potential

Although mucosal defects in CF highlight the pathophysiologic role played by CFTR, there are several other epithelial channels known to conduct chloride across the airway surface. These channels function in concert to generate a luminal chloride gradient that drives paracellular sodium and water to sustain isosmotic mucosal secretions. As such, epithelial chloride channels do not function in isolation, and they remain electrochemically in tandem with other apical Na+ channels, anion exchangers, basolateral Na+/K+ pumps, and K+ channels to maintain luminal osmolality.

Calcium-activated chloride channels (CaCC) are a large family of apical chloride channels that are activated in response to increased intracellular Ca2+ (93). Although initially associated with Ca2+ mobilization by muscarinic stimulation, they are now known to respond to a wide variety of Ca2+ agonists (94). Attempts to define molecular identity of CaCCs have identified several interesting candidates (e.g., CLCA1, CLC3, hTTYH3) over three decades (95). Three independent laboratories identified TMEM16A (also known as anactomin-1) with convincing properties of a CaCC (9698). Gene silencing and murine knockout models have since validated its pharmacologic selectivity and tissue distribution (99, 100). Nearly 50% of uridine 5′-triphosphate (UTP)-stimulated airway CaCC activity was attributed to TMEM16A, and its pharmacologic inhibitors were found to abolish only the transient CaCC currents (101). Identity of other CaCC channels remains elusive, and successful characterization of TMEM16A highlights the likelihood of discovering additional members using advanced experimental and theoretical approaches. Because alternated chloride channels continue to remain a prime focus for developing CF therapy, TMEM16A, with its established biology, has been at the forefront of such efforts (102, 103).

SLC26A is another large family of anion transporters known to mediate epithelial Cl and HCO3 secretion. Members of this family are of significant interest to CF research, because they directly interact with CFTR domains and mutually augment anion secretion by sharing regulatory proteins such as protein kinase A (104, 105). SLC26A9 is one such apical channel that is responsive to cAMP/protein kinase A–mediated stimulation and accounts for basal ion secretion after inhibition of epithelial sodium absorption (106). Interestingly, SLC26A9 is known to function synergistically with wild-type CFTR (but not with mutated CFTR) to conduct anion transport (105). Thus, interactions between CFTR and SLC26A channels might be exploited in acquired decrements of the CFTR channel. As such, alternate chloride channels are an attractive drug target that can augment anion secretion independent of CFTR (107), potentially circumventing acquired CFTR dysfunction. The potential of the epithelial sodium channel (ENaC), a key ion channel that regulates ASL depth and mucus hydration on the epithelial cell surface, as a therapy in COPD is reviewed elsewhere in this issue.

Conclusions

Previous work indicates that cigarette smoking, the most common cause of chronic bronchitis, results in a functional CFTR defect in the airways and also systemically. The success of CFTR modulators for the treatment of CF has resulted in significant interest in determining whether these agents may also be useful in chronic bronchitis. Preclinical studies demonstrate the capacity of CFTR potentiators to restore airway surface liquid defect and augment mucociliary transport in human cells and tissues by augmenting CFTR function, suggesting their potential as a therapeutic agent in the disease. Although disease heterogeneity is a challenge, several tools for patient selection are apparent, including targeting patients with COPD with chronic bronchitis or enriching the population for individuals with abnormal CFTR function. Tools to detect efficacy include measures of CFTR activity, physiologic assays of mucus clearance, and measures of clinical efficacy. The confluence of an improved understanding of airway biology, comprehensive patient phenotyping, and availability of new therapies provides new opportunities to explore CFTR modulators and other ion transport agonists as new therapeutic agents that target COPD at the level of the airway epithelium.

Footnotes

Supported by the National Institutes of Health grants R01 HL105487 (S.M.R.), P30 DK072482 (S.M.R.), and 5 UL1 RR025777; Cystic Fibrosis Foundation grants CLANCY09Y0 (S.M.R.), R464-CF (S.M.R.), and SORSCH15RO (G.M.S); and American Lung Association grant RG-305752 (S.V.R.) and the Flight Attendants Medical Research Association grant YFA130008 (S.V.R.).

The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript

Author Contributions: G.M.S., S.V.R., M.T.D., and S.M.R. all contributed to manuscript preparation and final editing. All authors approved the final manuscript draft before submission.

Author disclosures are available with the text of this article at www.atsjournals.org .

References

  • 1.Vestbo J, Hurd SS, Agustí AG, Jones PW, Vogelmeier C, Anzueto A, Barnes PJ, Fabbri LM, Martinez FJ, Nishimura M, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2013;187:347–365. doi: 10.1164/rccm.201204-0596PP. [DOI] [PubMed] [Google Scholar]
  • 2.American Thoracic Society; European Respiratory Society. American Thoracic Society/European Respiratory Society statement: standards for the diagnosis and management of individuals with alpha-1 antitrypsin deficiency. Am J Respir Crit Care Med. 2003;168:818–900. doi: 10.1164/rccm.168.7.818. [DOI] [PubMed] [Google Scholar]
  • 3.Kim V, Han MK, Vance GB, Make BJ, Newell JD, Hokanson JE, Hersh CP, Stinson D, Silverman EK, Criner GJ COPDGene Investigators. The chronic bronchitic phenotype of COPD: an analysis of the COPDGene Study. Chest. 2011;140:626–633. doi: 10.1378/chest.10-2948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kim V, Criner GJ. Chronic bronchitis and chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;187:228–237. doi: 10.1164/rccm.201210-1843CI. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kim V, Sternberg AL, Washko G, Make BJ, Han MK, Martinez F, Criner GJ National Emphysema Treatment Trial Research Group. Severe chronic bronchitis in advanced emphysema increases mortality and hospitalizations. COPD. 2013;10:667–678. doi: 10.3109/15412555.2013.827166. [DOI] [PubMed] [Google Scholar]
  • 6.Sloane PA, Shastry S, Wilhelm A, Courville C, Tang LP, Backer K, Levin E, Raju SV, Li Y, Mazur M, et al. A pharmacologic approach to acquired cystic fibrosis transmembrane conductance regulator dysfunction in smoking related lung disease. Plos One. 2012;7:e39809. doi: 10.1371/journal.pone.0039809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Welsh MJ. Cigarette smoke inhibition of ion transport in canine tracheal epithelium. J Clin Invest. 1983;71:1614–1623. doi: 10.1172/JCI110917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kreindler JL, Jackson AD, Kemp PA, Bridges RJ, Danahay H. Inhibition of chloride secretion in human bronchial epithelial cells by cigarette smoke extract. Am J Physiol Lung Cell Mol Physiol. 2005;288:L894–L902. doi: 10.1152/ajplung.00376.2004. [DOI] [PubMed] [Google Scholar]
  • 9.Cantin AM, Hanrahan JW, Bilodeau G, Ellis L, Dupuis A, Liao J, Zielenski J, Durie P. Cystic fibrosis transmembrane conductance regulator function is suppressed in cigarette smokers. Am J Respir Crit Care Med. 2006;173:1139–1144. doi: 10.1164/rccm.200508-1330OC. [DOI] [PubMed] [Google Scholar]
  • 10.Clunes LA, Davies CM, Coakley RD, Aleksandrov AA, Henderson AG, Zeman KL, Worthington EN, Gentzsch M, Kreda SM, Cholon D, et al. Cigarette smoke exposure induces CFTR internalization and insolubility, leading to airway surface liquid dehydration. FASEB J. 2012;26:533–545. doi: 10.1096/fj.11-192377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Dransfield MT, Wilhelm AM, Flanagan B, Courville C, Tidwell SL, Raju SV, Gaggar A, Steele C, Tang LP, Liu B, et al. Acquired cystic fibrosis transmembrane conductance regulator dysfunction in the lower airways in COPD. Chest. 2013;144:498–506. doi: 10.1378/chest.13-0274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Guimbellot JS, Fortenberry JA, Siegal GP, Moore B, Wen H, Venglarik C, Chen YF, Oparil S, Sorscher EJ, Hong JS. Role of oxygen availability in CFTR expression and function. Am J Respir Cell Mol Biol. 2008;39:514–521. doi: 10.1165/rcmb.2007-0452OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Le Gars M, Descamps D, Roussel D, Saussereau E, Guillot L, Ruffin M, Tabary O, Hong SS, Boulanger P, Paulais M, et al. Neutrophil elastase degrades cystic fibrosis transmembrane conductance regulator via calpains and disables channel function in vitro and in vivo. Am J Respir Crit Care Med. 2013;187:170–179. doi: 10.1164/rccm.201205-0875OC. [DOI] [PubMed] [Google Scholar]
  • 14.Zhao R, Liang X, Zhao M, Liu SL, Huang Y, Idell S, Li X, Ji HL. Correlation of apical fluid-regulating channel proteins with lung function in human COPD lungs. Plos One. 2014;9:e109725. doi: 10.1371/journal.pone.0109725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Raju SV, Jackson PL, Courville CA, McNicholas CM, Sloane PA, Sabbatini G, Tidwell S, Tang LP, Liu B, Fortenberry JA, et al. Cigarette smoke induces systemic defects in cystic fibrosis transmembrane conductance regulator function. Am J Respir Crit Care Med. 2013;188:1321–1330. doi: 10.1164/rccm.201304-0733OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Courville CA, Tidwell S, Liu B, Accurso FJ, Dransfield MT, Rowe SM. Acquired defects in CFTR-dependent β-adrenergic sweat secretion in chronic obstructive pulmonary disease. Respir Res. 2014;15:25. doi: 10.1186/1465-9921-15-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Courville CA, Raju SV, Liu B, Accurso FJ, Dransfield MT, Rowe SM. Recovery of acquired cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction following smoking cessation. Am J Respir Crit Care Med. 2015;192:1521–1524. doi: 10.1164/rccm.201502-0396LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rennolds J, Butler S, Maloney K, Boyaka PN, Davis IC, Knoell DL, Parinandi NL, Cormet-Boyaka E. Cadmium regulates the expression of the CFTR chloride channel in human airway epithelial cells. Toxicol Sci. 2010;116:349–358. doi: 10.1093/toxsci/kfq101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Hassan F, Nuovo GJ, Crawford M, Boyaka PN, Kirkby S, Nana-Sinkam SP, Cormet-Boyaka E. MiR-101 and miR-144 regulate the expression of the CFTR chloride channel in the lung. Plos One. 2012;7:e50837. doi: 10.1371/journal.pone.0050837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kelsen SG, Duan X, Ji R, Perez O, Liu C, Merali S. Cigarette smoke induces an unfolded protein response in the human lung: a proteomic approach. Am J Respir Cell Mol Biol. 2008;38:541–550. doi: 10.1165/rcmb.2007-0221OC. [DOI] [PubMed] [Google Scholar]
  • 21.Ribeiro CM, O’Neal WK. Endoplasmic reticulum stress in chronic obstructive lung diseases. Curr Mol Med. 2012;12:872–882. doi: 10.2174/156652412801318791. [DOI] [PubMed] [Google Scholar]
  • 22.Rasmussen JE, Sheridan JT, Polk W, Davies CM, Tarran R. Cigarette smoke-induced Ca2+ release leads to cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction. J Biol Chem. 2014;289:7671–7681. doi: 10.1074/jbc.M113.545137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Moran AR, Norimatsu Y, Dawson DC, MacDonald KD. Aqueous cigarette smoke extract induces a voltage-dependent inhibition of CFTR expressed in Xenopus oocytes. Am J Physiol Lung Cell Mol Physiol. 2014;306:L284–L291. doi: 10.1152/ajplung.00163.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Unwalla HJ, Ivonnet P, Dennis JS, Conner GE, Salathe M. Transforming growth factor-β1 and cigarette smoke inhibit the ability of β2-agonists to enhance epithelial permeability. Am J Respir Cell Mol Biol. 2015;52:65–74. doi: 10.1165/rcmb.2013-0538OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tamashiro E, Xiong G, Anselmo-Lima WT, Kreindler JL, Palmer JN, Cohen NA. Cigarette smoke exposure impairs respiratory epithelial ciliogenesis. Am J Rhinol Allergy. 2009;23:117–122. doi: 10.2500/ajra.2009.23.3280. [DOI] [PubMed] [Google Scholar]
  • 26.Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P, Fukuchi Y, Jenkins C, Rodriguez-Roisin R, van Weel C, et al. Global Initiative for Chronic Obstructive Lung Disease. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2007;176:532–555. doi: 10.1164/rccm.200703-456SO. [DOI] [PubMed] [Google Scholar]
  • 27.Mannino DM. COPD: epidemiology, prevalence, morbidity and mortality, and disease heterogeneity. Chest. 2002;121:121S–126S. doi: 10.1378/chest.121.5_suppl.121s. [DOI] [PubMed] [Google Scholar]
  • 28.Murray CJ, Lopez AD. Alternative projections of mortality and disability by cause 1990-2020: Global Burden of Disease Study. Lancet. 1997;349:1498–1504. doi: 10.1016/S0140-6736(96)07492-2. [DOI] [PubMed] [Google Scholar]
  • 29.Drummond MB, Dasenbrook EC, Pitz MW, Murphy DJ, Fan E. Inhaled corticosteroids in patients with stable chronic obstructive pulmonary disease: a systematic review and meta-analysis. JAMA. 2008;300:2407–2416. doi: 10.1001/jama.2008.717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, Yates JC, Vestbo J TORCH investigators. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med. 2007;356:775–789. doi: 10.1056/NEJMoa063070. [DOI] [PubMed] [Google Scholar]
  • 31.Tashkin DP, Celli B, Senn S, Burkhart D, Kesten S, Menjoge S, Decramer M UPLIFT Study Investigators. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543–1554. doi: 10.1056/NEJMoa0805800. [DOI] [PubMed] [Google Scholar]
  • 32.Crim C, Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, Willits LR, Yates JC, Vestbo J. Pneumonia risk in COPD patients receiving inhaled corticosteroids alone or in combination: TORCH study results. Eur Respir J. 2009;34:641–647. doi: 10.1183/09031936.00193908. [DOI] [PubMed] [Google Scholar]
  • 33.Singh S, Loke YK. Risk of pneumonia associated with long-term use of inhaled corticosteroids in chronic obstructive pulmonary disease: a critical review and update. Curr Opin Pulm Med. 2010;16:118–122. doi: 10.1097/MCP.0b013e328334c085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Decramer M, Rutten-van Mölken M, Dekhuijzen PN, Troosters T, van Herwaarden C, Pellegrino R, van Schayck CP, Olivieri D, Del Donno M, De Backer W, et al. Effects of N-acetylcysteine on outcomes in chronic obstructive pulmonary disease (Bronchitis Randomized on NAC Cost-Utility Study, BRONCUS): a randomised placebo-controlled trial. Lancet. 2005;365:1552–1560. doi: 10.1016/S0140-6736(05)66456-2. [DOI] [PubMed] [Google Scholar]
  • 35.Zheng JP, Kang J, Huang SG, Chen P, Yao WZ, Yang L, Bai CX, Wang CZ, Wang C, Chen BY, et al. Effect of carbocisteine on acute exacerbation of chronic obstructive pulmonary disease (PEACE Study): a randomised placebo-controlled study. Lancet. 2008;371:2013–2018. doi: 10.1016/S0140-6736(08)60869-7. [DOI] [PubMed] [Google Scholar]
  • 36.Poole PJ, Black PN. Mucolytic agents for chronic bronchitis or chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2006;3:CD001287. doi: 10.1002/14651858.CD001287.pub2. [DOI] [PubMed] [Google Scholar]
  • 37.Sadowska AM, Manuel-Y-Keenoy B, De Backer WA. Antioxidant and anti-inflammatory efficacy of NAC in the treatment of COPD: discordant in vitro and in vivo dose-effects: a review. Pulm Pharmacol Ther. 2007;20:9–22. doi: 10.1016/j.pupt.2005.12.007. [DOI] [PubMed] [Google Scholar]
  • 38.Rogers DF, Barnes PJ. Treatment of airway mucus hypersecretion. Ann Med. 2006;38:116–125. doi: 10.1080/07853890600585795. [DOI] [PubMed] [Google Scholar]
  • 39.Rogers DF. Mucociliary dysfunction in COPD: effect of current pharmacotherapeutic options. Pulm Pharmacol Ther. 2005;18:1–8. doi: 10.1016/j.pupt.2004.08.001. [DOI] [PubMed] [Google Scholar]
  • 40.Black PN, Morgan-Day A, McMillan TE, Poole PJ, Young RP. Randomised, controlled trial of N-acetylcysteine for treatment of acute exacerbations of chronic obstructive pulmonary disease [ISRCTN21676344] BMC Pulm Med. 2004;4:13. doi: 10.1186/1471-2466-4-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Vestbo J, Prescott E, Lange P Copenhagen City Heart Study Group. Association of chronic mucus hypersecretion with FEV1 decline and chronic obstructive pulmonary disease morbidity. Am J Respir Crit Care Med. 1996;153:1530–1535. doi: 10.1164/ajrccm.153.5.8630597. [DOI] [PubMed] [Google Scholar]
  • 42.Hogg JC, Chu FS, Tan WC, Sin DD, Patel SA, Pare PD, Martinez FJ, Rogers RM, Make BJ, Criner GJ, et al. Survival after lung volume reduction in chronic obstructive pulmonary disease: insights from small airway pathology. Am J Respir Crit Care Med. 2007;176:454–459. doi: 10.1164/rccm.200612-1772OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:2645–2653. doi: 10.1056/NEJMoa032158. [DOI] [PubMed] [Google Scholar]
  • 44.Kandel DB, Kiros GE, Schaffran C, Hu MC. Racial/ethnic differences in cigarette smoking initiation and progression to daily smoking: a multilevel analysis. Am J Public Health. 2004;94:128–135. doi: 10.2105/ajph.94.1.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.McGorrian C, Yusuf S, Islam S, Jung H, Rangarajan S, Avezum A, Prabhakaran D, Almahmeed W, Rumboldt Z, Budaj A, et al. INTERHEART Investigators. Estimating modifiable coronary heart disease risk in multiple regions of the world: the INTERHEART Modifiable Risk Score. Eur Heart J. 2011;32:581–589. doi: 10.1093/eurheartj/ehq448. [DOI] [PubMed] [Google Scholar]
  • 46.Mannino DM, Gagnon RC, Petty TL, Lydick E. Obstructive lung disease and low lung function in adults in the United States: data from the National Health and Nutrition Examination Survey, 1988-1994. Arch Intern Med. 2000;160:1683–1689. doi: 10.1001/archinte.160.11.1683. [DOI] [PubMed] [Google Scholar]
  • 47.Jones AP, Wallis C. Dornase alfa for cystic fibrosis. Cochrane Database Syst Rev. 2010;3:CD001127. doi: 10.1002/14651858.CD001127.pub2. [DOI] [PubMed] [Google Scholar]
  • 48.Wark P, McDonald VM. Nebulised hypertonic saline for cystic fibrosis. Cochrane Database Syst Rev. 2009;(2):CD001506. doi: 10.1002/14651858.CD001506.pub3. [DOI] [PubMed] [Google Scholar]
  • 49.Ziment I. Pharmacologic therapy of obstructive airway disease. Clin Chest Med. 1990;11:461–486. [PubMed] [Google Scholar]
  • 50.Rogers DF. Mucoactive drugs for asthma and COPD: any place in therapy? Expert Opin Investig Drugs. 2002;11:15–35. doi: 10.1517/13543784.11.1.15. [DOI] [PubMed] [Google Scholar]
  • 51.Dovey M, Aitken ML, Emerson J, McNamara S, Waltz DA, Gibson RL. Oral corticosteroid therapy in cystic fibrosis patients hospitalized for pulmonary exacerbation: a pilot study. Chest. 2007;132:1212–1218. doi: 10.1378/chest.07-0843. [DOI] [PubMed] [Google Scholar]
  • 52.Niewoehner DE, Erbland ML, Deupree RH, Collins D, Gross NJ, Light RW, Anderson P, Morgan NA Department of Veterans Affairs Cooperative Study Group. Effect of systemic glucocorticoids on exacerbations of chronic obstructive pulmonary disease. N Engl J Med. 1999;340:1941–1947. doi: 10.1056/NEJM199906243402502. [DOI] [PubMed] [Google Scholar]
  • 53.McIlwaine PM, Wong LT, Peacock D, Davidson AG. Long-term comparative trial of conventional postural drainage and percussion versus positive expiratory pressure physiotherapy in the treatment of cystic fibrosis. J Pediatr. 1997;131:570–574. doi: 10.1016/s0022-3476(97)70064-7. [DOI] [PubMed] [Google Scholar]
  • 54.Hardy KA, Anderson BD. Noninvasive clearance of airway secretions. Respir Care Clin N Am. 1996;2:323–345. [PubMed] [Google Scholar]
  • 55.Chakravorty I, Chahal K, Austin G. A pilot study of the impact of high-frequency chest wall oscillation in chronic obstructive pulmonary disease patients with mucus hypersecretion. Int J Chron Obstruct Pulmon Dis. 2011;6:693–699. doi: 10.2147/COPD.S22896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Saiman L, Marshall BC, Mayer-Hamblett N, Burns JL, Quittner AL, Cibene DA, Coquillette S, Fieberg AY, Accurso FJ, Campbell PW, III Macrolide Study Group. Azithromycin in patients with cystic fibrosis chronically infected with Pseudomonas aeruginosa: a randomized controlled trial. JAMA. 2003;290:1749–1756. doi: 10.1001/jama.290.13.1749. [DOI] [PubMed] [Google Scholar]
  • 57.Albert RK, Connett J, Bailey WC, Casaburi R, Cooper JA, Jr, Criner GJ, Curtis JL, Dransfield MT, Han MK, Lazarus SC, et al. COPD Clinical Research Network. Azithromycin for prevention of exacerbations of COPD. N Engl J Med. 2011;365:689–698. doi: 10.1056/NEJMoa1104623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Galietta LJ, Springsteel MF, Eda M, Niedzinski EJ, By K, Haddadin MJ, Kurth MJ, Nantz MH, Verkman AS. Novel CFTR chloride channel activators identified by screening of combinatorial libraries based on flavone and benzoquinolizinium lead compounds. J Biol Chem. 2001;276:19723–19728. doi: 10.1074/jbc.M101892200. [DOI] [PubMed] [Google Scholar]
  • 59.Van Goor F, Straley KS, Cao D, González J, Hadida S, Hazlewood A, Joubran J, Knapp T, Makings LR, Miller M, et al. Rescue of DeltaF508-CFTR trafficking and gating in human cystic fibrosis airway primary cultures by small molecules. Am J Physiol Lung Cell Mol Physiol. 2006;290:L1117–L1130. doi: 10.1152/ajplung.00169.2005. [DOI] [PubMed] [Google Scholar]
  • 60.Pedemonte N, Lukacs GL, Du K, Caci E, Zegarra-Moran O, Galietta LJ, Verkman AS. Small-molecule correctors of defective DeltaF508-CFTR cellular processing identified by high-throughput screening. J Clin Invest. 2005;115:2564–2571. doi: 10.1172/JCI24898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Van Goor F, Hadida S, Grootenhuis PD, Burton B, Cao D, Neuberger T, Turnbull A, Singh A, Joubran J, Hazlewood A, et al. Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770. Proc Natl Acad Sci USA. 2009;106:18825–18830. doi: 10.1073/pnas.0904709106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Van Goor F, Yu H, Burton B, Hoffman BJ. Effect of ivacaftor on CFTR forms with missense mutations associated with defects in protein processing or function. J Cyst Fibros. 2014;13:29–36. doi: 10.1016/j.jcf.2013.06.008. [DOI] [PubMed] [Google Scholar]
  • 63.Yu H, Burton B, Huang CJ, Worley J, Cao D, Johnson JP, Jr, Urrutia A, Joubran J, Seepersaud S, Sussky K, et al. Ivacaftor potentiation of multiple CFTR channels with gating mutations. J Cyst Fibros. 2012;11:237–245. doi: 10.1016/j.jcf.2011.12.005. [DOI] [PubMed] [Google Scholar]
  • 64.Rowe SM, Heltshe SL, Gonska T, Donaldson SH, Borowitz D, Gelfond D, Sagel SD, Khan U, Mayer-Hamblett N, Van Dalfsen JM, et al. GOAL Investigators of the Cystic Fibrosis Foundation Therapeutics Development Network. Clinical mechanism of the CFTR potentiator ivacaftor in G551D-mediated cystic fibrosis. Am J Respir Crit Care Med. 2014;190:175–184. doi: 10.1164/rccm.201404-0703OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Moss RB, Flume PA, Elborn JS, Cooke J, Rowe SM, McColley SA, Rubenstein RC, Higgins M VX11-770-110 (KONDUCT) Study Group. Efficacy and safety of ivacaftor in patients with cystic fibrosis who have an Arg117His-CFTR mutation: a double-blind, randomised controlled trial. Lancet Respir Med. 2015;3:524–533. doi: 10.1016/S2213-2600(15)00201-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ren HY, Grove DE, De La Rosa O, Houck SA, Sopha P, Van Goor F, Hoffman BJ, Cyr DM. VX-809 corrects folding defects in cystic fibrosis transmembrane conductance regulator protein through action on membrane-spanning domain 1. Mol Biol Cell. 2013;24:3016–3024. doi: 10.1091/mbc.E13-05-0240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Pedemonte N, Tomati V, Sondo E, Galietta LJ. Influence of cell background on pharmacological rescue of mutant CFTR. Am J Physiol Cell Physiol. 2010;298:C866–C874. doi: 10.1152/ajpcell.00404.2009. [DOI] [PubMed] [Google Scholar]
  • 68.Tarran R, Loewen ME, Paradiso AM, Olsen JC, Gray MA, Argent BE, Boucher RC, Gabriel SE. Regulation of murine airway surface liquid volume by CFTR and Ca2+-activated Cl- conductances. J Gen Physiol. 2002;120:407–418. doi: 10.1085/jgp.20028599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Tarran R. Regulation of airway surface liquid volume and mucus transport by active ion transport. Proc Am Thorac Soc. 2004;1:42–46. doi: 10.1513/pats.2306014. [DOI] [PubMed] [Google Scholar]
  • 70.Roomans GM, Kozlova I, Nilsson H, Vanthanouvong V, Button B, Tarran R. Measurements of airway surface liquid height and mucus transport by fluorescence microscopy, and of ion composition by X-ray microanalysis. J Cyst Fibros. 2004;3:135–139. doi: 10.1016/j.jcf.2004.05.029. [DOI] [PubMed] [Google Scholar]
  • 71.Matsui H, Randell SH, Peretti SW, Davis CW, Boucher RC. Coordinated clearance of periciliary liquid and mucus from airway surfaces. J Clin Invest. 1998;102:1125–1131. doi: 10.1172/JCI2687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Donaldson SH, Corcoran TE, Laube BL, Bennett WD. Mucociliary clearance as an outcome measure for cystic fibrosis clinical research. Proc Am Thorac Soc. 2007;4:399–405. doi: 10.1513/pats.200703-042BR. [DOI] [PubMed] [Google Scholar]
  • 73.Sood N, Bennett WD, Zeman K, Brown J, Foy C, Boucher RC, Knowles MR. Increasing concentration of inhaled saline with or without amiloride: effect on mucociliary clearance in normal subjects. Am J Respir Crit Care Med. 2003;167:158–163. doi: 10.1164/rccm.200204-293OC. [DOI] [PubMed] [Google Scholar]
  • 74.Cholon DM, Quinney NL, Fulcher ML, Esther CR, Jr, Das J, Dokholyan NV, Randell SH, Boucher RC, Gentzsch M. Potentiator ivacaftor abrogates pharmacological correction of ΔF508 CFTR in cystic fibrosis. Sci Transl Med. 2014;6:246ra96. doi: 10.1126/scitranslmed.3008680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Birket SE, Chu KK, Liu L, Houser GH, Diephuis BJ, Wilsterman EJ, Dierksen G, Mazur M, Shastry S, Li Y, et al. A functional anatomic defect of the cystic fibrosis airway. Am J Respir Crit Care Med. 2014;190:421–432. doi: 10.1164/rccm.201404-0670OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hoegger MJ, Fischer AJ, McMenimen JD, Ostedgaard LS, Tucker AJ, Awadalla MA, Moninger TO, Michalski AS, Hoffman EA, Zabner J, et al. Impaired mucus detachment disrupts mucociliary transport in a piglet model of cystic fibrosis. Science. 2014;345:818–822. doi: 10.1126/science.1255825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rennard SI, Calverley PM, Goehring UM, Bredenbröker D, Martinez FJ. Reduction of exacerbations by the PDE4 inhibitor roflumilast: the importance of defining different subsets of patients with COPD. Respir Res. 2011;12:18. doi: 10.1186/1465-9921-12-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Fabbri LM, Calverley PM, Izquierdo-Alonso JL, Bundschuh DS, Brose M, Martinez FJ, Rabe KF M2-127 and M2-128 study groups. Roflumilast in moderate-to-severe chronic obstructive pulmonary disease treated with longacting bronchodilators: two randomised clinical trials. Lancet. 2009;374:695–703. doi: 10.1016/S0140-6736(09)61252-6. [DOI] [PubMed] [Google Scholar]
  • 79.Lambert JA, Raju SV, Tang LP, McNicholas CM, Li Y, Courville CW, Faris RF, Coricor GE, Smoot LH, Mazur MM, et al. CFTR activation by roflumilast contributes to therapeutic benefit in chronic bronchitis. Am J Respir Cell Mol Biol. 2014;50:549–558. doi: 10.1165/rcmb.2013-0228OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Tyrrell J, Qian X, Freire J, Tarran R. Roflumilast combined with adenosine increases mucosal hydration in human airway epithelial cultures after cigarette smoke exposure. Am J Physiol Lung Cell Mol Physiol. 2015;308:L1068–L1077. doi: 10.1152/ajplung.00395.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Milara J, Armengot M, Bañuls P, Tenor H, Beume R, Artigues E, Cortijo J. Roflumilast N-oxide, a PDE4 inhibitor, improves cilia motility and ciliated human bronchial epithelial cells compromised by cigarette smoke in vitro. Br J Pharmacol. 2012;166:2243–2262. doi: 10.1111/j.1476-5381.2012.01929.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Rennard SI, Vestbo J. The many “small COPDs”: COPD should be an orphan disease. Chest. 2008;134:623–627. doi: 10.1378/chest.07-3059. [DOI] [PubMed] [Google Scholar]
  • 83.Martinez CH, Kim V, Chen Y, Kazerooni EA, Murray S, Criner GJ, Curtis JL, Regan EA, Wan E, Hersh CP, et al. COPDGene Investigators. The clinical impact of non-obstructive chronic bronchitis in current and former smokers. Respir Med. 2014;108:491–499. doi: 10.1016/j.rmed.2013.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Accurso FJ, Van Goor F, Zha J, Stone AJ, Dong Q, Ordonez CL, Rowe SM, Clancy JP, Konstan MW, Hoch HE, et al. Sweat chloride as a biomarker of CFTR activity: proof of concept and ivacaftor clinical trial data. J Cyst Fibros. 2014;13:139–147. doi: 10.1016/j.jcf.2013.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Raju SV, Tate JH, Peacock SK, Fang P, Oster RA, Dransfield MT, Rowe SM. Impact of heterozygote CFTR mutations in COPD patients with chronic bronchitis. Respir Res. 2014;15:18. doi: 10.1186/1465-9921-15-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Quinton P, Molyneux L, Ip W, Dupuis A, Avolio J, Tullis E, Conrad D, Shamsuddin AK, Durie P, Gonska T. β-adrenergic sweat secretion as a diagnostic test for cystic fibrosis. Am J Respir Crit Care Med. 2012;186:732–739. doi: 10.1164/rccm.201205-0922OC. [DOI] [PubMed] [Google Scholar]
  • 87.Yousef S, Solomon GM, Brody A, Rowe SM, Colin AA. Improved clinical and radiographic outcomes after treatment with ivacaftor in a young adult with cystic fibrosis with the P67L CFTR mutation. Chest. 2015;147:e79–e82. doi: 10.1378/chest.14-1198. [DOI] [PubMed] [Google Scholar]
  • 88.Rowe SM, Heltshe SL, Gonska T, Donaldson SH, Borowitz D, Gelfond D, Sagel SD, Khan U, Mayer-Hamblett N, Van Dalfsen JM, et al. GOAL Investigators of the Cystic Fibrosis Foundation Therapeutics Development Network. Clinical mechanism of the cystic fibrosis transmembrane conductance regulator potentiator ivacaftor in G551D-mediated cystic fibrosis. Am J Respir Crit Care Med. 2014;190:175–184. doi: 10.1164/rccm.201404-0703OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Crawford B, Monz B, Hohlfeld J, Roche N, Rubin B, Magnussen H, Nivens C, Ghafouri M, McDonald J, Tetzlaff K. Development and validation of a cough and sputum assessment questionnaire. Respir Med. 2008;102:1545–1555. doi: 10.1016/j.rmed.2008.06.009. [DOI] [PubMed] [Google Scholar]
  • 90.Leidy NK, Rennard SI, Schmier J, Jones MK, Goldman M. The breathlessness, cough, and sputum scale: the development of empirically based guidelines for interpretation. Chest. 2003;124:2182–2191. doi: 10.1378/chest.124.6.2182. [DOI] [PubMed] [Google Scholar]
  • 91.Leidy NK, Schmier JK, Jones MK, Lloyd J, Rocchiccioli K. Evaluating symptoms in chronic obstructive pulmonary disease: validation of the Breathlessness, Cough and Sputum Scale. Respir Med. 2003;97:S59–S70. [PubMed] [Google Scholar]
  • 92.Durmowicz AG, Witzmann KA, Rosebraugh CJ, Chowdhury BA. Change in sweat chloride as a clinical end point in cystic fibrosis clinical trials: the ivacaftor experience. Chest. 2013;143:14–18. doi: 10.1378/chest.12-1430. [DOI] [PubMed] [Google Scholar]
  • 93.Ferrera L, Caputo A, Galietta LJ. TMEM16A protein: a new identity for Ca(2+)-dependent Cl⁻ channels. Physiology (Bethesda) 2010;25:357–363. doi: 10.1152/physiol.00030.2010. [DOI] [PubMed] [Google Scholar]
  • 94.Marty A, Tan YP, Trautmann A. Three types of calcium-dependent channel in rat lacrimal glands. J Physiol. 1984;357:293–325. doi: 10.1113/jphysiol.1984.sp015501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Frizzell RA, Hanrahan JW. Physiology of epithelial chloride and fluid secretion. Cold Spring Harb Perspect Med. 2012;2:a009563. doi: 10.1101/cshperspect.a009563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Schroeder BC, Cheng T, Jan YN, Jan LY. Expression cloning of TMEM16A as a calcium-activated chloride channel subunit. Cell. 2008;134:1019–1029. doi: 10.1016/j.cell.2008.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Caputo A, Caci E, Ferrera L, Pedemonte N, Barsanti C, Sondo E, Pfeffer U, Ravazzolo R, Zegarra-Moran O, Galietta LJ. TMEM16A, a membrane protein associated with calcium-dependent chloride channel activity. Science. 2008;322:590–594. doi: 10.1126/science.1163518. [DOI] [PubMed] [Google Scholar]
  • 98.Wang Y, Soyombo AA, Shcheynikov N, Zeng W, Dorwart M, Marino CR, Thomas PJ, Muallem S. Slc26a6 regulates CFTR activity in vivo to determine pancreatic duct HCO3- secretion: relevance to cystic fibrosis. EMBO J. 2006;25:5049–5057. doi: 10.1038/sj.emboj.7601387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ousingsawat J, Martins JR, Schreiber R, Rock JR, Harfe BD, Kunzelmann K. Loss of TMEM16A causes a defect in epithelial Ca2+-dependent chloride transport. J Biol Chem. 2009;284:28698–28703. doi: 10.1074/jbc.M109.012120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Rock JR, O’Neal WK, Gabriel SE, Randell SH, Harfe BD, Boucher RC, Grubb BR. Transmembrane protein 16A (TMEM16A) is a Ca2+-regulated Cl- secretory channel in mouse airways. J Biol Chem. 2009;284:14875–14880. doi: 10.1074/jbc.C109.000869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Namkung W, Phuan PW, Verkman AS. TMEM16A inhibitors reveal TMEM16A as a minor component of calcium-activated chloride channel conductance in airway and intestinal epithelial cells. J Biol Chem. 2011;286:2365–2374. doi: 10.1074/jbc.M110.175109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Sondo E, Caci E, Galietta LJ. The TMEM16A chloride channel as an alternative therapeutic target in cystic fibrosis. Int J Biochem Cell Biol. 2014;52:73–76. doi: 10.1016/j.biocel.2014.03.022. [DOI] [PubMed] [Google Scholar]
  • 103.Ruffin M, Voland M, Marie S, Bonora M, Blanchard E, Blouquit-Laye S, Naline E, Puyo P, Le Rouzic P, Guillot L, et al. Anoctamin 1 dysregulation alters bronchial epithelial repair in cystic fibrosis. Biochim Biophys Acta. 2013;1832:2340–2351. doi: 10.1016/j.bbadis.2013.09.012. [DOI] [PubMed] [Google Scholar]
  • 104.Sun F, Hug MJ, Bradbury NA, Frizzell RA. Protein kinase A associates with cystic fibrosis transmembrane conductance regulator via an interaction with ezrin. J Biol Chem. 2000;275:14360–14366. doi: 10.1074/jbc.275.19.14360. [DOI] [PubMed] [Google Scholar]
  • 105.Kanelis V, Hudson RP, Thibodeau PH, Thomas PJ, Forman-Kay JD. NMR evidence for differential phosphorylation-dependent interactions in WT and DeltaF508 CFTR. EMBO J. 2010;29:263–277. doi: 10.1038/emboj.2009.329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Bertrand CA, Zhang R, Pilewski JM, Frizzell RA. SLC26A9 is a constitutively active, CFTR-regulated anion conductance in human bronchial epithelia. J Gen Physiol. 2009;133:421–438. doi: 10.1085/jgp.200810097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.El Khouri E, Touré A. Functional interaction of the cystic fibrosis transmembrane conductance regulator with members of the SLC26 family of anion transporters (SLC26A8 and SLC26A9): physiological and pathophysiological relevance. Int J Biochem Cell Biol. 2014;52:58–67. doi: 10.1016/j.biocel.2014.02.001. [DOI] [PubMed] [Google Scholar]

Articles from Annals of the American Thoracic Society are provided here courtesy of American Thoracic Society

RESOURCES