Next Article in Journal
Prelamin A Accumulation Attenuates Rac1 Activity and Increases the Intrinsic Migrational Persistence of Aged Vascular Smooth Muscle Cells
Previous Article in Journal
Self-Organization of Stem Cell Colonies and of Early Mammalian Embryos: Recent Experiments Shed New Light on the Role of Autonomy vs. External Instructions in Basic Body Plan Development
Previous Article in Special Issue
The Trypanosome Flagellar Pocket Collar and Its Ring Forming Protein—TbBILBO1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Airway Epithelial Cell Cilia and Obstructive Lung Disease

1
Firestone Research Aerosol Laboratory, Fontbonne Bldg. Room F132, Hamilton, ON L8N 4A6, Canada
2
Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
3
St. Joseph’s Healthcare, Firestone Institute for Respiratory Health, 50 Charlton Ave East, FIRH Room T2135, Hamilton, ON L8N 4A6, Canada
*
Author to whom correspondence should be addressed.
Cells 2016, 5(4), 40; https://doi.org/10.3390/cells5040040
Submission received: 1 August 2016 / Revised: 27 October 2016 / Accepted: 7 November 2016 / Published: 11 November 2016
(This article belongs to the Special Issue Cilia and Flagella: Biogenesis and Function)

Abstract

:
Airway epithelium is the first line of defense against exposure of the airway and lung to various inflammatory stimuli. Ciliary beating of airway epithelial cells constitutes an important part of the mucociliary transport apparatus. To be effective in transporting secretions out of the lung, the mucociliary transport apparatus must exhibit a cohesive beating of all ciliated epithelial cells that line the upper and lower respiratory tract. Cilia function can be modulated by exposures to endogenous and exogenous factors and by the viscosity of the mucus lining the epithelium. Cilia function is impaired in lung diseases such as COPD and asthma, and pharmacologic agents can modulate cilia function and mucus viscosity. Cilia beating is reduced in COPD, however, more research is needed to determine the structural-functional regulation of ciliary beating via all signaling pathways and how this might relate to the initiation or progression of obstructive lung diseases. Additionally, genotypes and how these can influence phenotypes and epithelial cell cilia function and structure should be taken into consideration in future investigations.

1. Introduction

The roles of cilia in development and disease are vast [1]. In the lung, cilia are tiny hair-like structures that move mucus and debris up the respiratory escalator. Identification of cilia in relation to lung disease goes back to the early 1900s. Kartagner recognized (1933) the clinical triad of situs inversus, chronic sinusitis, and bronchiectasis now known as Kartagner’s syndrome (KS). KS is inherited via an autosomal recessive pattern with symptoms resulting from defective cilia motility. However, KS is only part of a larger group of disorders known as primary ciliary dyskinesia (PCD) to differentiate them from the acquired types [2,3,4,5,6,7]. Ciliary beating of airway epithelial cells constitutes an important part of the mucociliary transport apparatus [8,9,10]. To be effective in transporting secretions out of the lung, the mucociliary transport apparatus must exhibit a cohesive beating of all ciliated epithelial cells that line the upper and lower respiratory tract [11]. Similar beat frequencies have been observed in nasal, tracheal, and bronchial epithelial cells [8,9,12]. Structure-function studies have helped identify the ciliary targets that participate in regulating ciliary beating, however, our understanding of such regulation is far from complete [3,13,14,15]. Many endogenous and exogenous factors can influence cilia function [16,17,18,19,20,21] with efficient mucociliary transport relying on effective regulation of ciliary beating [11,22]. The ultrastructure of cilia is well documented [23,24]. Yet little is known about the role of cilia in inflammatory and obstructive lung diseases [15] despite correlations between ciliary structure and ciliary function [4,22,25,26,27]. Recent studies of neonates and children with respiratory distress outlined clinical features of neonatal and childhood PCD and their associations with ultrastructural defects and genotype [28,29]. Additionally, links between genetics and ciliary function and structure are demonstrated in several studies [30,31,32,33]. For information on the relationship between genes and cilia structure and function, we refer the readers to recent extensive reviews on this subject [1,3,15,31,34,35]. Another important factor in the function of cilia is the mucus layer composition and thickness [36,37,38]. In this review, we will summarize what we know so far about the respiratory epithelium and specifically airway epithelial cell cilia and their role in inflammatory lung diseases specifically Chronic Obstructive Pulmonary Disease (COPD) and asthma. More research is required to determine the structural-functional regulation of ciliary beating via all signaling pathways and how this might relate to the initiation or progression of lung diseases such as COPD and asthma.

2. Airway Epithelium: A Specialized Physical, Secretory, and Regulatory Barrier

Airway epithelium is the first line of defense against exposure of the airway and lung to inflammatory stimuli and antigens. Epithelial cells form a physical barrier to bacteria and viruses, allergens, dust particles, and air pollutants. In addition, the airway epithelial cells provide an antimicrobial function via production of mucus, immunoglobulins, and defensins. The airway epithelium plays a regulatory role and a pro-inflammatory role through the release of neuropeptide degrading enzymes, endothelin, nitric oxide, TGF-β, arachidonic acid metabolites, and cytokines [22,39,40,41,42,43]. Epithelial cells line the airways form the nasal cavity down to the alveoli. A variety of cells make up the epithelium: the basal cells, which are the stem or progenitor cells for the epithelium and differentiate to form the other cells in injury and repair; the ciliated cells, which provide the mechanism for moving the mucus blanket and have also been reported to be involved in epithelial cell trans-differentiation and repair [44]; the goblet cells, which secrete the mucus; and the brush cells, which are involved in drug metabolism [45]. 50%–80% of epithelial cells lining the airways are ciliated; therefore they are the predominant cell type within the human airways [45] and specifically the smaller airways [46]. This contrasts to rodents where less cilia line the smaller compared to the larger airways [47]. The liquid periciliary layer (PCL) and the mucus layer on top of the PCL layer also line the airway epithelium from the upper regions of the lung to the alveoli, this layer gets thinner as the airways become smaller in diameter until the thickness is <0.1 μm in the alveoli [45]. Therefore, the airway epithelium is a specialized barrier with multiple functions. Any modifications of this barrier or its components can lead to lung disease.
The ciliated airway epithelium: The role of the cilia in traits (inflammation, exacerbation), symptoms (sputum production and cough), and progression of lung disease (relation to disease severity) is not fully understood. Cilia features that may contribute to lung disease include cilia function (ciliary beat frequency-CBF- and wave pattern), the length of the cilia, the ratio of ciliated to non-ciliated areas, cilia structure, and the susceptibility of the cilia to endogenous and exogenous factors that modify their CBF and quality of motion [11,13,18,20,22,24,48,49,50,51,52,53]. Random ciliary orientation has also been reported as a possible cause of respiratory disease [54,55].
Airway epithelial cell cilia are motile cilia, but these cilia have sensory functions and can respond to mechanical and chemical stimuli [56,57,58,59]. In bronchial explants of human respiratory mucosa, ciliary activity was significantly influenced by chemical (pH) and physical properties (viscosity) of the liquid medium in which the cilia beat [60]. In our studies, we demonstrated that tonicity and viscosity of the medium bathing human bronchial cilia can modify CBF [53]. Cilia are 6–7 µm long and 0.2–0.3 µm in diameter. Airway cilia move in a coordinated metachronous beat pattern moving mucus up the airway tree. A range of CBF of 12–16 Hz has been reported in nasal cilia of healthy subjects [61,62,63,64]. We have observed that cilia from nasal epithelial cells of healthy volunteers beat with a frequency of 14.2 ± 1.0 Hz (n = 10, ages 22–52 years) at 37 °C and exhibit the same metachronous beat pattern (laboratory data). Each beat constitutes of an effective and recovery stroke, and a rest phase which influence the amplitude, CBF and beat pattern [65]. Figure 1 shows a high-speed imaging setup for data acquisition of cilia function. Figure 2 illustrates how ciliary function can be evaluated by recording CBF and beat patterns. In terms of structure, cilia are microtubule-based cell organelles extending from the basal body, a centriole at the apical cell surface, containing axonemes. An axoneme is the microtubule cytoskeleton of the cilium, consisting of a ring of nine doublet microtubules surrounding a central pair (9+2). Inner and outer dynein arms (Figure 3) of each microtubule doublet generate the force needed for motility in an ATP-dependent process [24,65]. Inner dynein arms or radial spoke defects correlate with problems in ciliary bending, and outer dynein arms defects correlate with a decline in CBF, while the absence of the central microtubule pair has been correlated with a circular beat pattern [4,7,22,25,26,27]. The cilia are dynamic organelles. Functional and structural components of cilia are synthesized in the cytoplasm and these are transported up and down the cilium by a specialized system known as the intraflagellar transport system. Many ciliary proteins have been identified, yet their exact role in the cell remains unknown [14,66,67,68]. Cilia membranes are contiguous with the plasma membrane of the cell and contain specific receptors and ion channel proteins that initiate signalling pathways controlling motility, mechanical or chemical stimuli [13,24,56]. CBF in airway epithelial cells depends on the cyclic nucleotides cAMP and cGMP and intracellular calcium concentration [69,70,71,72]. In human airway epithelia, sAC is localized to motile airway cilia and it contributes to the regulation of human airway CBF [73]. Changes in epithelial cell intracellular pH [74] and intracellular bicarbonate are additional factors that might influence CBF through direct effects on dynein arms. The structural-functional regulation of CBF via many signalling pathways has been reviewed [13], yet a lot of research is required to determine how this regulation might influence the initiation or progression of obstructive lung diseases.

3. What Modulates Airway Cilia Function?

Ciliary beat frequency (CBF) and wave pattern are important measures of epithelial cell function and these can be modified by various exogenous and endogenous mediators. In 1981, Rossman and co-investigators observed that some patients with KS had motile cilia with abnormal ciliary structure and uncoordinated beating and used the term PCD which is now used to describe all congenital cilia disorders [4,7]. However, when structural cilia defects are not identified, cilia function can still be impaired by various exposures that can affect cilia function directly or can modify the secretions and viscosity of the mucus layer which is transported up the respiratory escalator by cilia [20,38,52,53,75].
  • The effect of exogenous and endogenous mediators on CBF: Airway cilia function can be modified by various endogenous and exogenous exposures (Figure 4) including infections such as rhinitis, rhinosinusitis [6,76,77], and pneumonia [78]; environmental pollutants [79,80,81]; cigarette smoke and oxidative stress [82,83,84,85], and the rheology of the mucus secretions [75,86,87]. Numerous agents and drugs (Table 1) have been shown to modulate CBF [16,17] with efficient mucociliary transport relying on effective regulation of ciliary beating [11,22]. In vitro, beta-adrenergic and cholinergic agents were demonstrated to stimulate CBF, while fluticasone, some preservatives, anaesthetics, and some bacterial toxins inhibited CBF [16,17,18,19]. In addition, the effect of hyperosmolar agents on the muco-ciliary transport apparatus has been documented [38,75]. In a study of the direct effect of mannitol on the CBF of human bronchial epithelial cells (Figure 5), we demonstrated a direct osmolarity-independent cilio-stimulatory effect, a unique mechanism of action for mannitol, compared to dextran and saline, on ciliary beating [53]. A systematic review of the literature that examines the effect of 229 drugs and other substances on CBF of airway epithelial cell cilia has been published [20].
  • Relationship of inflammation to reduced or variable CBF: earlier studies in patients with cystic fibrosis have demonstrated a link between airway inflammation and a reduction in CBF [88,89]. Similar links between inflammation and the impaired mucociliary transport rate were investigated with sputum collected from COPD patients [90], and in ciliated nasal epithelial cells from stable bronchiectatic subjects [91]. However, the effects of inflammation on CBF are not always direct and can reflect modifications of epithelial differentiation and proliferation, possibly related to local chronic inflammation. This occurred in nasal airway cells from Rhinitis patients indicating a possible effect of inflammation on epithelial cell integrity and the number of cilia [92]. In addition, a correlation between ciliary activity and ciliary ultrastructure of the nasal mucosa in chronic and recurrent sinusitis was demonstrated [93] and leukotrienes released during inflammation could slow down CBF [94,95]. Bronchial cilia abnormalities worsened with the disease possibly contributing to the impairment of the mucociliary clearance in chronic bronchitis patients [96]. Also infections such as P. aeruginosa and H. influenzae caused a significant decrease in CBF of human nasal cilia, while staphylococcal products did not [97]. In comparison, RSV infection of human bronchial epithelial cells in culture resulted in ciliostasis and loss of cilia from the cell surface [98]. Resolution of infection and/or inflammation could restore normal cilia function several weeks following upper respiratory viral infection [99] or acute bronchiolitis in infancy [100].
  • Nasal ciliary changes can reflect bronchial changes: Respiratory cilia from different locations have been studied. Correlation between nasal, tracheal, and bronchial CBF and between mucociliary clearance and CBF were demonstrated [9,61,101,102,103]. In addition, nasal and bronchial cilia were shown to be comparable in function and structure allowing the use of nasal epithelial cells as surrogates of bronchial epithelial cells in airway inflammation studies [12]. Patients with allergic rhinitis and asthma have stronger nasal responsiveness to cold, dry air compared with patients with rhinitis alone, indicating that upper airway responses could represent lower airway responsiveness [104]. However, the use of nasal cilia as representative of tracheal and bronchial cilia for study of respiratory diseases such as asthma or COPD should be done with caution and preferably in the absence of rhinitis and runny nose due to allergies or active respiratory infections as these directly influence the differentiation and proliferation of nasal cilia and could negate their use as surrogates of tracheal and bronchial cilia. Evidence for this restriction has been demonstrated when intermittent allergic rhinitis was associated with decreased expression of anti-inflammatory genes in nasal fluids obtained from intermittent allergic rhinitis patients [105]. Also, when compared to controls, rhinitis patients exhibited lower percentages of ciliated cells and higher percentages of goblet and basal cells in nasal epithelial brushings related to local chronic inflammation [92]. In addition, respiratory viruses lead to ciliary defects in nasal epithelium of children with recovery occurring within 2–10 weeks after infection [99] and 13–17 weeks following acute bronchiolitis in infancy [100]. All these studies stress the need to study ciliated epithelium specimens in the absence of active respiratory infections.

4. Pathophysiology of COPD

COPD is a leading cause of morbidity and mortality worldwide, and results in a vast economic and social burden that continues to increase. Prevalence and morbidity data greatly underestimate the total burden of COPD because the disease is usually not diagnosed until it is advanced [109]. COPD is a respiratory disorder characterized by progressive, non-reversible airflow limitation, associated with a chronic inflammatory response of the lung to harmful environmental agents (e.g., tobacco smoke, fumes). Inflammatory cells infiltrate the surface epithelium of large airways, contributing to two characteristic symptoms of COPD: cough and sputum production. Persistent inflammation in the small and large airways, and lung parenchyma and its vasculature occurs in COPD patients. The most prominent symptom of COPD is dyspnea, which is largely caused by hyperinflation of the lungs as a result of small airway collapse due to emphysema and narrowing due to fibrosis, so that the alveoli are not able to empty. Hyperinflation reduces exercise tolerance leading to immobility and poor health status [85,109,110,111,112]. Airway inflammation is present even in early disease and persists long after the stimulus (cigarette smoke, pollutants, and noxious gases) is withdrawn [113,114]. In COPD, cilia can be overwhelmed with secretions or they could be abnormal in terms of structure, but we can readily investigate ciliary function. Nasal brushings from COPD patients and healthy subjects are non-invasive and provide a quick method to study ciliary function and structure ex vivo. Brushings can be obtained from the right or left nasal turbinate and can be stored for up to seven days without any significant change in CBF measurements (Figure 6) [49].

5. Factors That Can Affect CBF in COPD

Hurst et al. reported a correlation between the degree of upper and lower airway inflammation in COPD highlighting the significance of studying ciliated nasal epithelial cells in this disease [115]. In addition to inflammation, another process thought to be important in the pathogenesis of COPD is an imbalance of proteinases and antiproteinases in the lung [43]. Roles for defensins, matrix metalloproteinases, and inflammatory mediators have been implicated in COPD [109,116,117,118,119]. Exhaled leukotrienes and prostaglandins are also elevated in COPD [120]. Physiological changes characteristic of the disease include mucus hypersecretion and ciliary dysfunction. To be effective in clearing secretions out of the lung, the mucociliary transport apparatus must exhibit coordinated beating of all ciliated epithelial cells that line the upper and lower respiratory tract. CBF and wave pattern, biologically important measures of epithelial cell function, may be altered during an exacerbation, leading to accumulation of secretions and impairment of beating. We speculate that this impairment may take the form of cilia unable to initiate the effector or return stroke of the beat cycle due to increased mucus viscosity or ineffective transport of mucus due to an uncoupling of the cilia tips from the increased depth of the secretion layer. When CBF and wave pattern are abnormal, electron micrographs of cilia ultrastructure may provide useful functional clues in COPD as was demonstrated for PCD and other respiratory diseases [4,64,121]. Differences in CBF have been documented in diverse patient populations [7,122] and numerous drugs and excipients have been shown to affect CBF and mucociliary clearance (MCC) [16,107,108]. As mentioned earlier in this review, beta-adrenergic and cholinergic agents stimulate CBF, while fluticasone, some preservatives, and some bacterial toxins inhibit CBF [16,17,18,19,21,106,107]. However, we do not know the effect of exacerbations on CBF in epithelial cells of COPD patients. Structure-function studies have helped identify the ciliary targets that participate in regulating ciliary beating; however, our understanding of such regulation is far from complete [13]. The ultrastructure of cilia is known [23,24] and correlations between the structure and function of cilia [4,22,25,26,27] have been documented. However, more research is needed to investigate the role of cilia in COPD. An earlier study of nasal CBF in stable COPD found that baseline CBF was reduced by 25% compared to healthy controls and that salmeterol produced a significant increase in CBF [78]. We recently found that CBF of nasal epithelial cell cilia is depressed in subjects with moderate and severe COPD (Figure 7) and that salmeterol could stimulate CBF in cilia from these subjects [52]. Additionally, we demonstrated that pharmacologic agents with various mechanisms of action could modulate CBF of nasal cilia obtained from COPD patients and that a thorough investigation of the various signaling pathways involved in cilia function is still needed [52]. Here we summarize the possible signaling pathways that could be involved in cilia function based on the literature and what we found in nasal cilia from COPD patients (Figure 8). Please note that this is only a suggested starting point for investigating cilia function in COPD. More investigations are needed to fully understand the regulation of cilia function in COPD.
  • Effect of smoking: Mucociliary clearance has been demonstrated to be affected in smoking [123,124] and while smoke caused cilia loss in bovine lungs studied in vitro [125], ciliary function has been shown to be normal in smoking asymptomatic subjects [64]. When investigating the ultrastructure (TEM) of bronchial epithelial cell cilia, significant ciliary abnormalities in smokers and ex-smokers compared to non-smokers and controls have been observed with the higher percentage of non-specific ciliary abnormalities associated with chronic tobacco smoke in those with chronic sputum production [126], a characteristic symptom of COPD [109,127]. Earlier studies into the effect of smoking on cilia abundance have yielded conflicting results. Rankin et al. (2007) observed a smoking-related difference in cilia abundance in explanted human bronchial tissue [128] similar to that previously described in bronchial [129] and tracheal explants [130]. Other studies, into cilia abnormalities in asymptomatic smokers and nonsmokers, have yielded conflicting results with no difference between asymptomatic smokers and nonsmokers observed in nasal brushings [7,55]. In contrast, Rutland et al. (1983) and Fox et al. (1983), studied cilia ultrastructure in nasal brushings and reported significantly more abnormal cilia in smokers than in non-smokers [131,132]. In a study of bronchial epithelium from normal smokers and matched nonsmokers, the small airway samples (10th to 12th order) had a higher proportion of ciliated cells compared to samples from large (second to third order) bronchi, and smoking caused a decline in the proportion of ciliated cells at both sites [46]. Another study of bronchial biopsies suggested that smoking-associated shorter airway epithelial cilia could play a role in the pathogenesis of smoking-induced lung disease [48]. Cigarette smoke exposure of C57BL/6 mice for 6–12 month was associated with slowed ciliary motility, decreased ciliated cell numbers, and an impaired ciliary response to beta-agonists [133]. In recent studies, exposure of the human epithelium in vitro to cigarette smoke extracts resulted in a broad suppression of genes involved in ciliogenesis [134], and evaluations of cilia lengths from bronchial biopsies or brushings of healthy smokers and nonsmokers showed that smoking is associated with shorter cilia [48,135]. In the large and small airway epithelium, cilia were significantly shorter in COPD smokers compared to healthy smokers and nonsmokers. However, when investigating the gene expression data of the intraflagellar transport system of cilia (which regulates cilia length), some IFT genes were downregulated in smokers but not in COPD smokers compared to healthy smokers [135]. In our COPD study, nasal CBF was significantly depressed in cilia obtained from moderate and severe COPD subjects compared to At Risk and Control subjects (Figure 7). This significant decline in CBF was not due to a difference in the quality of samples (abundance of cilia), or to cell size, cilia length, or cilia coordination. In addition, since At Risk, and COPD (Moderate, Severe) subjects were smokers/ex-smokers with minimum of 32 pack years, the reduced CBF in COPD cilia only could not be attributed just to the effect of cigarette smoke [52]. In addition, various agents could modulate in vitro cilia function of COPD patients confirming a complicated role for cilia in COPD [52] (Figure 8). We also demonstrated that mannitol has a direct effect on CBF of bronchial cilia [53]. Additionally, airway surface properties (hydration, viscosity) can modulate cilia function in vivo as demonstrated in studies of inhaled mannitol in lung disease patients [38,136,137]. This indicates that further studies of the function and ultrastructure of cilia in COPD are needed.

6. Factors That Can Affect CBF in Asthma

When investigating the ultrastructure of bronchial mucosa in lung biopsies of children with asthma, the luminal surfaces of ciliated cells showed cytoplasmic blebs and abnormal cilia [138]. Application of Sputum from asthma patients to frogs’ isolated palates to determine the effect on CBF resulted in ciliostasis which disappeared with clinical improvement [139,140]. In comparison, samples obtained from normal volunteers and from most patients, excluding those with asthma, had no effect on ciliary beating. The inhibitory effect on CBF was independent of medications used and was shown in atopic and intrinsic types of asthmatic patients. In addition, the effect was not pH dependent or related to the degree of eosinophilia [140]. Additionally, tracheal mucus velocity (TMV) was significantly less (6.3 ± 2.3 mm per min, mean ± SD, n = 6) in asymptomatic asthmatic patients than in normal subjects (11.6 ± 3.6 mm per min, n = 7) [141], and tracheal mucociliary transport rates were decreased in patients with allergic asthma [142,143]. Mucociliary function in the airways of patients with bronchial asthma showed abnormalities [144]. Additionally, inhalation of hypertonic saline aerosol or dry-powder mannitol increased mucociliary clearance in asthmatics [145,146], and inhaled steroids restored most of the ciliated surfaces of bronchial epithelial cells [147,148,149]. Therefore, structural and functional ciliary abnormalities have been observed in asthma. Further investigation of cilia structure and function in asthma is needed.

7. Summary and Conclusions

COPD is a respiratory disease characterized by airflow limitation that is not fully reversible, is usually progressive, and associated with an abnormal inflammatory response of the lungs to noxious particles or gases. Cigarette smoke is the primary risk factor for COPD. Patients with a smoking history presenting with cough and sputum production, exertional dyspnea, and frequent respiratory infections are likely to be diagnosed with COPD. Exacerbations (whether infectious -bacterial/viral- or non-infectious—second hand smoke/pollution) contribute to the progression of disease severity in COPD. Repeated exacerbations may result in repeated injuries to the airway epithelium and if frequent, could shorten the time available for epithelial repair, resulting in further damage to the integrity of the epithelium due to ongoing inflammation. Inflammation due to exacerbation could alter CBF through mediator release from the inflammatory cells and/or the epithelium or by contributing to cilia damage, epithelial cell loss, and damage to the basement membrane. In addition, the ciliated epithelial cells can spread and transdifferentiate into distinct epithelial cell types to repair the airway epithelium after injury, and this process could be modified in COPD and asthma. Ciliary beating of epithelial cells constitutes an important part of the mucociliary transport apparatus. We demonstrated a significant decrease in CBF of COPD patients classified in the GOLD 2–4 categories implying that impaired ciliary function can impact mucociliary clearance in COPD, potentially contributing to retention of secretions and infection. Pharmacologic agents with different mechanisms of action can similarly increase CBF of nasal cilia of COPD subjects compared to healthy subjects. Further investigation of the signaling pathways involved is needed. Additionally, the acute and prolonged effects of mannitol on CBF of HBEC suggest a unique mechanism of action for mannitol on ciliary function. Continued investigation of the function and structure of airway epithelial cilia in COPD could lead to a better understanding of the mechanisms of disease and to improved therapies. The investigation of epithelial repair and the role of cilia abnormalities in the initiation and progression of COPD is still needed. Specifically, aspects of ciliary function and structure that may affect COPD, namely CBF, coordinated ciliary beating, the ratio of ciliated to non-ciliated areas, and the susceptibility of the cilia to intrinsic and extrinsic agents that modify their rate and quality of motion should be further investigated. Additionally, fewer investigations on the above parameters have been published in patients with asthma including the effects of chronic treatments (e.g., steroids) on ciliary function and structure. Finally, genotypes and how these can influence phenotypes and epithelial cell cilia function and structure should be taken into consideration in future investigations. We believe that the use of models of human airway mucosa is very much needed in respiratory research and will generate answers relevant to the role of cilia in human lung diseases such as COPD and asthma. Use of primary human cells in these models is essential since these cells will mimic more closely the in vivo conditions and allow further investigation of epithelial cell cilia in lung disease.

Acknowledgments

The authors did not receive funds for covering the costs to publish in open access.

Author Contributions

A.Y. and M.D. conceived and designed the review; A.Y. wrote first draft; M.D. contributed references and reviewed the document.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Powles-Glover, N. Cilia and ciliopathies: Classic examples linking phenotype and genotype-an overview. Reprod. Toxicol. 2014, 48, 98–105. [Google Scholar] [CrossRef] [PubMed]
  2. Werner, C.; Onnebrink, J.G.; Omran, H. Diagnosis and management of primary ciliary dyskinesia. Cilia 2015, 4, 2. [Google Scholar] [CrossRef] [PubMed]
  3. Knowles, M.R.; Daniels, L.A.; Davis, S.D.; Zariwala, M.A.; Leigh, M.W. Primary ciliary dyskinesia. Recent advances in diagnostics, genetics, and characterization of clinical disease. Am. J. Respir. Crit. Care Med. 2013, 188, 913–922. [Google Scholar] [CrossRef] [PubMed]
  4. Rossman, C.M.; Forrest, J.B.; Lee, R.M.; Newhouse, A.F.; Newhouse, M.T. The dyskinetic cilia syndrome; abnormal ciliary motility in association with abnormal ciliary ultrastructure. Chest 1981, 80 (Suppl. S6), 860–865. [Google Scholar] [PubMed]
  5. Leigh, M.W.; Pittman, J.E.; Carson, J.L.; Ferkol, T.W.; Dell, S.D.; Davis, S.D.; Knowles, M.R.; Zariwala, M.A. Clinical and genetic aspects of primary ciliary dyskinesia/Kartagener syndrome. Genet. Med. 2009, 11, 473–487. [Google Scholar] [CrossRef] [PubMed]
  6. Gudis, D.; Zhao, K.Q.; Cohen, N.A. Acquired cilia dysfunction in chronic rhinosinusitis. Am. J. Rhinol. Allergy 2012, 26, 1–6. [Google Scholar] [CrossRef] [PubMed]
  7. Rossman, C.M.; Lee, R.M.; Forrest, J.B.; Newhouse, M.T. Nasal cilia in normal man, primary ciliary dyskinesia and other respiratory diseases: Analysis of motility and ultrastructure. Eur. J. Respir. Dis. Suppl. 1983, 127, 64–70. [Google Scholar] [PubMed]
  8. Rutland, J.; Griffin, W.M.; Cole, P.J. Human ciliary beat frequency in epithelium from intrathoracic and extrathoracic airways. Am. Rev. Respir. Dis. 1982, 125, 100–105. [Google Scholar] [PubMed]
  9. Yager, J.A.; Ellman, H.; Dulfano, M.J. Human ciliary beat frequency at three levels of the tracheobronchial tree. Am. Rev. Respir. Dis. 1980, 121, 661–665. [Google Scholar] [PubMed]
  10. Wanner, A.; Salathe, M.; O’Riordan, T.G. Mucociliary clearance in the airways. Am. J. Respir. Crit. Care Med. 1996, 154 Pt 1, 1868–1902. [Google Scholar] [CrossRef] [PubMed]
  11. Braiman, A.; Priel, Z. Efficient mucociliary transport relies on efficient regulation of ciliary beating. Respir. Physiol. Neurobiol. 2008, 163, 202–207. [Google Scholar] [CrossRef] [PubMed]
  12. McDougall, C.M.; Blaylock, M.G.; Douglas, J.G.; Brooker, R.J.; Helms, P.J.; Walsh, G.M. Nasal Epithelial Cells as Surrogates of Bronchial Epithelial Cells in Airway Inflammation Studies. Am. J. Respir. Cell Mol. Biol. 2008, 39, 560–568. [Google Scholar] [CrossRef] [PubMed]
  13. Salathe, M. Regulation of mammalian ciliary beating. Annu. Rev. Physiol. 2007, 69, 401–422. [Google Scholar] [CrossRef] [PubMed]
  14. Ishikawa, H.; Marshall, W.F. Mechanobiology of Ciliogenesis. BioScience 2014, 64, 1084–1091. [Google Scholar] [CrossRef] [PubMed]
  15. Tilley, A.E.; Walters, M.S.; Shaykhiev, R.; Crystal, R.G. Cilia dysfunction in lung disease. Annu. Rev. Physiol. 2015, 77, 379–406. [Google Scholar] [CrossRef] [PubMed]
  16. Merkus, P.; Romeijn, S.G.; Verhoef, J.C.; Merkus, F.W.; Schouwenburg, P.F. Classification of cilio-inhibiting effects of nasal drugs. Laryngoscope 2001, 111 Pt 1, 595–602. [Google Scholar] [CrossRef] [PubMed]
  17. Rusznak, C.; Devalia, J.L.; Lozewicz, S.; Davies, R.J. The assessment of nasal mucociliary clearance and the effect of drugs. Respir. Med. 1994, 88, 89–101. [Google Scholar] [CrossRef]
  18. Dowling, R.B.; Wilson, R. Bacterial toxins which perturb ciliary function and respiratory epithelium. Symp. Ser. Soc. Appl. Microbiol. 1998, 27, 138S–148S. [Google Scholar] [CrossRef] [PubMed]
  19. Hofmann, T.; Gugatschga, M.; Koidl, B.; Wolf, G. Influence of preservatives and topical steroids on ciliary beat frequency in vitro. Arch. Otolaryngol. Head Neck Surg. 2004, 130, 440–445. [Google Scholar] [CrossRef] [PubMed]
  20. Workman, A.D.; Cohen, N.A. The effect of drugs and other compounds on the ciliary beat frequency of human respiratory epithelium. Am. J. Rhinol. Allergy 2014, 28, 454–464. [Google Scholar] [CrossRef] [PubMed]
  21. Salathe, M. Effects of beta-agonists on airway epithelial cells. J. Allergy Clin. Immunol. 2002, 110 (Suppl. S6), S275–S281. [Google Scholar] [CrossRef] [PubMed]
  22. Stannard, W.; O’Callaghan, C. Ciliary function and the role of cilia in clearance. J. Aerosol Med. 2006, 19, 110–115. [Google Scholar] [CrossRef] [PubMed]
  23. Marshall, W.F.; Nonaka, S. Cilia: Tuning in to the cell’s antenna. Curr. Biol. 2006, 16, R604–R614. [Google Scholar] [CrossRef] [PubMed]
  24. Satir, P.; Christensen, S.T. Overview of structure and function of mammalian cilia. Annu. Rev. Physiol 2007, 69, 377–400. [Google Scholar] [CrossRef] [PubMed]
  25. Chilvers, M.A.; Rutman, A.; O’Callaghan, C. Ciliary beat pattern is associated with specific ultrastructural defects in primary ciliary dyskinesia. J. Allergy Clin. Immunol. 2003, 112, 518–524. [Google Scholar] [CrossRef]
  26. Willems, T.; Jorissen, M. Correlations between ciliary structure and ciliary function. Acta Otorhinolaryngol. Belg. 2000, 54, 299–308. [Google Scholar] [PubMed]
  27. Jorissen, M. Correlations among mucociliary transport, ciliary function, and ciliary structure. Am. J. Rhinol. 1998, 12, 53–58. [Google Scholar] [CrossRef] [PubMed]
  28. Davis, S.D.; Ferkol, T.W.; Rosenfeld, M.; Lee, H.S.; Dell, S.D.; Sagel, S.D.; Milla, C.; Zariwala, M.A.; Pittman, J.E.; Shapiro, A.J.; et al. Clinical features of childhood primary ciliary dyskinesia by genotype and ultrastructural phenotype. Am. J. Respir. Crit. Care Med. 2015, 191, 316–324. [Google Scholar] [CrossRef] [PubMed]
  29. Mullowney, T.; Manson, D.; Kim, R.; Stephens, D.; Shah, V.; Dell, S. Primary ciliary dyskinesia and neonatal respiratory distress. Pediatrics 2014, 134, 1160–1166. [Google Scholar] [CrossRef] [PubMed]
  30. Knowles, M.R.; Ostrowski, L.E.; Loges, N.T.; Hurd, T.; Leigh, M.W.; Huang, L.; Wolf, W.E.; Carson, J.L.; Hazucha, M.J.; Yin, W.; et al. Mutations in SPAG1 cause primary ciliary dyskinesia associated with defective outer and inner dynein arms. Am. J. Hum. Genet. 2013, 93, 711–720. [Google Scholar] [CrossRef] [PubMed]
  31. Kim, R.H.; Hall, A.; Cutz, E.; Knowles, M.R.; Nelligan, K.A.; Nykamp, K.; Zariwala, M.A.; Dell, S.D. The role of molecular genetic analysis in the diagnosis of primary ciliary dyskinesia. Ann. Am. Thorac. Soc. 2014, 11, 351–359. [Google Scholar] [CrossRef] [PubMed]
  32. Knowles, M.R.; Ostrowski, L.E.; Leigh, M.W.; Sears, P.R.; Davis, S.D.; Wolf, W.E.; Hazucha, M.J.; Carson, J.L.; Olivier, K.N.; Sagel, S.D.; et al. Mutations in RSPH1 cause primary ciliary dyskinesia with a unique clinical and ciliary phenotype. Am. J. Respir. Crit. Care Med. 2014, 189, 707–717. [Google Scholar] [CrossRef] [PubMed]
  33. Shapiro, A.J.; Davis, S.D.; Ferkol, T.; Dell, S.D.; Rosenfeld, M.; Olivier, K.N.; Sagel, S.D.; Milla, C.; Zariwala, M.A.; Wolf, W.; et al. Laterality defects other than situs inversus totalis in primary ciliary dyskinesia: Insights into situs ambiguus and heterotaxy. Chest 2014, 146, 1176–1186. [Google Scholar] [CrossRef] [PubMed]
  34. Fretzayas, A.; Moustaki, M. Clinical spectrum of primary ciliary dyskinesia in childhood. World J. Clin. Pediatr. 2016, 5, 57–62. [Google Scholar] [CrossRef] [PubMed]
  35. Milla, C.E. The evolving spectrum of ciliopathies and respiratory disease. Curr. Opin. Pediatr. 2016, 28, 339–347. [Google Scholar] [CrossRef] [PubMed]
  36. Hill, D.B.; Vasquez, P.A.; Mellnik, J.; McKinley, S.A.; Vose, A.; Mu, F.; Henderson, A.G.; Donaldson, S.H.; Alexis, N.E.; Boucher, R.C.; et al. A biophysical basis for mucus solids concentration as a candidate biomarker for airways disease. PLoS ONE 2014, 9, e87681. [Google Scholar] [CrossRef] [PubMed]
  37. Button, B.; Boucher, R.C. Role of mechanical stress in regulating airway surface hydration and mucus clearance rates. Respir. Physiol. Neurobiol. 2008, 163, 189–201. [Google Scholar] [CrossRef] [PubMed]
  38. Daviskas, E.; Rubin, B.K. Effect of inhaled dry powder mannitol on mucus and its clearance. Expert. Rev. Respir. Med. 2013, 7, 65–75. [Google Scholar] [CrossRef] [PubMed]
  39. Bals, R.; Hiemstra, P.S. Innate immunity in the lung: How epithelial cells fight against respiratory pathogens. Eur. Respir. J. 2004, 23, 327–333. [Google Scholar] [CrossRef] [PubMed]
  40. Hamilton, L.M.; Davies, D.E.; Wilson, S.J.; Kimber, I.; Dearman, R.J.; Holgate, S.T. The bronchial epithelium in asthma--much more than a passive barrier. Monaldi Arch. Chest Dis. 2001, 56, 48–54. [Google Scholar] [PubMed]
  41. Hermans, C.; Bernard, A. Lung epithelium-specific proteins: Characteristics and potential applications as markers. Am. J. Respir. Crit. Care Med. 1999, 159, 646–678. [Google Scholar] [CrossRef] [PubMed]
  42. Mills, P.R.; Davies, R.J.; Devalia, J.L. Airway epithelial cells, cytokines, and pollutants. Am. J. Respir. Crit. Care Med. 1999, 160 (Suppl. S1), S38–S43. [Google Scholar] [CrossRef] [PubMed]
  43. Hiemstra, P.S.; van Wetering, S.; Stolk, J. Neutrophil serine proteinases and defensins in chronic obstructive pulmonary disease: Effects on pulmonary epithelium. Eur. Respir. J. 1998, 12, 1200–1208. [Google Scholar] [CrossRef] [PubMed]
  44. Park, K.S.; Wells, J.M.; Zorn, A.M.; Wert, S.E.; Laubach, V.E.; Fernandez, L.G.; Whitsett, J.A. Transdifferentiation of ciliated cells during repair of the respiratory epithelium. Am. J. Respir. Cell Mol. Biol. 2006, 34, 151–157. [Google Scholar] [CrossRef] [PubMed]
  45. Patton, J.S.; Byron, P.R. Inhaling medicines: Delivering drugs to the body through the lungs. Nat. Rev. Drug Discov. 2007, 6, 67–74. [Google Scholar] [CrossRef] [PubMed]
  46. Harvey, B.G.; Heguy, A.; Leopold, P.L.; Carolan, B.J.; Ferris, B.; Crystal, R.G. Modification of gene expression of the small airway epithelium in response to cigarette smoking. J. Mol. Med. 2007, 85, 39–53. [Google Scholar] [CrossRef] [PubMed]
  47. Rawlins, E.L.; Hogan, B.L. Ciliated epithelial cell lifespan in the mouse trachea and lung. Am. J. Physiol. Lung Cell. Mol. Physiol. 2008, 295, L231–L234. [Google Scholar] [CrossRef] [PubMed]
  48. Leopold, P.L.; O’Mahony, M.J.; Lian, X.J.; Tilley, A.E.; Harvey, B.G.; Crystal, R.G. Smoking is associated with shortened airway cilia. PLoS ONE 2009, 4, e8157. [Google Scholar] [CrossRef] [PubMed]
  49. Yaghi, A.; Dolovich, M. Investigating the Function of Epithelial Cell Cilia Using High Speed Digital Imaging. Am. J. Respir. Crit. Care Med. 2008, 177, A206. [Google Scholar]
  50. Iida, H.; Matsuura, S.; Shirakami, G.; Tanimoto, K.; Fukuda, K. Differential effects of intravenous anesthetics on ciliary motility in cultured rat tracheal epithelial cells. Can. J. Anaesth. 2006, 53, 242–249. [Google Scholar] [CrossRef] [PubMed]
  51. Chilvers, M.A.; O’Callaghan, C. Local mucociliary defence mechanisms. Paediatr. Respir. Rev. 2000, 1, 27–34. [Google Scholar] [CrossRef] [PubMed]
  52. Yaghi, A.; Zaman, A.; Cox, G.; Dolovich, M.B. Ciliary beating is depressed in nasal cilia from chronic obstructive pulmonary disease subjects. Respir. Med. 2012, 106, 1139–1147. [Google Scholar] [CrossRef] [PubMed]
  53. Yaghi, A.; Zaman, A.; Dolovich, M.B. The direct effect of hyperosmolar agents on ciliary beating of human bronchial epithelial cells. J. Aerosol Med. Pulm. Drug Deliv. 2012, 25, 88–95. [Google Scholar] [CrossRef] [PubMed]
  54. Rutland, J.; de Iongh, R.U. Random ciliary orientation. A cause of respiratory tract disease. N. Engl. J. Med. 1990, 323, 1681–1684. [Google Scholar] [CrossRef] [PubMed]
  55. De Iongh, R.U.; Rutland, J. Ciliary defects in healthy subjects, bronchiectasis, and primary ciliary dyskinesia. Am. J. Respir. Crit. Care Med. 1995, 151, 1559–1567. [Google Scholar] [CrossRef] [PubMed]
  56. Shah, A.S.; Ben-Shahar, Y.; Moninger, T.O.; Kline, J.N.; Welsh, M.J. Motile Cilia of Human Airway Epithelia Are Chemosensory. Science 2009, 325, 1131–1134. [Google Scholar] [CrossRef] [PubMed]
  57. Horvath, G.; Sorscher, E.J. Luminal fluid tonicity regulates airway ciliary beating by altering membrane stretch and intracellular calcium. Cell Motil. Cytoskeleton 2008, 65, 469–475. [Google Scholar] [CrossRef] [PubMed]
  58. Kanthakumar, K.; Taylor, G.W.; Cundell, D.R.; Dowling, R.B.; Johnson, M.; Cole, P.J.; Wilson, R. The effect of bacterial toxins on levels of intracellular adenosine nucleotides and human ciliary beat frequency. Pulm. Pharmacol. 1996, 9, 223–230. [Google Scholar] [CrossRef] [PubMed]
  59. Agius, A.M.; Wake, M.; Pahor, A.L.; Smallman, A. The effects of in vitro cotitine on nasal ciliary beat frequency. Clin. Otolaryngol. Allied Sci. 1995, 20, 465–469. [Google Scholar] [CrossRef] [PubMed]
  60. Luk, C.K.; Dulfano, M.J. Effect of pH, viscosity and ionic-strength changes on ciliary beating frequency of human bronchial explants. Clin. Sci. (Lond.) 1983, 64, 449–451. [Google Scholar] [CrossRef] [PubMed]
  61. Roth, Y.; Aharonson, E.F.; Teichtahl, H.; Baum, G.L.; Priel, Z.; Modan, M. Human in vitro nasal and tracheal ciliary beat frequencies: Comparison of sampling sites, combined effect of medication, and demographic relationships. Ann. Otol. Rhinol. Laryngol. 1991, 100 Pt 1, 378–384. [Google Scholar] [CrossRef] [PubMed]
  62. Yager, J.; Chen, T.M.; Dulfano, M.J. Measurement of frequency of ciliary beats of human respiratory epithelium. Chest 1978, 73, 627–633. [Google Scholar] [CrossRef] [PubMed]
  63. Chilvers, M.A.; Rutman, A.; O’Callaghan, C. Functional analysis of cilia and ciliated epithelial ultrastructure in healthy children and young adults. Thorax 2003, 58, 333–338. [Google Scholar] [CrossRef] [PubMed]
  64. Rossman, C.M.; Lee, R.M.; Forrest, J.B.; Newhouse, M.T. Nasal ciliary ultrastructure and function in patients with primary ciliary dyskinesia compared with that in normal subjects and in subjects with various respiratory diseases. Am. Rev. Respir. Dis. 1984, 129, 161–167. [Google Scholar] [PubMed]
  65. Satir, P.; Dirksen, E.R. Function-structure correlations in cilia from mammalian respiratory tract. In The Respiratory System Volume 1: Circulation and Nonrespiratory Functions; Weibel, E.R., Ed.; The American Physiological Society: Bethesda, MD, USA, 1985; pp. 473–494. [Google Scholar]
  66. Pazour, G.J.; Rosenbaum, J.L. Intraflagellar transport and cilia-dependent diseases. Trends Cell Biol. 2002, 12, 551–555. [Google Scholar] [CrossRef]
  67. Rosenbaum, J.L.; Witman, G.B. Intraflagellar transport. Nat. Rev. Mol. Cell Biol. 2002, 3, 813–825. [Google Scholar] [CrossRef] [PubMed]
  68. Badano, J.L.; Mitsuma, N.; Beales, P.L.; Katsanis, N. The Ciliopathies: An Emerging Class of Human Genetic Disorders. Annu. Rev. Genomics Hum. Genet. 2006, 7, 125–148. [Google Scholar] [CrossRef] [PubMed]
  69. Wyatt, T.A.; Forget, M.A.; Adams, J.M.; Sisson, J.H. Both cAMP and cGMP are required for maximal ciliary beat stimulation in a cell-free model of bovine ciliary axonemes. Am. J. Physiol Lung Cell. Mol. Physiol. 2005, 288, L546–L551. [Google Scholar] [CrossRef] [PubMed]
  70. Wyatt, T.A.; Spurzem, J.R.; May, K.; Sisson, J.H. Regulation of ciliary beat frequency by both PKA and PKG in bovine airway epithelial cells. Am. J. Physiol 1998, 275 Pt 1, L827–L835. [Google Scholar] [PubMed]
  71. Zhang, L.; Sanderson, M.J. Oscillations in ciliary beat frequency and intracellular calcium concentration in rabbit tracheal epithelial cells induced by ATP. J. Physiol. 2003, 546 Pt 3, 733–749. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, L.; Sanderson, M.J. The role of cGMP in the regulation of rabbit airway ciliary beat frequency. J. Physiol. 2003, 551 Pt 3, 765–776. [Google Scholar] [CrossRef] [PubMed]
  73. Schmid, A.; Sutto, Z.; Nlend, M.C.; Horvath, G.; Schmid, N.; Buck, J.; Levin, L.R.; Conner, G.E.; Fregien, N.; Salathe, M. Soluble adenylyl cyclase is localized to cilia and contributes to ciliary beat frequency regulation via production of cAMP. J. Gen. Physiol. 2007, 130, 99–109. [Google Scholar] [CrossRef] [PubMed]
  74. Sutto, Z.; Conner, G.E.; Salathe, M. Regulation of human airway ciliary beat frequency by intracellular pH. J. Physiol. 2004, 560, 519–532. [Google Scholar] [CrossRef] [PubMed]
  75. Daviskas, E.; Anderson, S.D. Hyperosmolar agents and clearance of mucus in the diseased airway. J. Aerosol Med. 2006, 19, 100–109. [Google Scholar] [CrossRef] [PubMed]
  76. Braunstahl, G.J.; Fokkens, W.J.; Overbeek, S.E.; Kleinjan, A.; Hoogsteden, H.C.; Prins, J.B. Mucosal and systemic inflammatory changes in allergic rhinitis and asthma: A comparison between upper and lower airways. Clin. Exp. Allergy 2003, 33, 579–587. [Google Scholar] [CrossRef] [PubMed]
  77. Bertrand, B.; Collet, S.; Eloy, P.; Rombaux, P. Secondary ciliary dyskinesia in upper respiratory tract. Acta Otorhinolaryngol. Belg. 2000, 54, 309–316. [Google Scholar] [PubMed]
  78. Piatti, G.; Ambrosetti, U.; Santus, P.; Allegra, L. Effects of salmeterol on cilia and mucus in COPD and pneumonia patients. Pharmacol. Res. 2005, 51, 165–168. [Google Scholar] [CrossRef] [PubMed]
  79. BeruBe, K.; Balharry, D.; Sexton, K.; Koshy, L.; Jones, T. Combustion-derived nanoparticles: Mechanisms of pulmonary toxicity. Clin. Exp. Pharmacol. Physiol. 2007, 34, 1044–1050. [Google Scholar] [CrossRef] [PubMed]
  80. Duffin, R.; Mills, N.L.; Donaldson, K. Nanoparticles-a thoracic toxicology perspective. Yonsei Med. J. 2007, 48, 561–572. [Google Scholar] [CrossRef] [PubMed]
  81. Monteiller, C.; Tran, L.; MacNee, W.; Faux, S.; Jones, A.; Miller, B.; Donaldson, K. The pro-inflammatory effects of low-toxicity low-solubility particles, nanoparticles and fine particles, on epithelial cells in vitro: The role of surface area. Occup. Environ. Med. 2007, 64, 609–615. [Google Scholar] [CrossRef] [PubMed]
  82. Pace, E.; Ferraro, M.; Di, V.S.; Gerbino, S.; Bruno, A.; Lanata, L.; Gjomarkaj, M. Oxidative stress and innate immunity responses in cigarette smoke stimulated nasal epithelial cells. Toxicol. In Vitro 2014, 28, 292–299. [Google Scholar] [CrossRef] [PubMed]
  83. Lam, H.C.; Cloonan, S.M.; Bhashyam, A.R.; Haspel, J.A.; Singh, A.; Sathirapongsasuti, J.F.; Cervo, M.; Yao, H.; Chung, A.L.; Mizumura, K.; et al. Histone deacetylase 6-mediated selective autophagy regulates COPD-associated cilia dysfunction. J. Clin. Investig. 2013, 123, 5212–5230. [Google Scholar] [CrossRef] [PubMed]
  84. Pierrou, S.; Broberg, P.; O’donnell, R.A.; Pawlowski, K.; Virtala, R.; Lindqvist, E.; Richter, A.; Wilson, S.J.; Angco, G.; Moller, S.; et al. Expression of genes involved in oxidative stress responses in airway epithelial cells of smokers with chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2007, 175, 577–586. [Google Scholar] [CrossRef] [PubMed]
  85. Yoshida, T.; Tuder, R.M. Pathobiology of cigarette smoke-induced chronic obstructive pulmonary disease. Physiol. Rev. 2007, 87, 1047–1082. [Google Scholar] [CrossRef] [PubMed]
  86. Daviskas, E.; Anderson, S.D.; Young, I.H. Effect of mannitol and repetitive coughing on the sputum properties in bronchiectasis. Respir. Med. 2010, 104, 371–377. [Google Scholar] [CrossRef] [PubMed]
  87. Daviskas, E.; Robinson, M.; Anderson, S.D.; Bye, P.T. Osmotic stimuli increase clearance of mucus in patients with mucociliary dysfunction. J. Aerosol Med. 2002, 15, 331–341. [Google Scholar] [CrossRef] [PubMed]
  88. Newhouse, M.T.; Rossman, C.M.; Dolovich, J.; Dolovich, M.B.; Wilson, W.M. Impairment of mucociliary transport in cystic fibrosis. Mod. Probl. Paediatr. 1976, 19, 190–198. [Google Scholar] [PubMed]
  89. Rossman, C.; Dolovich, J.; Dolovich, M.; Wilson, W.; Newhouse, M. Cystic fibrosis--related inhibition of mucociliary clearance in vivo in man. J. Pediatr. 1977, 90, 579–584. [Google Scholar] [CrossRef]
  90. Puchelle, E.; Zahm, J.M. Influence of rheological properties of human bronchial secretions on the ciliary beat frequency. Biorheology 1984, 21, 265–272. [Google Scholar] [PubMed]
  91. Veale, D.; Rodgers, A.D.; Griffiths, C.J.; Ashcroft, T.; Gibson, G.J. Variability in ciliary beat frequency in normal subjects and in patients with bronchiectasis. Thorax 1993, 48, 1018–1020. [Google Scholar] [CrossRef] [PubMed]
  92. Chapelin, C.; Coste, A.; Gilain, L.; Poron, F.; Verra, F.; Escudier, E. Modified epithelial cell distribution in chronic airways inflammation. Eur. Respir. J. 1996, 9, 2474–2478. [Google Scholar] [CrossRef] [PubMed]
  93. Joki, S.; Toskala, E.; Saano, V.; Nuutinen, J. Correlation between ciliary beat frequency and the structure of ciliated epithelia in pathologic human nasal mucosa. Laryngoscope 1998, 108, 426–430. [Google Scholar] [CrossRef] [PubMed]
  94. Hisamatsu, K.; Nakajima, M. Pranlukast protects leukotriene C4- and D4-induced epithelial cell impairment of the nasal mucosa in vitro. Life Sci. 2000, 67, 2767–2773. [Google Scholar] [CrossRef]
  95. Sabater, J.R.; Wanner, A.; Abraham, W.M. Montelukast prevents antigen-induced mucociliary dysfunction in sheep. Am. J. Respir. Crit. Care Med. 2002, 166, 1457–1460. [Google Scholar] [CrossRef] [PubMed]
  96. Trevisani, L.; Sartori, S.; Pazzi, P.; Potena, A.; Aleotti, A.; Grandi, E.; Stabellini, G. Acquired ciliary defects in bronchial epithelium of patients with chronic bronchitis. Respiration 1990, 57, 6–13. [Google Scholar] [CrossRef] [PubMed]
  97. Wilson, R.; Roberts, D.; Cole, P. Effect of bacterial products on human ciliary function in vitro. Thorax 1985, 40, 125–131. [Google Scholar] [CrossRef] [PubMed]
  98. Tristram, D.A.; Hicks, W., Jr.; Hard, R. Respiratory syncytial virus and human bronchial epithelium. Arch. Otolaryngol. Head Neck Surg. 1998, 124, 777–783. [Google Scholar] [CrossRef] [PubMed]
  99. Carson, J.L.; Collier, A.M.; Hu, S.S. Acquired ciliary defects in nasal epithelium of children with acute viral upper respiratory infections. N. Engl. J. Med. 1985, 312, 463–468. [Google Scholar] [CrossRef] [PubMed]
  100. Wong, J.Y.; Rutman, A.; O’Callaghan, C. Recovery of the ciliated epithelium following acute bronchiolitis in infancy. Thorax 2005, 60, 582–587. [Google Scholar] [CrossRef] [PubMed]
  101. Low, P.M.; Luk, C.K.; Dulfano, M.J.; Finch, P.J. Ciliary beat frequency of human respiratory tract by different sampling techniques. Am. Rev. Respir. Dis. 1984, 130, 497–498. [Google Scholar] [PubMed]
  102. Boek, W.M.; Graamans, K.; Natzijl, H.; van Rijk, P.P.; Huizing, E.H. Nasal mucociliary transport: New evidence for a key role of ciliary beat frequency. Laryngoscope 2002, 112, 570–573. [Google Scholar] [CrossRef] [PubMed]
  103. Verra, F.; Fleury-Feith, J.; Boucherat, M.; Pinchon, M.C.; Bignon, J.; Escudier, E. Do nasal ciliary changes reflect bronchial changes? An ultrastructural study. Am. Rev. Respir. Dis. 1993, 147, 908–913. [Google Scholar] [CrossRef] [PubMed]
  104. Hanes, L.S.; Issa, E.; Proud, D.; Togias, A. Stronger nasal responsiveness to cold air in individuals with rhinitis and asthma, compared with rhinitis alone. Clin. Exp. Allergy 2006, 36, 26–31. [Google Scholar] [CrossRef] [PubMed]
  105. Benson, M.; Jansson, L.; Adner, M.; Luts, A.; Uddman, R.; Cardell, L.O. Gene profiling reveals decreased expression of uteroglobin and other anti-inflammatory genes in nasal fluid cells from patients with intermittent allergic rhinitis. Clin. Exp. Allergy 2005, 35, 473–478. [Google Scholar] [CrossRef] [PubMed]
  106. Devalia, J.L.; Sapsford, R.J.; Rusznak, C.; Toumbis, M.J.; Davies, R.J. The effects of salmeterol and salbutamol on ciliary beat frequency of cultured human bronchial epithelial cells, in vitro. Pulm. Pharmacol. 1992, 5, 257–263. [Google Scholar] [CrossRef]
  107. Hermens, W.A.; Merkus, F.W. The influence of drugs on nasal ciliary movement. Pharm. Res. 1987, 4, 445–449. [Google Scholar] [CrossRef] [PubMed]
  108. Romeijn, S.G.; Verhoef, J.C.; Marttin, E.; Merkus, F.W. The effect of nasal drug formulations on ciliary beating in vitro. Int. J. Pharm. 1996, 135, 137–145. [Google Scholar] [CrossRef]
  109. WatermarkedGlobal-Strategy-2016.pdf. From the Global Strategy for the Diagnosis, Management and Prevention of COPD, Global Initiative for Chronic Obstructive Lung Disease (GOLD) 2016. Available online: http://goldcopd.org/ (accessed on 8 November 2016).
  110. Barnes, P.J.; Stockley, R.A. COPD: Current therapeutic interventions and future approaches. Eur. Respir. J. 2005, 25, 1084–1106. [Google Scholar] [CrossRef] [PubMed]
  111. Barnes, P.J. Emerging pharmacotherapies for COPD. Chest 2008, 134, 1278–1286. [Google Scholar] [CrossRef] [PubMed]
  112. O’donnell, R.A.; Peebles, C.; Ward, J.A.; Daraker, A.; Angco, G.; Broberg, P.; Pierrou, S.; Lund, J.; Holgate, S.T.; Davies, D.E.; et al. Relationship between peripheral airway dysfunction, airway obstruction, and neutrophilic inflammation in COPD. Thorax 2004, 59, 837–842. [Google Scholar] [CrossRef] [PubMed]
  113. Lapperre, T.S.; Postma, D.S.; Gosman, M.M.; Snoeck-Stroband, J.B.; ten Hacken, N.H.; Hiemstra, P.S.; Timens, W.; Sterk, P.J.; Mauad, T. Relation between duration of smoking cessation and bronchial inflammation in COPD. Thorax 2006, 61, 115–121. [Google Scholar] [CrossRef] [PubMed]
  114. O’Donnell, R.; Breen, D.; Wilson, S.; Djukanovic, R. Inflammatory cells in the airways in COPD. Thorax 2006, 61, 448–454. [Google Scholar] [CrossRef] [PubMed]
  115. Hurst, J.R.; Wilkinson, T.M.; Perera, W.R.; Donaldson, G.C.; Wedzicha, J.A. Relationships among bacteria, upper airway, lower airway, and systemic inflammation in COPD. Chest 2005, 127, 1219–1226. [Google Scholar] [CrossRef]
  116. Tamaoki, J.; Sakai, N.; Kobayashi, K.; Kanemura, T.; Takizawa, T. Stimulation of airway ciliary motility by immunologically activated canine pulmonary macrophages: Role of leukotrienes. Acta Physiol. Scand. 1991, 141, 415–420. [Google Scholar] [CrossRef] [PubMed]
  117. Devalia, J.L.; Davies, R.J. Human nasal and bronchial epithelial cells in culture: An overview of their characteristics and function. Allergy Proc. 1991, 12, 71–79. [Google Scholar] [CrossRef] [PubMed]
  118. Taggart, C.C.; Greene, C.M.; Carroll, T.P.; O’neill, S.J.; McElvaney, N.G. Elastolytic proteases: Inflammation resolution and dysregulation in chronic infective lung disease. Am. J. Respir. Crit. Care Med. 2005, 171, 1070–1076. [Google Scholar] [CrossRef] [PubMed]
  119. Van Wetering, S.; Sterk, P.J.; Rabe, K.F.; Hiemstra, P.S. Defensins: Key players or bystanders in infection, injury, and repair in the lung? J. Allergy Clin. Immunol. 1999, 104, 1131–1138. [Google Scholar] [CrossRef]
  120. Montuschi, P.; Kharitonov, S.A.; Ciabattoni, G.; Barnes, P.J. Exhaled leukotrienes and prostaglandins in COPD. Thorax 2003, 58, 585–588. [Google Scholar] [CrossRef] [PubMed]
  121. Rossman, C.M.; Newhouse, M.T. Primary ciliary dyskinesia: Evaluation and management. Pediatr. Pulmonol. 1988, 5, 36–50. [Google Scholar] [CrossRef] [PubMed]
  122. Dolovich, M.B.; Mahony, J.B.; Chambers, C.; Newhouse, M.T.; Chernesky, M.A. Ciliary function, cell viability, and in vitro effect of ribavirin on nasal epithelial cells in acute rhinorrhea. Chest 1992, 102, 284–287. [Google Scholar] [CrossRef] [PubMed]
  123. Wolff, R.K. Effects of airborne pollutants on mucociliary clearance. Environ. Health Perspect. 1986, 66, 223–237. [Google Scholar] [CrossRef] [PubMed]
  124. Stanley, P.J.; Wilson, R.; Greenstone, M.A.; MacWilliam, L.; Cole, P.J. Effect of cigarette smoking on nasal mucociliary clearance and ciliary beat frequency. Thorax 1986, 41, 519–523. [Google Scholar] [CrossRef] [PubMed]
  125. Sisson, J.H.; Papi, A.; Beckmann, J.D.; Leise, K.L.; Wisecarver, J.; Brodersen, B.W.; Kelling, C.L.; Spurzem, J.R.; Rennard, S.I. Smoke and viral infection cause cilia loss detectable by bronchoalveolar lavage cytology and dynein ELISA. Am. J. Respir. Crit. Care Med. 1994, 149, 205–213. [Google Scholar] [CrossRef] [PubMed]
  126. Verra, F.; Escudier, E.; Lebargy, F.; Bernaudin, J.F.; De, C.H.; Bignon, J. Ciliary abnormalities in bronchial epithelium of smokers, ex-smokers, and nonsmokers. Am. J. Respir. Crit. Care Med. 1995, 151 Pt 1, 630–634. [Google Scholar] [CrossRef] [PubMed]
  127. O’Donnell, D.E.; Aaron, S.; Bourbeau, J.; Hernandez, P.; Marciniuk, D.; Balter, M.; Ford, G.; Gervais, A.; Goldstein, R.; Hodder, R.; et al. State of the Art Compendium: Canadian Thoracic Society recommendations for the management of chronic obstructive pulmonary disease. Can. Respir. J. 2004, 11 (Suppl. B), 7B–59B. [Google Scholar] [CrossRef] [PubMed]
  128. Rankin, H.V.; Moody, A.J.; Moate, R.M.; Macnaughton, P.D.; Rahamim, J.; Smith, M.E.; Sneyd, J.R. Elevated oxygen fraction reduces cilial abundance in explanted human bronchial tissue. Ultrastruct. Pathol. 2007, 31, 339–346. [Google Scholar] [CrossRef] [PubMed]
  129. Auerbach, O.; Hammond, E.C.; Garfinkel, L. Bronchial epithelium and cigarette smoking. N. Engl. J. Med. 1979, 300, 1395–1396. [Google Scholar] [PubMed]
  130. Hobson, J.; Wright, J.; Churg, A. Histochemical evidence for generation of active oxygen species on the apical surface of cigarette-smoke-exposed tracheal explants. Am. J. Pathol. 1991, 139, 573–580. [Google Scholar] [PubMed]
  131. Rutland, J.; Cox, T.; Dewar, A.; Cole, P. Screening for ciliary dyskinesia - a spectrum of defects of motility and structure. Eur. J. Respir. Dis. Suppl. 1983, 127, 71–77. [Google Scholar] [PubMed]
  132. Fox, B.; Bull, T.B.; Oliver, T.N. The distribution and assessment of electron-microscopic abnormalities of human cilia. Eur. J. Respir. Dis. Suppl 1983, 127, 11–18. [Google Scholar] [PubMed]
  133. Wyatt, T.A.; Sisson, J.H.; Pavlik, J.A.; Devasure, J.; Boyer, C.; Liu, X.; Kawasaki, S.; Rennard, S.I. Long-Term Cigarette Smoke Exposure in a Mouse Model of Ciliated Epithelial Cell Function. Am. J. Respir. Crit. Care Med. 2010, 43, 634–640. [Google Scholar]
  134. Brekman, A.; Walters, M.S.; Tilley, A.E.; Crystal, R.G. FOXJ1 prevents cilia growth inhibition by cigarette smoke in human airway epithelium in vitro. Am. J. Respir. Cell Mol. Biol. 2014, 51, 688–700. [Google Scholar] [CrossRef] [PubMed]
  135. Hessel, J.; Heldrich, J.; Fuller, J.; Staudt, M.R.; Radisch, S.; Hollmann, C.; Harvey, B.G.; Kaner, R.J.; Salit, J.; Yee-Levin, J.; et al. Intraflagellar transport gene expression associated with short cilia in smoking and COPD. PLoS ONE 2014, 9, e85453. [Google Scholar] [CrossRef] [PubMed]
  136. Daviskas, E.; Anderson, S.D.; Gomes, K.; Briffa, P.; Cochrane, B.; Chan, H.K.; Young, I.H.; Rubin, B.K. Inhaled mannitol for the treatment of mucociliary dysfunction in patients with bronchiectasis: Effect on lung function, health status and sputum. Respirology 2005, 10, 46–56. [Google Scholar] [CrossRef] [PubMed]
  137. Daviskas, E.; Anderson, S.D.; Jaques, A.; Charlton, B. Inhaled Mannitol Improves the Hydration and Surface properties of Sputum in patients with Cystic Fibrosis. Chest 2010, 137, 861–868. [Google Scholar] [CrossRef] [PubMed]
  138. Cutz, E.; Levison, H.; Cooper, D.M. Ultrastructure of airways in children with asthma. Histopathology 1978, 2, 407–421. [Google Scholar] [CrossRef] [PubMed]
  139. Dulfano, M.J.; Luk, C.K.; Beckage, M.; Wooten, O. Ciliary inhibitory effects of asthma patients’ sputum. Clin. Sci. (Lond.) 1982, 63, 393–396. [Google Scholar] [CrossRef] [PubMed]
  140. Dulfano, M.J.; Luk, C.K. Sputum and ciliary inhibition in asthma. Thorax 1982, 37, 646–651. [Google Scholar] [CrossRef] [PubMed]
  141. Mezey, R.J.; Cohn, M.A.; Fernandez, R.J.; Januszkiewicz, A.J.; Wanner, A. Mucociliary transport in allergic patients with antigen-induced bronchospasm. Am. Rev. Respir. Dis. 1978, 118, 677–684. [Google Scholar] [CrossRef] [PubMed]
  142. Wanner, A. Allergic mucociliary dysfunction. Laryngoscope 1983, 93, 68–70. [Google Scholar] [CrossRef] [PubMed]
  143. Wanner, A. Allergic mucociliary dysfunction. J. Allergy Clin. Immunol. 1983, 72, 347–350. [Google Scholar] [CrossRef]
  144. Wanner, A. The role of mucociliary dysfunction in bronchial asthma. Am. J. Med. 1979, 67, 477–485. [Google Scholar] [CrossRef]
  145. Daviskas, E.; Anderson, S.D.; Gonda, I.; Eberl, S.; Meikle, S.; Seale, J.P.; Bautovich, G. Inhalation of hypertonic saline aerosol enhances mucociliary clearance in asthmatic and healthy subjects. Eur. Respir. J. 1996, 9, 725–732. [Google Scholar] [CrossRef] [PubMed]
  146. Daviskas, E.; Anderson, S.D.; Brannan, J.D.; Chan, H.K.; Eberl, S.; Bautovich, G. Inhalation of dry-powder mannitol increases mucociliary clearance. Eur. Respir. J. 1997, 10, 2449–2454. [Google Scholar] [CrossRef] [PubMed]
  147. Heino, M.; Karjalainen, J.; Ylikoski, J.; Laitinen, A.; Laitinen, L.A. Bronchial ciliogenesis and oral steroid treatment in patients with asthma. Br. J. Dis. Chest 1988, 82, 175–178. [Google Scholar] [CrossRef]
  148. Agnew, J.E.; Bateman, J.R.; Sheahan, N.F.; Lennard-Jones, A.M.; Pavia, D.; Clarke, S.W. Effect of oral corticosteroids on mucus clearance by cough and mucociliary transport in stable asthma. Bull. Eur. Physiopathol. Respir. 1983, 19, 37–41. [Google Scholar] [PubMed]
  149. Lundgren, R.; Soderberg, M.; Horstedt, P.; Stenling, R. Morphological studies of bronchial mucosal biopsies from asthmatics before and after ten years of treatment with inhaled steroids. Eur. Respir. J. 1988, 1, 883–889. [Google Scholar] [PubMed]
Figure 1. High Speed Digital Imaging Setup for data acquisition. The Motion Analyzer System attached to the microscope (MotionScope 1000 S monochrome, MotionScope PCI High Speed Digital Imaging System from Redlake MASD Inc., San Diego, CA, USA). A ciliated Epithelium specimen is placed in a perfusion chamber (Dvorak-Stotler Controlled Environment Culture System, Nicholson Precision Instruments Inc., Bethesda, MD, USA) attached to a gravity-fed perfusion system which delivers perfusate at the rate of 0.25 mL/min. The chamber is placed under the phase contrast microscope (E. Leitz, Wetzlar, Germany). A Plexiglas enclosure and an air stream stage heater (Nicholson Precision Instruments Inc., Bethesda, MD, USA) maintain the 37 °C temperature of the preparation. A MotionScope high-speed digital camera and PCI application software, running in a Windows environment (Redlake MASD Inc.), is used for image acquisition. A video segment is recorded for each area. Each video segment is stored in a file for later retrieval and analysis (Figure 2).
Figure 1. High Speed Digital Imaging Setup for data acquisition. The Motion Analyzer System attached to the microscope (MotionScope 1000 S monochrome, MotionScope PCI High Speed Digital Imaging System from Redlake MASD Inc., San Diego, CA, USA). A ciliated Epithelium specimen is placed in a perfusion chamber (Dvorak-Stotler Controlled Environment Culture System, Nicholson Precision Instruments Inc., Bethesda, MD, USA) attached to a gravity-fed perfusion system which delivers perfusate at the rate of 0.25 mL/min. The chamber is placed under the phase contrast microscope (E. Leitz, Wetzlar, Germany). A Plexiglas enclosure and an air stream stage heater (Nicholson Precision Instruments Inc., Bethesda, MD, USA) maintain the 37 °C temperature of the preparation. A MotionScope high-speed digital camera and PCI application software, running in a Windows environment (Redlake MASD Inc.), is used for image acquisition. A video segment is recorded for each area. Each video segment is stored in a file for later retrieval and analysis (Figure 2).
Cells 05 00040 g001
Figure 2. Video analysis and calculation of CBF in a nasal epithelial strip of cells. (a) Cilia are outlined; (b) Grayscale of beating cilia expressed per time and indicating 15 beats/s (i.e., Hz). Magnification 320×; (c) Ciliary beat frequency patterns illustrating amplitude and beat patterns. The video recordings are obtained and analyzed using ProAnalyst video imaging processing software (XCitex, Cambridge, MA, USA). Briefly, areas on the edge of the epithelium with beating cilia are analyzed using the one-dimensional tracking algorithm included in the motion analysis software ProAnalyst. A line is drawn across a segment of cilia (Figure 2a). The software captures the motion history within the analysis grid for the duration of the video and records the gray-scale intensity variation as a function of time (Figure 2b,c). The resulting files are calibrated as pixels/unit. This tracking process allows the plotting of the sinusoidal waveform generated by the beating cilia and the determination of the frequency (Hz). Figure 2c Evaluation of ciliary function (CBF and waveform) shows a 1 s CBF recording of 11 beats/s and CBF waveform panels of 8.7 beats/s and 3.5 beats/s. The resolution of the obtained images at a microscope magnification of 320× and a sampling rate of 250 frames per second is 480 × 420 pixels, where one pixel corresponds to 0.158 μm.
Figure 2. Video analysis and calculation of CBF in a nasal epithelial strip of cells. (a) Cilia are outlined; (b) Grayscale of beating cilia expressed per time and indicating 15 beats/s (i.e., Hz). Magnification 320×; (c) Ciliary beat frequency patterns illustrating amplitude and beat patterns. The video recordings are obtained and analyzed using ProAnalyst video imaging processing software (XCitex, Cambridge, MA, USA). Briefly, areas on the edge of the epithelium with beating cilia are analyzed using the one-dimensional tracking algorithm included in the motion analysis software ProAnalyst. A line is drawn across a segment of cilia (Figure 2a). The software captures the motion history within the analysis grid for the duration of the video and records the gray-scale intensity variation as a function of time (Figure 2b,c). The resulting files are calibrated as pixels/unit. This tracking process allows the plotting of the sinusoidal waveform generated by the beating cilia and the determination of the frequency (Hz). Figure 2c Evaluation of ciliary function (CBF and waveform) shows a 1 s CBF recording of 11 beats/s and CBF waveform panels of 8.7 beats/s and 3.5 beats/s. The resolution of the obtained images at a microscope magnification of 320× and a sampling rate of 250 frames per second is 480 × 420 pixels, where one pixel corresponds to 0.158 μm.
Cells 05 00040 g002
Figure 3. Ultrastructure of airway cilia. An illustration of Transmission Electron Microscopy (TEM) demonstrating axonemes. TEM sections of airway cilia can be analyzed for the number of inner and outer dynein arms, central tubules, and orientation of cilia.
Figure 3. Ultrastructure of airway cilia. An illustration of Transmission Electron Microscopy (TEM) demonstrating axonemes. TEM sections of airway cilia can be analyzed for the number of inner and outer dynein arms, central tubules, and orientation of cilia.
Cells 05 00040 g003
Figure 4. The effect of exogenous and endogenous mediators on CBF of airway epithelium. Many drugs and excipients have been shown to affect CBF and mucus clearance. Beta-adrenergic and cholinergic agents stimulate CBF, while fluticasone, some preservatives, and some bacterial toxins inhibit CBF [13,16,18,21,50,58,106,107,108].
Figure 4. The effect of exogenous and endogenous mediators on CBF of airway epithelium. Many drugs and excipients have been shown to affect CBF and mucus clearance. Beta-adrenergic and cholinergic agents stimulate CBF, while fluticasone, some preservatives, and some bacterial toxins inhibit CBF [13,16,18,21,50,58,106,107,108].
Cells 05 00040 g004
Figure 5. Effect of mannitol, dextran, and saline on CBF of ciliated HBEC. CBF (Hz) was measured from the same sites (10–12 epithelial cell strips) before and after perfusion with mannitol, dextran, or saline for 10 min or 30 min. CBF increased after acute (10 and 30 min) exposure to mannitol and decreased with dextran. No significant change in CBF occurred with hypertonic saline. A fresh batch of HBEC was used per agent. Data shown as % baseline CBF (mean ± SEM). * # ANOVA plus Tukey’s test, p < 0.01 for 10 min; p < 0.05 for 30 min.
Figure 5. Effect of mannitol, dextran, and saline on CBF of ciliated HBEC. CBF (Hz) was measured from the same sites (10–12 epithelial cell strips) before and after perfusion with mannitol, dextran, or saline for 10 min or 30 min. CBF increased after acute (10 and 30 min) exposure to mannitol and decreased with dextran. No significant change in CBF occurred with hypertonic saline. A fresh batch of HBEC was used per agent. Data shown as % baseline CBF (mean ± SEM). * # ANOVA plus Tukey’s test, p < 0.01 for 10 min; p < 0.05 for 30 min.
Cells 05 00040 g005
Figure 6. Nasal brushings are non-invasive and reliable to study cilia function and structure. (a) CBF (Hz) of ciliated epithelium obtained by brushing the inferior nasal turbinate of healthy subjects: comparison of cilia from the right (n = 7) and left (n = 3) nasal turbinate. There is no significant difference in CBF of ciliated epithelium obtained from the right and left nasal turbinate; (b) CBF (Hz) of ciliated epithelium obtained by brushing of the right inferior nasal turbinate of healthy subjects (n = 3). CBF was measured from 8 to 10 sites on the day the sample was obtained (DAY 1), then the sample was stored in the fridge (4 °C) for seven days before another CBF measurement was obtained. Ciliated epithelium can be stored at 4 °C for seven days in Earle’s balanced Salt Solution without any significant change in CBF. Note: Methods as illustrated in Figure 1 and Figure 2. For all epithelial samples, each CBF value is the average CBF measured from 8 to 10 individual sites. Data are expressed as the mean ± SEM (standard error of the mean) of n where n = number of subjects.
Figure 6. Nasal brushings are non-invasive and reliable to study cilia function and structure. (a) CBF (Hz) of ciliated epithelium obtained by brushing the inferior nasal turbinate of healthy subjects: comparison of cilia from the right (n = 7) and left (n = 3) nasal turbinate. There is no significant difference in CBF of ciliated epithelium obtained from the right and left nasal turbinate; (b) CBF (Hz) of ciliated epithelium obtained by brushing of the right inferior nasal turbinate of healthy subjects (n = 3). CBF was measured from 8 to 10 sites on the day the sample was obtained (DAY 1), then the sample was stored in the fridge (4 °C) for seven days before another CBF measurement was obtained. Ciliated epithelium can be stored at 4 °C for seven days in Earle’s balanced Salt Solution without any significant change in CBF. Note: Methods as illustrated in Figure 1 and Figure 2. For all epithelial samples, each CBF value is the average CBF measured from 8 to 10 individual sites. Data are expressed as the mean ± SEM (standard error of the mean) of n where n = number of subjects.
Cells 05 00040 g006
Figure 7. CBF (Hz) of nasal cilia was significantly depressed in cilia obtained from moderate and severe COPD subjects compared to At Risk and Control subjects. CBF (Hz) of ciliated nasal epithelium of Control (n = 6), At Risk (cough and phlegm but no COPD) (n = 7) and COPD subjects (Moderate, n = 5 and Severe, n = 7). * p = 0.029, Kruskal-Wallis test. For all epithelial samples, each CBF value is the average CBF measured from 10 to 12 individual sites. Data are the mean ± SEM of n where n = number of subjects.
Figure 7. CBF (Hz) of nasal cilia was significantly depressed in cilia obtained from moderate and severe COPD subjects compared to At Risk and Control subjects. CBF (Hz) of ciliated nasal epithelium of Control (n = 6), At Risk (cough and phlegm but no COPD) (n = 7) and COPD subjects (Moderate, n = 5 and Severe, n = 7). * p = 0.029, Kruskal-Wallis test. For all epithelial samples, each CBF value is the average CBF measured from 10 to 12 individual sites. Data are the mean ± SEM of n where n = number of subjects.
Cells 05 00040 g007
Figure 8. Possible modulation of CBF by pharmacologic agents in COPD. Representative schemes of the possible sites of action of the pharmacologic agents tested on nasal cilia obtained from Control, At Risk, and COPD subjects. Information presented here is derived from the review of signaling pathways involved in mammalian ciliary beating [13], and references in our COPD paper [52] and in this review. Beta agonists cause an immediate cAMP-dependent increase in CBF, followed by a cAMP-dependent increase in intracellular calcium, which in turn increases CBF. Salmeterol (1 µM), a beta-adrenergic agonist, increases CBF in a cAMP-dependent manner via beta two receptors (G-alpha-s coupled R) expressed at the apical membranes of epithelial cells—i.e., acts via increased cAMP, PKA activation, and increased CBF due to phosphorylation of axonemal proteins. Tiotropium Bromide (5 µM) results in muscarinic AchR inhibition (i.e., inhibits muscarinic M3 receptors on parasympathetic postganglionic effector cell junctions) and plays a role in increased intracellular calcium leading to increased CBF; also, increased intracellular calcium activates NOS leading to increased cGMP and increased cGMP-dependent protein kinase and increased CBF. The effect is on a G-alpha-q coupled R and IP3 release which leads to increased calcium influx. Licofelone (10 µM), a dual inhibitor of cyclooxygenase and 5-lipoxygenase pathways—i.e., inhibits 5-LO LTs (LTB4, C4 and D4), prostaglandins, and thromboxanes. Inhibition of both pathways leads to a shift into the metabolism of AA to CYP metabolites (EETs and 20-HETE) which activate influx of calcium into the cell and increased CBF. PKA and PKG activation are also possible. Luteolin (100 µM) inhibits activity of metalloproteinases MMP-2 and MMP-9. MMPs are not expressed in normal healthy tissues but in diseased tissues that are inflamed or undergoing remodeling and repair and MMPs may be compartmentalized in close proximity to the cell surface—i.e., soluble and cell surface forms. YM976 (100 nM) inhibits PDE4 (PDEIV) resulting in increased cAMP and PKA activation and increased CBF. HNP-1 (6 µM), human neutrophil protein-1 inhibits serpins and also acts as an endogenous antimicrobial agent. Note that increased intracellular calcium leads to activation of ca-dependent cyclases (AC1, 3 and 8) which increase cAMP and lead to increased CBF. In summary, whether the agent modulates a receptor (adrenergic/cholinergic), an enzyme (5-LO, COX, PDE4, serpins), or structural components (MMPs soluble and cell surface forms)—i.e., different mechanisms of action—the final outcome is an increased CBF. From the literature, we know that the main second messengers following receptor stimulation are cAMP and calcium and the final step in increased CBF is phosphorylation of axonemal proteins, suggesting a common signaling pathway among all the agents tested. Further investigation of the intracellular signaling pathways is needed. R: receptor; IP3: inositol tris-phosphate; P450: cytochrome P450; 5-LO: 5-lipoxynase; COX: cyclooxygenase; EETs and 20-HETE cytochrome P450 metabolites that result in increased calcium influx into the cell; PDEIV, phosphodiesterase type 4; cAMP, cyclic adenosine monophosphate; cGMP, cyclic guanosine monophosphate; NOS, nitric oxide synthase; PKA, protein kinase A; PKG, cGMP-dependent protein kinase; sAC, soluble adenylyl cyclase; tmACs, transmembrane adenylyl cyclases; Serpins, serine protease inhibitors.
Figure 8. Possible modulation of CBF by pharmacologic agents in COPD. Representative schemes of the possible sites of action of the pharmacologic agents tested on nasal cilia obtained from Control, At Risk, and COPD subjects. Information presented here is derived from the review of signaling pathways involved in mammalian ciliary beating [13], and references in our COPD paper [52] and in this review. Beta agonists cause an immediate cAMP-dependent increase in CBF, followed by a cAMP-dependent increase in intracellular calcium, which in turn increases CBF. Salmeterol (1 µM), a beta-adrenergic agonist, increases CBF in a cAMP-dependent manner via beta two receptors (G-alpha-s coupled R) expressed at the apical membranes of epithelial cells—i.e., acts via increased cAMP, PKA activation, and increased CBF due to phosphorylation of axonemal proteins. Tiotropium Bromide (5 µM) results in muscarinic AchR inhibition (i.e., inhibits muscarinic M3 receptors on parasympathetic postganglionic effector cell junctions) and plays a role in increased intracellular calcium leading to increased CBF; also, increased intracellular calcium activates NOS leading to increased cGMP and increased cGMP-dependent protein kinase and increased CBF. The effect is on a G-alpha-q coupled R and IP3 release which leads to increased calcium influx. Licofelone (10 µM), a dual inhibitor of cyclooxygenase and 5-lipoxygenase pathways—i.e., inhibits 5-LO LTs (LTB4, C4 and D4), prostaglandins, and thromboxanes. Inhibition of both pathways leads to a shift into the metabolism of AA to CYP metabolites (EETs and 20-HETE) which activate influx of calcium into the cell and increased CBF. PKA and PKG activation are also possible. Luteolin (100 µM) inhibits activity of metalloproteinases MMP-2 and MMP-9. MMPs are not expressed in normal healthy tissues but in diseased tissues that are inflamed or undergoing remodeling and repair and MMPs may be compartmentalized in close proximity to the cell surface—i.e., soluble and cell surface forms. YM976 (100 nM) inhibits PDE4 (PDEIV) resulting in increased cAMP and PKA activation and increased CBF. HNP-1 (6 µM), human neutrophil protein-1 inhibits serpins and also acts as an endogenous antimicrobial agent. Note that increased intracellular calcium leads to activation of ca-dependent cyclases (AC1, 3 and 8) which increase cAMP and lead to increased CBF. In summary, whether the agent modulates a receptor (adrenergic/cholinergic), an enzyme (5-LO, COX, PDE4, serpins), or structural components (MMPs soluble and cell surface forms)—i.e., different mechanisms of action—the final outcome is an increased CBF. From the literature, we know that the main second messengers following receptor stimulation are cAMP and calcium and the final step in increased CBF is phosphorylation of axonemal proteins, suggesting a common signaling pathway among all the agents tested. Further investigation of the intracellular signaling pathways is needed. R: receptor; IP3: inositol tris-phosphate; P450: cytochrome P450; 5-LO: 5-lipoxynase; COX: cyclooxygenase; EETs and 20-HETE cytochrome P450 metabolites that result in increased calcium influx into the cell; PDEIV, phosphodiesterase type 4; cAMP, cyclic adenosine monophosphate; cGMP, cyclic guanosine monophosphate; NOS, nitric oxide synthase; PKA, protein kinase A; PKG, cGMP-dependent protein kinase; sAC, soluble adenylyl cyclase; tmACs, transmembrane adenylyl cyclases; Serpins, serine protease inhibitors.
Cells 05 00040 g008
Table 1. Summary of Modulators of CBF *.
Table 1. Summary of Modulators of CBF *.
Agents/Factors that Increase CBF Agents/Factors that Decrease CBFAgents/Factors that Have No Effect
-
PKA and PKG activation ↑ CBF
-
Osmolar agents modulating mucus layer (dextran, mannitol, saline)
-
mannitol (direct effect on cilia)
-
Bitter compounds (denatonium and thujone) via T2R4 receptors
-
Ach (directly)-role for Ca++ and M3 receptors
-
Nicotine (direct effect)
-
Substance P
-
Theophylline inhibits PDE4 leading to increased cAMP
-
Inflammatory agents: PGE1, PGE2, LTD4, LTC4
-
Beta2-adrenergic agonists: Salmeterol and salbutamol via PKA activation and ↑ cAMP
-
ATP
-
NO
-
Hyaluronic acid
-
Relaxin (↑ cAMP, increases PKA)
-
Temperature rise: 22 °C to 37 °C
-
Increased intracellular pH
-
Increased intracellular bicarbonate
-
Radiation
-
Resection of airway (trachea)
-
PKC activation ↓ CBF
-
Fluticasone
-
Preservatives
-
Bacterial toxins: P. aeruginosa LPS, H. influenza LPS
-
RSV infections
-
Anesthetics; bipivicaine: ciliotoxic, irreversible
-
Some allergies reduce CBF: allergic rhinitis
-
Anti-allergy drugs vary—some increase and some decrease CBF
-
Hog barn suppresses cilia response to stimulation
-
Cotinine (metabolite of nicotine)
-
Propranolol
-
Midazolam
-
Ribavirin
-
Histamine
-
PGF2-alpha
-
Staphylococcus
-
Some allergies
* Summarized from references in this review.

Share and Cite

MDPI and ACS Style

Yaghi, A.; Dolovich, M.B. Airway Epithelial Cell Cilia and Obstructive Lung Disease. Cells 2016, 5, 40. https://doi.org/10.3390/cells5040040

AMA Style

Yaghi A, Dolovich MB. Airway Epithelial Cell Cilia and Obstructive Lung Disease. Cells. 2016; 5(4):40. https://doi.org/10.3390/cells5040040

Chicago/Turabian Style

Yaghi, Asma, and Myrna B. Dolovich. 2016. "Airway Epithelial Cell Cilia and Obstructive Lung Disease" Cells 5, no. 4: 40. https://doi.org/10.3390/cells5040040

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop