American Journal of Respiratory and Critical Care Medicine

Introduction

Nitric Oxide

 Source of NO in exhaled air

 Measurement

 Asthma

 COPD

 Cystic fibrosis

 Bronchiectasis

 Primary ciliary dyskinesia

 Rhinitis

 Interstitial lung diseases

 Pulmonary hypertension

 Occupational diseases

 Infections

 Chronic cough

 Lung cancer

 Lung transplant rejection

 Adult respiratory distress syndrome

 Diffuse Panbronchiolitis

Carbon Monoxide

 Source of exhaled CO

 Measurement

 Asthma

 COPD

 Bronchiectasis

 Cystic fibrosis

 Interstitial lung disease

 Allergic rhinitis

 Infections

 Other conditions

Exhaled Hydrocarbons

 Origin

 Measurement

 Asthma

 COPD

 Cystic Fibrosis

 Other lung diseases

Exhaled Breath Condensate

 Origin

 Hydrogen peroxide

 Eicosanoids

 Products of lipid peroxidation

 Vasoactive amines

 NO-related products

 Ammonia

 Electrolytes

 Hydrogen ions

 Proteins and cytokines

Other Methods

 Exhaled temperature

 Combined gas chromatography/spectroscopy

 The selected ion flow tube (SIFT) technique

 Polymer-coated surface-acoustic-wave resonators

Future Directions

 Standardization of measurements

 Clinical application

 Profiles of mediators

 Measuring devices

 New markers

There has recently been an explosion of interest in the analysis of breath constituents as a way of monitoring inflammation and oxidative stress in the lungs. Here we review the use of exhaled breath analysis in the diagnosis and monitoring of lung disease. Although most studies have focused on exhaled nitric oxide (NO), recently several other volatile gases (carbon monoxide, ethane, pentane) have also been used. In addition, several endogenous substances (inflammatory mediators, cytokines, oxidants) may be detected in expired breath condensates, opening up new perspectives for exhaled breath analysis.

Many lung diseases, including asthma, chronic obstructive pulmonary disease (COPD), bronchiectasis, cystic fibrosis, and interstitial lung disease, involve chronic inflammation and oxidative stress. Yet these are not measured directly in routine clinical practice because of the difficulties in monitoring inflammation. In asthma fiberoptic bronchial biopsies have become the “gold standard” for measuring inflammation in the airway wall, but this is an invasive procedure that is not suitable for routine clinical practice and cannot be repeated often. It is also unsuitable for use in children and patients with severe disease. Symptoms may not accurately reflect the extent of underlying inflammation because of differences in perception and masking by bronchodilators in airway disease. In asthma measurement of airway hyperresponsiveness by histamine or methacholine challenge has been used as a surrogate marker of inflammation, but interpretation may be confounded by the use of bronchodilator therapy. Furthermore, it is difficult to perform this measurement in children and in patients with severe disease. This has led to the use of induced sputum to detect inflammation. This method is relatively reproducible and allows the quantification of inflammatory cells and mediators (1). However, this technique is somewhat invasive as it involves inhalation of hypertonic saline, which may induce coughing and bronchoconstriction, and it is difficult to use in small children. Furthermore, the technique itself induces an inflammatory response so that it is not possible to repeat measurements in less than 24 h (2). The need to monitor inflammation in the lungs has led to the exploration of exhaled gases and condensates. Noninvasive monitoring may assist in differential diagnosis of pulmonary diseases, assessment of disease severity and response to treatment. Because these techniques are completely noninvasive, they can be used repeatedly to give information about kinetics, they can be used in patients with severe disease, which has been previously difficult to monitor, and they can be used to monitor disease in children, including infants. Breath analysis is currently a research procedure, but there is increasing evidence that it may have an important place in the diagnosis and management of lung diseases in the future (3). This will drive the development of cheaper and more convenient analyzers, which can be used in a hospital and later in a family practice setting, then eventually to the development of personal monitoring devices for use by patients.

NO is the most extensively studied exhaled marker and abnormalities in exhaled NO have been documented in several lung diseases (3), particularly asthma (4-6).

Source of NO in Exhaled Air

Nitric oxide synthases. Endogenous NO is derived from l-arginine by the enzyme NO synthase (NOS), of which at least three distinct isoforms exist (7) (Figure 1, panel A). Two of these enzymes are constitutively expressed and are activated by small rises in intracellular calcium concentration, secondary to cell activation. Neuronal NOS (NOS1, nNOS) is predominantly expressed in neurones and endothelial NOS (NOS3, eNOS) mainly in endothelial cells, although other cell types also express both of these isoforms. A third enzyme is inducible (NOS2, iNOS), has a much greater level of activity, and is independent of calcium concentration. NOS2 may be induced by inflammatory cytokines, endotoxin, and viral infections and may show increased expression in inflammatory diseases (8-10). Genetic polymorphisms of all three isoforms of NOS have been detected. Surprisingly, associations have been found between polymorphisms in the NOS1 gene and asthma in Caucasian populations (11, 12). In patients with mild asthma there is a significant association between the length of the AAT repeat polymorphism in intron 20 of the NOS1 gene and exhaled NO levels (13).

Cellular sources in airways. The cellular source of NO gas in the lower respiratory tract is not yet certain. Studies with perfused porcine lungs suggest that exhaled NO originates at the alveolar surface, rather than from the pulmonary circulation (14), and it may be derived from NOS3 expressed in the alveolar walls of normal lungs. Studies in ventilated perfused lungs of guinea pigs have shown that exhaled NO is reduced during perfusion with calcium-free solutions, suggesting that NO is derived from a constitutive NOS, which is calcium- dependent (15). Airway epithelial cells may express both NOS3 and NOS1 and therefore may contribute to NO in the lower respiratory tract (16, 17). There is some expression of NOS2 even in airway epithelial cells from normal subjects (18), and NOS2 appears to be an important isoform contributing to exhaled NO in healthy mice (19). In inflammatory diseases such as asthma it is likely that the increase in exhaled NO reflects further induction of NOS2 in response to inflammatory signals such as proinflammatory cytokines. Indeed, increased NOS activity has been demonstrated in lung tissue of patients with asthma, cystic fibrosis, and obliterative bronchiolitis (20). In asthmatic patients there is evidence for increased expression of NOS2 in airway epithelial cells (21), and this is likely to be due to increased transcription mediated via the transcription factors STAT-1 and nuclear factor-κB (NF-κB), and increased availability of l-arginine (22, 23). Proinflammatory cytokines induce the expression of NOS2 in cultured human airway epithelial cells (24, 25), and it is likely that these same cytokines are released in asthmatic inflammation. NOS2 may be expressed in other cell types such as alveolar macrophages, eosinophils, and other inflammatory cells (26). Further evidence that the increase in exhaled NO is derived from increased NOS2 expression is the observation that corticosteroids inhibit inflammatory induction of NOS2 in epithelial cells (22, 27), decrease expression in bronchial biopsies of asthmatic patients (26), and also reduce exhaled NO concentrations in asthmatic patients (28) (Figure 1, panel B).

Nonenzymatic sources of NO. NOS is not the only source of NO in exhaled air, and exhaled NO is not therefore a direct measure of NOS activity in the lower respiratory tract. NO reacts with thiol-containing molecules such as cysteine and glutathione to form S-nitrosoproteins and S-nitrosothiols (29). Approximately 70 to 90% of NO is released by S-nitrosothiols, which therefore provide a major source of NO in tissues (30). S-nitrosothiols are potent relaxants of human airways and may play an important role in sequestration, releasing, and transportation of NO to its site of action (29).

NO in exhaled air may also be derived from nitrite protonation to form nitrous acid, which releases NO gas with acidification (31). This pH-related pathway has been implicated in acute asthma, when pH in expired condensate is low (32).

Anatomic origin. NO is produced along the entire length of human airways. The conducting airways secrete NO into the lumen, which mixes with alveoli NO during exhalation, resulting in the observed expiratory concentration. The levels of NO derived from the upper respiratory tract (200 to 1,000 ppb) (33-35) and sinuses (1,000 to 30,000 ppb) (36) are a hundred-fold higher than exhaled NO measured in the lower respiratory tract (1 to 9 ppb) (33, 34, 37-42). Several factors may contribute to high nasal levels. The paranasal sinuses produce a high level of NO (43). There is a dense innervation with NOS1-immunoreactive nerve fibers around nasal blood vessels (44). Vasculature-derived NO, however, is not the major source of NO in nasal mucosa, as neuropeptide Y, a powerful vasoconstrictor, reduces nasal blood flow by 37%, but NO by only 7% (45). There appears to be constitutive expression of NOS2 (46) and the transcription factor NF-κB in nasal mucosa (47). Interestingly, the NO outputs from the nostrils are significantly lower on the operated side (site with the reduced NO-generating surface) in patients who have undergone unilateral medial maxillectomy (48).

The source of NO in the lower respiratory tract is also of mixed origin and may be derived from airway and alveolar epithelial cells, which express both NOS3 and NOS1. The contribution of endothelial-derived NO is minimal, as inhaled NOS inhibitors are able to reduce exhaled NO by 40 to 70% (49– 51) without any effect on the systemic circulation. By contrast, l-NMMA infusion modulates blood pressure and heart rate but has only a minimal effect on exhaled NO (49).

Simultaneous measurement of expired CO2 and NO demonstrate that exhaled NO precedes the peak value of CO2 (end-tidal), suggesting that NO is derived from airways rather than from alveoli (33, 52). Direct sampling via fiberoptic bronchoscopy in normal subjects shows a similar levels of NO in trachea and main bronchi to that recorded at the mouth, thus indicating that there is NO derived from the lower airways (33, 42). Exhaled NO is therefore most likely to be of epithelial rather than of endothelial origin, and most NO is derived from airways rather than from alveoli.

Measurement

Expiratory flow, soft palate closure, and dead space air may all influence exhaled NO levels. Therefore, exhaled NO is usually determined during single-breath exhalations against a resistance (38) (Figure 2, panel A) (28, 40, 53) to prevent contamination with nasal NO (54, 55), or using reservoir collection with discarding of the dead space (56). However, this method has proven difficult for some children, who may have trouble maintaining a constant flow, and recently a simple flow-driven method for online NO measurements has been developed that does not require active patient cooperation (57). Recently, single breath analysis of exhaled NO has been successfully performed in the newborn when exhaled air was sampled from the tip of a thin nasal catheter placed in the hypopharynx (58). The most commonly used method to measure nasal NO is to sample nasal air directly from one nostril using the intrinsic flow of the chemiluminescence analyzer (36). A novel method of measuring exhaled NO at several exhalation flow rates has recently been described that can be used to approximate alveolar and airway NO production (59). NO is continuously formed in the airways. Mixing during exhalation between the NO produced by the alveoli and the conducting airways, explains its flow dependency (55) and accumulation during a breathhold (33). A relatively simple and robust two-compartment model of NO has been developed that is capable of simulating many important features of NO exchange in the lungs (60). The model assumes that the lung consists of two well-defined, separate regions: a rigid airway compartment and a well-mixed, expansile alveolar compartment. Both compartments seem to contribute to exhaled NO, and the relative contributions of each seems to be a function of minute ventilation (60). Finally, the model suggests that the relationship between exhaled NO at end-exhalation may be a simple, effective, and reproducible technique for determining the relative contribution of the airways and alveoli to exhaled NO.

It is therefore important to register the flow rate if NO is expressed as a concentration The flow rate recommended in 1997 by a Task Force of the European Respiratory Society is 10 to 15 L/min or 167 to 250 ml/s (53). Most investigators have used about 100 ml/s, but a more recent recommendation from the American Thoracic Society suggests 50 ml/s (61).

Factors Affecting Exhaled NO Measurements

Exhaled and nasal NO in healthy subjects is independent of age, sex, and lung function (34, 62). There is no evidence for significant diurnal variation (63), and exhaled NO measurements are highly reproducible in normal subjects (64, 65). Different phases of the menstrual cycle may influence exhaled NO (66), as estrogen activates NOS3 in airway epithelial cells (67).

There are several major factors, which may change NO levels in normal subjects (Table 1). Either intravenous, or inhaled, or digested l-arginine, the substrate for NOS, increase exhaled NO levels in normal subjects (68-70). Conversely nebulized l-NMMA and l-NAME, nonspecific inhibitors of NOS, reduce exhaled NO (28, 50) and nasal NO (71, 72). Some routinely used tests can transiently reduce exhaled NO; for example, repeated spirometry (73, 74), physical exercise (75), sputum induction (76). Environmental factors such as NO ozone and chlorine dioxide are known to increase exhaled NO levels (77-79). Habitual factors such as smoking (80, 81) and alcohol ingestion (82, 83) reduce exhaled NO. Upper respiratory infection significantly increases exhaled NO (84, 85) and nasal NO (86).

Table 1.  FACTORS AFFECTING EXHALED AND NASAL NO  MEASUREMENTS IN HEALTHY SUBJECTS

Increased NODecreased NO
Pharmacologic
 Papaverin (71)Oxymetazoline (71, 72)
 Sodium nitroprusside (458)NOS inhibitors (50, 51, 71, 72)
l-arginine (68, 186)
 ACE inhibitors (enalapril) (216)
Physiologic and procedural
 Arginine ingestion, nitrite/Repeated spirometry (73, 74)
  nitrate-enriched food (70)Acute and transient after  forced exhalation (74)
Physical exercise (75)
Menstrual cycle (66)
Sputum induction (76)
Body temperature reduction (459)
Environmental, occupational
 Air pollution (NO, ozone) (77)Water vapour, CO2,  nitrous oxide, heptane (462)
 Occupational hazards:100% inspired O2 (463)
  Fluoride, dust (221)Moderate altitude (464)
  Ozone, chlorine dioxide (78)
  Rubber latex (222)
 Formaldehyde (domestic)   exposure (460)
 Electromagnetic field   generated by cellular   phone (nasal NO) (461)
Habitual
Smoking (80, 81)
Alcohol ingestion (82, 83)
Infections
 URTI (84-86)

Definition of abbreviations: ACE = angiotensin-converting enzyme; URTI = upper respiratory tract infection.

Asthma

Increased levels of exhaled NO have been widely documented in patients with asthma (Figure 2, panel B) (28, 87). The increased levels of exhaled NO in asthma have a predominant lower airway origin (33, 42) and are most likely due to activation of NOS2 in airway epithelial and inflammatory cells (21, 26). However, there may be a small contribution from NOS1 as polymorphisms of NOS1 gene are correlated with exhaled NO (13). Exhaled NO may be further elevated by NO substrate l-arginine (69).

Diagnosis and epidemiology. An elevation of exhaled NO is not specific for asthma, but an increased level may be useful in differentiating asthma from other causes of chronic cough (88). The diagnostic value of exhaled NO measurements to differentiate between healthy subjects with or without respiratory symptoms and patients with confirmed asthma has been recently analyzed by Dupont and colleagues (89) with 90% specificity and 95% positive predictive value when exhaled NO > 15 ppb is used as a cutoff for asthma. The intraindividual coefficient of variation (CoV) of exhaled NO in normal subjects was 15.8% within an interval of 7 d, and 16.8% within 23 d, suggesting that the change of exhaled NO by 30 to 35% or more within the interval of 1 to 3 wk would be abnormal (62). Exhaled and nasal NO may be used to identify subjects with atopy, because nonatopic asthmatics have normal exhaled NO (90). There is a strong association between elevated exhaled and nasal NO and skin prick test scores, total IgE (91), and blood eosinophilia (92) in mild asthma. Elevated nasal NO is also related to the size of skin test reactivity in asymptomatic asthmatic subjects (93). This may denote “subclinical” airway inflammation.

Another potential use of exhaled NO levels in patient management is the prediction of future asthma. An elevated exhaled NO may be found in patients with “subclinical” forms of asthma (normal lung function, negative bronchodilator tests, and elevated sputum eosinophilic cationic protein concentrations) (94, 95). Elevated levels of NO in patients with “subclinical asthma” are not in conflict with the specificity of exhaled NO as a marker to diagnose asthma, as lack of current asthma symptoms does not exclude the diagnosis of asthma. Perhaps, this subclinical airway inflammation, which is reflected by elevated levels of exhaled NO in adolescent asymptomatic patients with asthma remission (96), should be treated with corticosteroids to prevent the risk of becoming clinically manifest again. This category of patients with “subclinical” forms of asthma, especially children, may be predisposed to develop asthma in the future (97). This may be studied in epidemiologic studies, in which the reservoir collection of exhaled NO has proved to be useful (98, 99). Airway responsiveness measurements (PC20) in this “high risk” group make the combination of exhaled NO and PC20 a more specific test for allergic asthma. This has recently been demonstrated in a study of more than 8,000 adolescents in Norway (100). Because of the noninvasive character and practicality of exhaled and nasal NO measurements they may be used cost effectively for screening of large populations.

Atopy and exposure to proinflammatory stimuli. Exhaled NO is elevated in allergic/atopic adults and children (97, 101, 102). It is further increased as a result of allergen exposure such as during the late phase response to allergen challenge (103, 104), during the grass pollen season (105), or during exposure to indoor allergens (106, 107). In subjects sensitive to house dust mites (HDM) the wheal size for HDM correlates with exhaled and nasal NO levels (93). Both adults (97) and children (102) with atopic asthma have higher levels of exhaled NO than do patients with nonatopic asthma, even without airway hyperresponsiveness (108).

Exhaled NO may represent a useful biomarker of individual exposure to air pollutants, as even healthy subjects may have elevated exhaled NO levels on days with high outdoor air pollution (79, 109). This may reflect an airway inflammatory response to ozone and nitrogen dioxide (110).

Asthma monitoring. It is difficult to monitor the response of different classes of anti-inflammatory drugs in asthma, as there is no single test that can be used to quantify airway inflammation. Peripheral blood markers are unlikely to be adequate as the most important mediator and cellular responses occur locally within airways. Eosinophils in induced sputum originate from more proximal rather than small airway (111). It is clear that different markers of airway inflammation should be considered together to monitor asthma (3).

Exhaled NO has been used to monitor the effect of anti- inflammatory treatment in asthma (6, 112) and asthma exacerbations, both spontaneous (40) and induced by steroid reduction (113, 114). There is a lack of long-term serial studies of exhaled NO, together with other markers of airway inflammation in sputum and exhaled condensate, lung function and symptoms. Exhaled NO behaves as a “rapid response” marker, which is extremely sensitive to steroid treatment, as it may be significantly reduced even after 6 h following a single treatment with a nebulized corticosteroid steroid (115), or within 2 to 3 d after inhaled corticosteroids (112), reaching maximal effect after 2 to 4 wk of treatment (112, 113, 116-120).

An important issue in asthma management is to prevent overtreatment of patients with steroids. The high sensitivity of exhaled NO to corticosteroid treatment is an advantage, as higher doses of inhaled steroids are not necessary to improve asthma control, e.g., in mild persistent asthma (3). We have demonstrated a dose-dependent reduction in exhaled NO and improvement in asthma symptoms in patients with mild asthmatics after treatment with low doses of inhaled corticosteroids (120), whereas the reduction in sputum eosinophils and similar improvement in symptoms was observed only after the higher dose of steroids (117). This suggests that exhaled NO levels may be too sensitive to determine whether inflammation is adequately controlled (3).

Although exhaled NO levels are normal in patients with moderate asthma treated with corticosteroids (28), increased levels have been observed in patients with severe asthma, despite treatment with oral corticosteroids (98, 121). Individual NO values such as individual peak expiratory flows should be established and monitored, and when the levels are above or below a certain reference level, steroid treatment should be either reduced or increased.

A considerable advantage of exhaled NO is that NO levels may increase before any significant changes in other parameters such as lung function and sputum eosinophils and may therefore serve as an early warning of loss of control (4). Thus, exhaled NO levels increase by 40 and 100% after 2 and 4 wk, respectively, after the reduction in steroid treatment (114). This increase in exhaled NO levels is accompanied by lung function deterioration and asthma symptoms. Although the baseline high number of eosinophils in sputum of patients who eventually develop exacerbations is a good predictor of asthma deterioration, the changes in eosinophils after the steroid reduction are slow (114). Prospective studies, which look at asthma outcomes over a prolonged period of time, where NO is used as a decision point for modifying inhaled corticosteroid treatment will be needed to evaluate the value of exhaled NO as a useful way of monitoring asthma.

Disease severity and control. Treatment with inhaled corticosteroids reduces exhaled NO levels, and therefore exhaled NO cannot be directly related to asthma severity.

Exhaled NO levels are almost three times higher in children with recent symptoms than in symptom-free subjects (122), and are further elevated during the asthma attack in both adults (123) and children (124, 125). In fact, the levels of NO in children with acute severe asthma (125) are more than 2-fold higher than in children with less severe wheezing exacerbations and almost 4-fold higher than in children with first-time wheeze (124). A reduction in exhaled NO (by 65% after 5 d of corticosteroid therapy) is accompanied by clinical and FEV1 improvement from asthma exacerbations in children (126), and NO has been a more sensitive marker of asthma activity than serum ECP or soluble interleukin-2 receptors (127). Higher exhaled NO levels are related to asthma symptoms and β2-agonist use in patients with difficult severe asthma (98). Exhaled NO is increased in patients who remain symptomatic despite oral steroids and who have a relative steroid resistance, and may therefore be useful to quantify steroid resistance in asthma.

It is most likely that exhaled NO is related to asthma control rather than to asthma severity (3), and that serial NO measurements in individual patients over time may be useful to identify patients requiring changes in therapy. In a recent study, Sippel and coworkers (128) have shown that exhaled NO was significantly correlated with markers of asthma control such as asthma symptoms within the previous 2 wk, dyspnea score, daily use of rescue medication, and reversibility of airflow obstruction. However, exhaled NO levels were not correlated with the following markers of asthma severity: history of respiratory failure, health care use, or fixed airflow obstruction.

It is reasonable to believe that subclinical airway inflammation, which is reflected by elevated levels of exhaled NO in adolescent asymptomatic patients with asthma remission (96), should be treated with corticosteroids to prevent this continuous risk of becoming clinically manifest again. However, only longitudinal studies can answer the question whether exhaled NO and bronchial hyperresponsiveness, for example, each reflecting different aspects of the inflammatory process, may guide the anti-inflammatory treatment to prevent asthma relapse later in life.

Although research in asthma has concentrated on complex proinflammatory mechanisms, it is likely that defective expression of cytokines that inhibit allergic inflammation such as interleukin 10 (IL-10), interleukin 12 (IL-12), and interferon gamma might also be important, particularly in determining disease severity and persistence of inflammation in the airways (129). Therapy based on these cytokines might also be useful, with the advantage that it restores the balance of endogenous cytokines. Recently, it has been shown that adenovirus-mediated human IL-10 gene transfer in vivo into lung isografts ameliorates subsequent ischemia-reperfusion injury and results in reduced neutrophil sequestration, and down-regulation of iNOS mRNA expression (130). Potentially, exhaled NO may be useful to monitor this type of treatment.

Relationship to other markers of asthma. The traditional means of monitoring asthma have limitations. Lung function and PC20, measurements are not directly related to airway inflammation, have little room for improvement in mild asthma (FEV1), and are affected by bronchodilators. Both parameters are slow to change and are not able to distinguish the effect of different doses of steroids. There are several areas in which exhaled NO measurements may be advantageous over the traditional means of asthma monitoring: screening for atopy, monitoring the impact of hazardous environmental factors, identification and monitoring of asthma exacerbations, and assessment of the adequacy of anti-inflammatory treatment.

Exhaled NO in patients with asthma is correlated with sputum eosinophils (117, 131, 132) and methacholine reactivity (133, 134), as well as peak flow variability (113, 116). However, the relationship between exhaled NO and airway inflammation is still uncertain, and in smaller studies no significant relationship is seen between exhaled NO and eosinophils in bronchial biopsies or bronchoalveolar lavage (116), and the induction of sputum eosinophils by inhaled LTE4 is not associated with increased exhaled NO (135, 136). This may indicate that increased exhaled NO reflects some, but not all, aspects of airway inflammation, and further work is needed to determine how it relates to some other markers of airway inflammation. On the other hand, a more comprehensive spectrum of inflammatory markers (for example, IL-4, IL-5, IL-6, IL-8, IL-10, and TNF-α) can be measured in induced sputum, and in the future these should be correlated with changes in exhaled NO.

Corticosteroids. Systemic corticosteroids have no effect on exhaled NO in normal subjects, but they decease its levels in patients with asthma (40, 50). Oral dexamethasone (4 mg/d for 2 d) similarly has no effect on exhaled NO or on serum concentrations of interferon-γ and IL-1β in normal subjects (137).

A large dose (1 mg/kg/d for 5 d) of oral prednisolone normalized exhaled NO in infants and young children with wheezing exacerbations (124), whereas the same dose in children with more severe asthma only shifted their exhaled NO down to the levels of mild-to-moderate asthma, in spite of the improvement in lung function (125). A cumulative dose of methylprednisolone (180 to 500 mg) causes 36% reduction within 50 h in the majority of severe adult patients with severe, acute asthma (40), and a combination of oral prednisolone and inhaled steroids reduces exhaled NO by 65% in children with acute asthma (126).

Recently, it has been shown that NO levels correlate with the percentage improvement in FEV1 from baseline to the poststeroid (30 mg prednisolone/d for 14 d) postbronchodilator value. A NO level of > 10 ppb at baseline has a positive predictive value of 83% for an improvement in FEV1 of ⩾ 15%, and therefore may be useful in predicting the response to a trial of oral steroid in asthma (138).

A key question is why has it been so difficult to show a dose-dependent effect of inhaled corticosteroids in the treatment of asthma? First, it is possible that the small change in doses makes it difficult to detect changes in asthma symptoms and lung function (FEV1). Secondly, the currently recommended doses may be at the upper end of the dose-response curve, making it difficult to detect a relatively small change in dose. In view of concerns about systemic effects and the better effects of adding an inhaled long-acting β2-agonist compared with doubling the dose of inhaled steroid, there is now a trend towards use of lower doses of inhaled corticosteroids. Exhaled NO as an inflammatory marker sensitive to corticosteroids may be the ideal tool to demonstrate a dose-response effect and to adjust the dose in clinical practice. It may also be useful in patients using a fixed combination inhalers (corticosteroids and long acting β2-agonist) to ensure that inflammation is controlled, as this may be difficult to assess from symptoms when a long-acting bronchodilator is taken. On the other hand, caution should be exercised as once-daily combination therapy

In fact, inhaled corticosteroids reduce exhaled NO in asthmatic patients (112) and this effect is dose-related (117). However, a plateau effect on exhaled NO measured after 6 to 12 h since the last treatment may be seen at a dose of 400 μg budesonide and higher (117, 139) in contrast to dose-related improvements in adenosine monophosphate and methacholine reactivity up to 1,600 μg in patients with mild-to-moderate asthma (120, 140). The effect of inhaled steroids on exhaled NO is very rapid and may occur within 6 h after a single high-dose (8 mg) of budesonide (Pulmicort Respules) in symptomatic moderate asthma (115). Therefore, chronic and acute reduction in exhaled NO may be of a different magnitude. Recently, it has been shown that the onset of action of inhaled BUD on exhaled NO and the time to reach the maximal reduction were also dose-dependent (120). A gradual reduction in exhaled NO is seen during the first week of regular treatment (112, 119, 120) with maximal effect between 3 wk (112, 118) or 4 wk (116, 117).

It is still uncertain whether exhaled NO is useful to direct changes in asthma therapy. Recently, it has been shown that exhaled NO values above 13 ppb had a sensitivity of 0.67 and a specificity of 0.65 to predict a step up in therapy (141), but clearly more studies are needed using exhaled NO to direct therapy.

Corticosteroids may reduce exhaled NO by directly inhibiting the induction of NOS2 (22) or by suppressing the proinflammatory cytokines that induce NOS2. There is inhibition of NOS2 immunoreactivity with inhaled corticosteroid treatment in asthmatic patients and a parallel reduction in immunoreactivity for nitrotyrosine, which may reflect local production of peroxynitrite from an interaction of NO and superoxide anions (26).

β2-agonists. Neither short-acting (112, 125, 142-145) nor long- acting (125, 139, 142, 144, 146) β2-agonists reduce exhaled NO. This is consistent with the fact that they do not have any anti-inflammatory effects in asthma, although it has been shown that regular treatment with inhaled formoterol reduces inflammatory cells in the mucosa of asthmatic patients (147). There may even be a short-term increase in exhaled NO after β2-agonists, which may be due to opening up of airways with higher local NO concentrations (148).

Antileukotrienes. The leukotriene receptor antagonist pranlukast blocks the increase in exhaled NO when inhaled corticosteroids are withdrawn (149), and montelukast rapidly reduces exhaled NO by 15 to 30% in children with asthma (150). Antileukotrienes have a moderate effect in patients with asthma and seasonal allergic rhinitis (151, 152). Both formoterol and zafirlukast were equally effective in maintaining asthma control, and zafirlukast caused a significant reduction in exhaled NO (143).

NOS inhibitors. Nebulized l-NMMA and l-NAME, which are nonselective inhibitors of NOS, both reduce exhaled NO in asthmatic patients, although this is not accompanied by any changes in lung function (50, 153). Aminoguanidine, a more selective inhibitor of NOS2, reduces exhaled NO in asthmatic patients, but it has little effect in normal subjects, indicating that NOS2 is an important source of the increased exhaled NO in asthma (51).

Prostaglandins. Prostaglandin (PG)E2 down-regulates NOS2 expression (154) and inhaled PGE2 and PGF decrease exhaled NO in normal and in asthmatic subjects (155).

Other drugs. The immunosuppressive drugs cyclosporin and rapamycin inhibit NOS2 expression (156), suggesting that exhaled NO can be used to monitor their effect. Ibuprofen, a cyclooxygenase inhibitor, reduces the elevated levels of exhaled NO in normal subjects after intravenous administration of endotoxin (157), and indomethacin partially prevents an increase in exhaled NO and asthma symptoms in patients whose dose of steroids was reduced (158). A low dose of theophylline has no effect on exhaled NO levels in asthmatic patients (159). Nebulized IL-4 receptor (altrakincept) reduces exhaled NO in patients with moderate asthma (160).

COPD

Exhaled NO levels in patients with stable COPD (80, 81, 161) and chronic bronchitis (162) are lower than in either smoking or nonsmoking asthmatics (163) and are not different from those in normal subjects. This reduction in exhaled NO is due to the effect of tobacco smoking, which down-regulates eNOS (164) and reduces exhaled NO (80), suggesting that this may contribute to the high risk of pulmonary and cardiovascular disease in cigarette smokers. In addition to the effects of cigarette smoking, a relatively low value of exhaled NO in COPD may reflect more peripheral inflammation than in asthma, low NOS2 expression (161), and increased oxidative stress that may consume NO in the formation of peroxynitrite (165).

Patients with unstable COPD, however, have high NO levels compared with stable smokers or ex-smokers with COPD (166), which may be explained by increased neutrophilic inflammation and oxidant/antioxidant imbalance. Eosinophils that are capable of expressing NOS2 and producing NO are present in exacerbations of COPD (167). Acidosis, which is frequently associated with exacerbations of COPD, may increase the release of NO (32). Pulmonary hypertension has the opposite effect, as COPD patients with cor pulmonale have low exhaled NO levels (168), which may reflect their impaired endothelial NO release.

A small proportion of patients with COPD appear to response to corticosteroids, and these patients, who are likely to have coexistent asthma, have an increased proportion of eosinophils in induced sputum (169). These patients also have an increased in exhaled NO (170). This suggests that exhaled NO may be useful in predicting which patients with COPD will respond to long-term inhaled corticosteroid treatment.

Cystic Fibrosis

Surprisingly, exhaled and nasal NO levels are significantly lower in patients with cystic fibrosis (CF) than in normal subjects, despite the intense neutrophilic inflammation in the airways (35) (Figure 3) (171) leading to the release of superoxide anions, which convert NO to nitrate and may result in the formation of peroxynitrite (172). Increased oxidative stress in CF is likely to be a consequence of this neutrophilic inflammation, malnutrition, and IL-10 deficiency (173, 174). Although there is a trend toward both exhaled and nasal NO being higher in patients who were not homozygous for the ΔF508 CF transmembrane regulator mutation (175, 176), there is no strong association between exhaled NO and disease severity in CF (176) or infection with Pseudomonas (35).

There are several possible reasons for the low levels of NO in patients with CF. First, there is a deficiency of NOS2 in patients with CF (177). Constitutive expression of NOS2, which has been demonstrated in normal human airway epithelium, and of non-CF mouse is essentially absent in the epithelium of CF airways (178). Neutrophils enhance expression of NOS2 in normal human bronchial epithelial cells but not in CF epithelial cells (179). The low expression of NOS2 would account for the low levels of NO in nasal as well as exhaled air. Secondly, an association between the length of a repeat polymorphism in the NOS1 gene and exhaled NO in patients with CF has recently been demonstrated, and exhaled NO is significantly lower in patients with CF with two alleles with a high number of repeats than in those alleles with fewer repeats at this locus (180). Interestingly, Pseudomonas aeruginosa colonization is more common in patients with CF with high numbers of repeats in the NOS1 gene, hence with lower exhaled NO.

Sexual hormones have impact on cystic fibrosis transmembrane mRNA expression (181) and it is not unusual that female patients with CF have reported worsening of lung symptoms prior to menstruation. Changes in exhaled NO during the menstrual cycle with the lowest NO levels during menstruation have been observed (66). Although NO is a weak bronchodilator and the physiologic significance of this finding is still not known, it has been shown that FEV1 was significantly lower during menstruation in female patients with CF (182).

Bronchiectasis

An increase in exhaled NO is found in bronchiectasis and the increase in NO is related to the extent of disease as measured by a computerized tomography score (183). As in asthma, the elevation of exhaled NO is not seen in patients treated with inhaled corticosteroids. This suggests that exhaled NO in bronchiectasis may reflect active inflammation in the lower airways and may be used to monitor disease activity. This is supported by increased NOS2 expression in lungs from patients with bronchiectasis (184). However, in another study, exhaled NO levels were not elevated compared with normal subjects in clinically stable patients with bronchiectasis, and it was suggested that NO is either trapped in viscous airway secretions or removed by reaction with reactive oxygen species (185).

Primary Ciliary Dyskinesia

Primary ciliary dyskinesia (PCD), including Kartagener's syndrome, is a genetic disease characterized by defective motility of cilia, in which the levels of exhaled NO are very low compared with normal subjects (186) (Figure 3) (187, 188). Such low values of exhaled and nasal NO are not seen in any other condition and are therefore of diagnostic value. Measurement of exhaled NO might be used as a screening procedure to detect PCD among patients with recurrent chest infections or male infertility caused by immotile spermatozoa, and the diagnosis of PCD is then confirmed by the saccharine test, nasal nitric oxide, ciliary beat frequency, and electron microscopy (189). NO plays an important role in bactericidal activity in the lungs, sodium and chloride transport in nasal epithelium, and ciliary beating (190), so that a lack of endogenous NO production might contribute to the characteristic recurrent chest infections in patients with PCD. Low levels of exhaled and nasal NO in patients with PCD are related to mucociliary dysfunction (186, 191), and treatment with NO donor l-arginine increases nasal NO and also improves mucociliary transport in patients with PCD (3, 186). The mechanism for such a low NO production by nasal and airway epithelia in PCD is unknown, but it might be linked to genetic abnormalities in NOS2 gene expression as in CF.

Rhinitis

The levels of NO derived from the upper respiratory tract are more than 100-fold higher than those from lower airways. This fact is mostly due to its high production in human paranasal sinuses (43), which is due to high basal activity of constitutively expressed form of NOS2 (192), and nasal NO may be significantly reduced by l-NAME in normal subjects (72), and this inhibition of NOS may induce hyperresponsiveness of the nasal airway (193). Strong NOS3- and weak NOS2-immunoreactivity are found in nasal epithelium and submucosal glands of normal subjects, but NOS2 reactivity is increased in patients with allergic rhinitis (46). There is increased immunoreactivity of nitrotyrosine in the nasal mucosa of patients with perennial rhinitis and is related to the severity of the nasal symptoms (194). However, an increased expression of iNOS is not necessarily associated with a higher 3-nitrotyrosine-labeling intensity (195), suggesting that iNOS-derived NO may have a role in the pathophysiology of rhinitis, but the production of peroxynitrite in patients with rhinitis is not dependent on the level of iNOS alone. Eotaxin causes chemotaxis of eosinophils, an increase of nitrotyrosine-immunoreactivity in nasal mucosa and increased levels of nasal NO in clinically symptomatic patients with allergic rhinitis (196). Instillation of LTB4 into the nasal segment caused a time-dependent increase in the volume of airway fluid and in the recruitment of neutrophils in dogs, and was prevented by l-NAME (197). Recently, it has been shown that the nasal decongestants oxymetazoline and xylometazoline, frequently used in the topical treatment of rhinitis and sinusitis, may have a dose dependent inhibitory effect on total iNOS activity (198).

Elevated nasal NO has been reported in allergic and perennial rhinitis (199, 200), which is reduced by treatment with nasal corticosteroids (200). Similar results are seen in children with allergic rhinitis (201). In addition, exhaled NO is also significantly elevated in allergic rhinitis in the nonpollen season and is increased further in the pollen season (202). However, the differences between the levels of nasal NO in rhinitis compared with those in normal subjects and much less marked than the differences between exhaled NO between patients with asthma and normal subjects because of the very high baseline values. This makes nasal NO less useful for diagnosis and monitoring treatment in rhinitis than exhaled NO in asthma.

Interstitial Lung Diseases

Systemic sclerosis. In patients with systemic sclerosis who have developed pulmonary hypertension, there is a reduction in exhaled NO compared with that in normal subjects and with that in patients with interstitial lung disease without pulmonary hypertension (203, 204). This may be due to reduced expression of NOS3 in pulmonary vessels, or a reduction in the pulmonary vascular endothelial surface. However, the presence of NOS3 in pulmonary vessels is variable, and it has been found to be either reduced (205-207), increased (208), variable (209), or unaltered (210).

Fibrosing alveolitis. There is strong expression of nitrotyrosine and NOS2 in macrophages, neutrophils, and alveolar epithelium in lungs of patients with idiopathic pulmonary fibrosis with active inflammation during the early to intermediate stage of the disease (211). This is consistent with elevated levels of exhaled NO in patients with fibrosing alveolitis. Increased exhaled NO levels are associated with disease activity, as assessed by BAL lymphocyte counts, and are reduced in patients treated with corticosteroids (212).

Sarcoidosis. Cytokines, including TNF-α and interferon-γ, are increased in the pulmonary inflammation of sarcoidosis and there is an up-regulation of NOS2 in respiratory epithelium and granulomata in patients with sarcoidosis (213). The magnitude of the rise in exhaled NO in sarcoidosis may be related to the activity of the disease and is reduced by steroid therapy. This is, perhaps, the reason behind two conflicting observations reporting either elevated (213) or normal (214) exhaled NO in patients with active pulmonary sarcoidosis.

Pulmonary Hypertension

The pathogenesis of pulmonary hypertension remains poorly understood. Vasoconstriction is likely to be a major factor in the initial stages of the disease, and a reduction in endogenous NO may contribute to the development of pulmonary hypertension. In fact, nebulized epoprostenol increased exhaled NO in patients with pulmonary hypertension, but not in normal control subjects, suggesting that this effect on the hypertensive circulation has a NO-related mechanism (215). In contrast, the angiotensin-converting enzyme (ACE) inhibitor enalapril, used to treat pulmonary hypertension, increases exhaled NO levels in normotensive subjects, but not in patients with systemic hypertension (216).

Biochemical reaction products of NO are inversely correlated with pulmonary artery pressures in patients with primary pulmonary hypertension and with years since the diagnosis (217). This may reflect reduced expression of NOS3 in patients with pulmonary hypertension, as reduced NOS3 expression has been reported in patients with primary pulmonary hypertension (205-207). In fact, aerosolized NOS2 gene transfer increases pulmonary NO production and reduces hypoxic pulmonary hypertension in rats (218) and may be a promising future strategy to target pulmonary vascular disorders.

However, interpretation of these low NO levels should be made cautiously and in the context of potential influence of Hb on NO. Although stimulation of NO production by pulmonary vascular endothelial cells in response to shear stress has been described, it is not an important determinant of NO production. Low exhaled NO in patients with pulmonary hypertension may be consistent with flow redistribution from alveolar septal capillaries to extra-alveolar vessels and decreased surface area or a direct, stretch-mediated depression of lung epithelial NO production (219), or increased Hb NO scavenging. It may be difficult to use exhaled NO changes as an accurate measure of lung tissue NO production.

Occupational Diseases

Allergens from rats, mice, guinea pigs, or rabbits cause as much as 30% of exposed persons to develop specific immunoglobulin E (IgE) responses. Laboratory animal allergy (LAA) is among the highest occupational risks for asthma. Exhaled NO is raised in subjects with LAA symptoms and correlates with symptom severity (97). The progressive increase in exhaled NO from asymptomatic to early LAA to symptomatic asthma suggests that exhaled NO measurements may be useful in monitoring occupational asthmas, and of environmental health effects of air pollution (220) in epidemiologic surveys. Recently, measurement of exhaled NO and induced sputum were evaluated in occupational asthma. Aluminum potroom workers (exposure to dust and fluorides) with asthmalike symptoms had higher concentrations of exhaled NO than did those with no symptoms (221), suggesting that exhaled NO may be an early marker of airway inflammation in potroom workers. High levels of exhaled NO and asthmalike symptoms in subjects with occupational exposure to high levels of ozone and chlorine dioxide (78), or in swine confinement workers (162), may indicate the presence of chronic airway inflammation. Latex sensitivity is an increasing problem among healthcare workers. Although allergen challenge with natural rubber latex increased exhaled NO levels after 22 h in some subjects with suspected occupational asthma (222), further studies are needed to demonstrate a clear relationship between exhaled NO and routine latex workplace exposure.

Infections

NO may play an important role in nonspecific host defenses against bacterial, viral and fungal infections. One of the general mechanisms of antimicrobial defenses involving NO is S-nitrosylation by NO of cysteine proteases, which are critical for virulence, or replication of many viruses, bacteria, and parasites. The reduced endogenous NO production, resulting in low exhaled and nasal NO levels, may contribute to recurrent chest infections in patients with PCD or CF, as discussed above. Low nasal NO is associated with colonization of the upper respiratory tract with Staphylococcus aureus in active Wegener's granulomatosis (223).

Viral infections. Exhaled, but nasal, NO is elevated during viral infections in adults and in children (84, 86). Exhaled NO is also increased in experimental human influenza (224) and rhinovirus infection (225). The increase in NO production during viral infection is likely to be protective, as NO inhibits virus replication either by inhibiting viral RNA synthesis, or/ and by S-nitrosylation of the cysteine proteases that are critical for virulence and replication of viruses (226). Viral infection may also induce the expression of NOS2 via activation of NF-κB and other transcription factors (227). Exhaled (228) and nasal NO (229) in HIV positive patients is less than in control subjects, and NO synthesis is further depressed in terminally ill patients with HIV (230), suggesting that low NO may indicate a mechanism of impaired host defense in HIV infection. This may be explained by an inhibitory role of the HIV type 1 regulatory protein Tat on NOS2 activity in a murine macrophage cell line (231).

Tuberculosis. NO plays an important role in resistance to Mycobacterium tuberculosis infection, and exposure of extra-cellular M. tuberculosis to < 100 ppm of NO for a short period (< 24 h) results in microbial killing (232). Elevated exhaled NO and NOS2 expression in alveolar macrophages is found in patients with active tuberculosis and is reduced with antituberculosis therapy (233).

Bacterial infections. Nitrate concentrations are significantly higher in BAL in immunosuppressed children with pneumonia than in normal control subjects (234), and elevated exhaled NO levels are found in patients with lower respiratory tract inflammation and chronic bronchitis (162).

Chronic Cough

Increased levels of exhaled NO do not accompany all forms of airway inflammation. Patients with chronic cough that is not attributable to asthma have lower NO values than do healthy volunteers and patients with asthma (88, 134), including those with cough caused by gastroesophageal reflux (235). Measurement of exhaled NO may therefore be a useful screening procedure for patients with chronic cough and would readily identify those patients with cough caused by asthma (88).

Lung Cancer

The levels of nitrite in epithelial lining fluid and exhaled NO are significantly higher in patients with lung cancer than in control subjects, and they are correlated with the intensity of NOS2 expression in alveolar macrophages (236). The level of nitrite was also significantly higher in epithelial lining fluid from patients with cancer, but the increased NO production is not specific to the tumor side and might be attributed to a tumor-associated nonspecific immunologic and inflammatory mechanism.

Lung Transplant Rejection

Monitoring endogenous NO release may be useful in lung transplantation. Loss of endogenous production of NO by cadaver lung allografts in the perioperative period (237), and the fact that reduced exhaled NO after hypoxia-reoxygenation might reflect bronchial epithelial dysfunction (238), may provide a rationale for interventions to restore NO production and, therefore, to improve the outcome of the surgery. The development of postlung transplant obliterative bronchiolitis is the commonest cause of late graft failure and is characterized by intense airway inflammation and high exhaled NO, which are higher than in either control subjects or stable lung transplant recipients (239). In stable lung transplant recipients, exhaled NO concentrations are highly dependent upon the severity of BAL neutrophilia and the intensity and extent of expression of NOS2 in the bronchial epithelium, but not in the subepithelial area (240). This suggests that serial exhaled NO measurements may have a role in the early detection of obliterative bronchiolitis (240) or of acute rejection (241).

Adult Respiratory Distress Syndrome

Adult respiratory distress syndrome (ARDS) is associated with a neutrophilic alveolar inflammation. In animal models of ARDS induced by endotoxin there is increased production of NO (242). Exhaled NO values are low, presumably because of the concomitant oxidative stress and consumption of NO by superoxide anions to form peroxynitrite (243). Association of reduced exhaled NO levels with the increases in pulmonary artery pressure and alveolar-arterial oxygen pressure and the decrease in lung compliance (244) suggests that exhaled NO may be an indicator of lung injury in adult patients after cardiopulmonary bypass.

Diffuse Panbronchiolitis

Diffuse panbronchiolitis (DPB), a pulmonary disease of unknown origin with chronic inflammation in the respiratory bronchioles leading to chronic chest infections resulting from mucociliary dysfunction, is the third disease (after primary ciliary dyskinesia, and cystic fibrosis) with diagnostically low nasal NO levels (245). Airway impaired NOS activity may be involved in its pathogenesis, and NO measurements may serve as a noninvasive test in the diagnosis of DPB.

Carbon monoxide (CO) is a gas that may be formed endogenously and is detectable in exhaled air.

Source of Exhaled CO

There are three major sources of CO in exhaled air: enzymatic degradation of heme, non–heme-related release (lipid peroxidation, xenobiotics, bacteria) and exogenous CO. The predominant endogenous source of CO (∼ 85%) in the body is from the degradation of hemoglobin by the enzyme heme oxygenase (HO), and approximately 15% arises from degradation of myoglobin, catalase, NO synthases, guanylyl cyclase and cytochromes (246). Several bacteria produce CO (247), but this does not play an appreciable role in the turnover of CO that is inhaled or endogenously produced. Approximately 85% of the CO in the body is bound to hemoglobin in circulating erythrocytes and the remaining 15% is bound to other compounds (such as myoglobin) or in tissues, and less than 1% is unbound and dissolved in body fluid (248). Approximately 80% of the CO formed from heme degradation is exhaled (249). CO uptake or excretion across the skin is minimal, except in premature infants, and the amount of CO consumed by the tissues is very small (3% of the rate of endogenous CO production) (250).

There are several reasons to consider that the alveoli are the predominant site of exhaled CO in normal subjects. First, levels of exhaled CO measured at the end of exhalation are similar to those measured via a bronchoscope at the level of main bronchus (251). Second, exhaled CO levels are less flow- or breathhold-dependent than exhaled NO (252), suggesting less airway contribution. Third, maximal CO levels are seen close to the end of exhalation, as for CO2. There is also a small proportion of CO derived from the airways, which is higher after allergen challenge measured either via bronchoscope (251), or at the mouth (104). The fact that breathing through the nose increases the CO levels obtained in the exhaled air (253) suggests that nose and paranasal sinuses may also contribute to the CO production of the human airways. Indeed, HO-like immunoreactivity is seen in the respiratory epithelium, in connection with seromucous glands and in the vascular smooth muscle of the nose (253).

Heme oxygenase. CO is a by-product of rate-limited oxidative cleavage of hemoglobin by HO, which exists in three isoforms, i.e., HO-1, HO-2, and HO-3. HO-2 is constitutively expressed in most tissues, whereas HO-3 is, so far, only described in rats (254). HO-1 has been identified as the major 32 kD heat shock (stress) protein (255). Like other stress proteins HO-1 can be induced by a variety of stimuli, such as proinflammatory cytokines, bacterial toxins, heme, ozone, hyperoxia, hypoxia, reactive oxygen species, and reactive nitrogen species. Both HO-1 and HO-2 are expressed in human airways and are found in most cell types, with particularly strong immunfluorescence in airway epithelial cells (256). Heme is converted by HO to biliverdin and thence to bilirubin, with the formation of CO and ferritin (Figure 4).

Interactions with NO. Like NO, CO is also capable of up-regulating cyclic guanine monophosphate (cGMP) via activation of guanylyl cyclase causing vasodilatation, smooth-muscle relaxation, and platelet disaggregation. The vasodilatory effect of CO may be important in maintaining adequate tissue oxygenation and perfusion in the lung during normal physiology and in hypoxic conditions that result from pulmonary vascular diseases and acute lung injury. It has been suggested that the HO pathway exerts important counter-regulatory effects on the NOS pathway and, when blocked, the underlying NOS pathway is unmasked leading to increased and prolonged release of NO (257). In contrast, exogenously administered or endogenously released NO stimulates HO-1 gene expression and CO production in vascular smooth muscle cells resulting in a higher resistance to oxidant damage (258). This effect of NO is related to the release of free heme from heme proteins, which are able to transcriptionally up-regulate HO-1 and lead to their own degradation. CO also directly inhibits NOS2 activity by binding to the heme moiety of the enzyme (259). The effect of hemoglobin scavenging, as a function of the extent of bronchial arterial neovascularization (e.g. bronchiectasis, thromboembolic disease) may play an important role in the reaction between erythrocytic hemoglobin and NO. This interaction has been generally considered in the context of mechanisms that safely detoxify NO. More recently, hemoglobin-dependent mechanisms that preserve, not destroy, NO bioactivity in vivo have also been proposed (260). The emerging picture suggests that the interplay between NO and erythrocytic hemoglobin is important in regulating the functions of both these molecules in vivo. Hemoglobins modified for therapeutic use as either hemoglobin-based oxygen carriers or scavengers of nitric oxide are currently being evaluated in clinical trials. One such product, pyridoxalated hemoglobin polyoxyethylene conjugate (PHP), is a human-derived and chemically modified hemoglobin that has been successfully studied in Phase II clinical trials, and may be used for the treatment of shock associated with the systemic inflammatory response syndrome (261). The redox activity of modified hemoglobins can be attenuated, so that modified hemoglobins containing endogenous antioxidants such as PHP may have reduced pro-oxidant potential. These antioxidant properties, in addition to the NO-scavenging properties, may allow the use of PHP in other indications in which excess NO, superoxide, or hydrogen peroxide is involved, including severe asthma, CF, COPD, and bronchiectasis.

Effect of oxidative stress. There is a close link between the reactive oxygen and nitrogen species and CO. Thus, a dose-dependent increase in exhaled CO has been shown after a 1-h exposure to different concentrations of O2 (262). HO-1 activation can be diminished by N-acetylcysteine, a precursor of glutathione with antioxidant properties (263). Both, superoxide anions and peroxynitrite can stimulate HO-1 activation (264), and subsequent release of CO is an important negative-feedback regulatory mechanism limiting the release of these cytotoxic substances (265). Animals exposed to a low concentration of CO exhibit a marked tolerance of the lungs to lethal concentrations of hyperoxia in vivo (266).

The precise mechanisms for this protection are not fully understood, but both the degradation of heme (with removal of iron and induction of ferritin) and the generation of bilirubin (an antioxidant) may be involved. There is evidence that the deleterious effects of ROS, such as superoxide and H2O2, are dependent on the presence of iron. The intracellular pool of free iron can react with both H2O2 and superoxide, giving rise to the OH radical via the Fenton reaction. The free iron that is not metabolized intracellularly sequestered in cells as ferritin. Thus, ferritin serves as a reservoir to restrict iron from participating in the Fenton reaction. It has been shown that free iron released from heme by HO may induce ferritin synthesis, and heme-induced HO-1 protein also activates ferritin via mRNA expression (267). Furthermore, the metabolite of heme degradation, bilirubin, is itself an effective antioxidant of peroxynitrite-mediated protein oxidation and may be even more effective than vitamin E in preventing lipid peroxidation (268). Moderate overexpression of HO-1 improves the resistance of cells to oxygen toxicity (269). However, there is cytotoxicity associated with HO-1 overexpression.

HO-2 may also protect against oxidative stress. HO-2 knockout mice are sensitized to hyperoxia-induced oxidative injury, have a higher mortality, and increased lung iron content without increased ferritin, suggesting accumulation of available redox-active iron (270).

Measurement

Exhaled CO as a marker to assess different diseases (cardiovascular, diabetes, and nephritis) was first described in Russia 1972 (271). Over the last 20 yr exhaled CO has been measured to identify current and passive smokers, to monitor bilirubin production, including hyperbilirubinemia in newborns, and in the assessment of the lung diffusion capacity.

CO can be quantified by a number of different techniques. Most of the measurements in humans have been made using electrochemical CO sensors. The sensor is selective, gives reproducible results (272), and is inexpensive. However, these instruments are susceptible to interference from a large number of substances, for example, hydrogen, which is present in exhaled breath and may be increased after glucose ingestion. H2-insensitive CO sensors, which are now available, are therefore recommended.

Exhaled CO can also be measured (at ppb level) by adjustable laser spectrophotometer (262, 273), or by a near-infrared CO analyzer (274). Near-infrared instruments, are used for continuous monitoring of atmospheric CO, and are fairly sensitive and stable. However, they are larger than electrochemical CO sensors, sensitive to water and CO2 concentrations, and require large sample volumes (275). This may explain the low CO levels detected by these instruments even after a prolonged breathhold time of 20s (274). Gas chromatography is a reference method for CO measurements, but its use is limited to specialized laboratories.

End-tidal exhaled CO measurements can be made during a single exhalation and is a routine in cooperative adults. It can also be easily performed in children older than 5 yr of age (276). A method for measuring CO in nasally sampled exhaled air in noncooperative neonates has been developed that involves the relatively noninvasive placement of a small catheter into the posterior of the nasopharynx and the collection of breath samples either manually or automatically (249).

Factors Affecting Exhaled CO Measurements

CO exists in the atmosphere as a by-product of incomplete combustion and oxidation of hydrocarbons, and is oxidized to CO2 by hydroxyl radicals, or eliminated either by soil microorganisms or by stratospheric diffusion. Regional and local levels of CO in ambient air can vary significantly depending on time of the day and season, on wind velocity, industrialization, traffic, and altitude. Although some exposure to CO may occur in normal day-to-day life because of environmental pollution, active or passive smoking are the most likely reason for high levels of exhaled CO. After inhalation, CO displaces oxygen in the erythrocyte to form carboxyhemoglobin (COHb), which has a half-life of about 5 to 6 h in this form. A cutoff level of 6 ppm (277) effectively separates nonsmokers from smokers, and the previously used cutoff 8 ppm (278) or 10 ppm (279) may be too high. Other individual factors, which can markedly affect the amount of CO that a person may inhale, are type and location of home and occupation, cooking/ heating appliances, and mode of transportation.

Many pathologic conditions and factors can increase the rate of hemoprotein breakdown and potentially increase the levels of exhaled CO, including anemias, hematomas, and preeclampsia. Nonpathologic factors may also increase endogenous CO production, including fasting, dehydration, some drugs (phenobarbitone), and xenobiotic compounds (paint remover) (280) (Table 2).

Table 2.  FACTORS INFLUENCING EXHALED CO

Exhaled COReference
Miscellaneous
↑ Smoking (277, 465)
↑ Airway pollution(466, 467)
↑ Airway obstruction (468)
↑ Hyperbilirubinemia(469)
Sex (cyclic variations in women)(470)
Race (↑ COHb in Japanese newborn)(471)
Disease
↑ Allergen challenge (early and late response) (104)
↑ Asthma (mild-moderate)(281, 282, 284)
↔ Asthma (mild)(285)
↑ Asthma (severe)(285)
↑ Atopy(101)
↑ Asthma in children (persistant asthma)(276)
↑ Allergic rhinitis(297)
↑ COPD (ex-smokers)(288)
↑ Upper respiratory tract infections(276, 298)
↑ Bronchiectasis and lower respiratory tract infections(290, 291)
↑ Interstitial lung disease(295)
↑ CF(176, 292–294)
↑ Citically ill patients (299)

Definition of abbreviations: ↓ = decrease; ↑ = increase; ↔ = no change.

Asthma

Elevated levels of exhaled CO have been reported in stable asthma (281, 282) with normal levels in patients treated with inhaled corticosteroids (282). The difference in exhaled CO between normal and asthmatic subjects, however, is much less than in exhaled NO (283), and the effect of inhaled steroids on exhaled CO in patients with mild asthma, as it has been reported recently, is negligible (256). Both HO-1 and HO-2 are extensively distributed in airways of normal and asthmatic subjects (256). The increased levels in stable asthma are likely to be due to preferential increase of HO-1 expression, which is seen in alveolar macrophages in induced sputum of patients with asthma (263). There is also an increase in the concentration of bilirubin in induced sputum, indicating increased HO-1 activity (263). Further evidence that exhaled CO increases may reflect HO activity is the demonstration that inhaled hemin, which is a substrate for HO, results in a significant increase in exhaled CO concentration in normal and asthmatic subjects (263). Increased levels of exhaled CO are seen in acute exacerbations of asthma, and are reduced after treatment with oral corticosteroids (284). Significantly elevated CO levels are found in patients with severe asthma (285), including patients treated with 30 mg of prednisolone for 2 wk (286). In view of the simplicity of CO measurements and the portability of CO analyzers, exhaled CO may be useful in noninvasive monitoring of pediatric asthma. For example, children with persistent asthma despite treatment with steroids, which reduce their NO levels, have significantly higher exhaled CO than do those with infrequent episodic asthma (276).

COPD

A major limitation of exhaled CO in COPD is the marked effects of cigarette smoking, which masks any increase that may occur because of the disease process. There is no difference in exhaled CO in patients with chronic bronchitis (without airflow obstruction) when compared with normal subjects (287). However, exhaled CO levels are elevated in ex-smoking patients with COPD (288), suggesting ongoing oxidative stress or inflammation. HO is induced in fibroblasts exposed to cigarette smoke (289). There is an increase in exhaled CO during acute exacerbations of COPD, with a decline after recovery (290).

Bronchiectasis

Exhaled CO levels are elevated in patients with bronchiectasis, irrespective of whether they are treated with inhaled corticosteroids (291).

Cystic Fibrosis

In contrast to NO, exhaled CO levels were markedly elevated in patients with stable CF (292-294) and increased further during exacerbations and reduced with antibacterial treatment (Figure 5) (176). This suggests that exhaled CO is not only a marker of oxidative stress/inflammation in CF, but is also a marker of disease severity. This is further confirmed by the finding of lower CO levels in patients receiving oral corticosteroid treatment (292-294). In fact, by reducing airway inflammation and the release of oxidants by inflammatory cells steroids may attenuate HO-1 expression and the synthesis of CO. We have shown that patients homozygous for the CF transmembrane regulator ΔF508 mutation have higher exhaled CO levels than do heterozygous patients (292). Considering the growing interest in gene therapy in cystic fibrosis, further studies are needed to investigate the role of CO levels in the assessment of effective therapeutic gene delivery or to confirm the diagnosis in patients with borderline sweat tests where more extensive genetic analysis is not available.

Interstitial Lung Disease

Elevation of exhaled CO is related to lung function deterioration (295) and impaired gas transfer in patients with cryptogenic fibrosing alveolitis and scleroderma (296). Elevated levels of exhaled CO in patients with fibrosing alveolitis are also associated with disease activity as assessed by BAL cell counts (212). This suggests that exhaled CO may be used to monitor disease progression and response to therapy in interstitial lung diseases.

Allergic Rhinitis

Stable levels of CO are recorded during continuous sampling from one nostril during normal breathing through the mouth in normal subjects (253). Sampling through a drainage tube inserted into the maxillary sinus reveals CO levels comparable to the levels obtained by sampling through the nose. In patients with allergic rhinitis exhaled CO is increased during the pollen season and returns to normal values after the season (297). The levels of exhaled CO are significantly higher in patients with symptoms than in those without. However, there is no correlation between nasal and exhaled samples, suggesting that the increase is derived from the lower respiratory tract. We did not measure any direct nasal CO production in either normal or asthmatic subjects (283).

Infections

HO-1 is induced by many infectious agents, and HO-1 may provide protection to cells against attack by infectious agents. Upper respiratory tract viral infections may induce the expression of HO-1, resulting in increased exhaled CO in adults (298) and in children (276). Elevated exhaled CO levels might provide an early warning signal for an acute infective episode, which may lead to exacerbation of asthma and COPD. Elevated levels of CO have been measured in patients in general practice with lower respiratory tract infection, which were significantly reduced after 5 d of treatment with antibiotics (290).

Other Conditions

Critically ill patients have a significantly higher CO concentration in exhaled air as well as total CO production than do healthy control subjects (299), but inspired oxygen concentration has to be measured, as it can influence CO excretion in mechanically ventilated patients (300). Interestingly, the levels of exhaled CO in these patients are similar to the levels seen in severe asthma and may be a reflection of systemic rather than the local oxidative stress.

Exhaled CO levels are also increased in diabetes, and the level is significantly related to the level of hyperglycemia (301). The mechanism is unclear, but hyperglycemia and oxidative stress in uncontrolled diabetes may activate HO-1.

Almost 30 yr ago Pauling and co-workers (302) reported that normal human breath contains a mixture of several hundred volatile organic compounds. Exhaled hydrocarbons have been measured in a variety of conditions, ranging from the monitoring of lipid peroxidation in cosmonauts during long-term space flights (303) to patients undergoing cardiopulmonary bypass operations (304). Hydrocarbons are non-specific markers of lipid peroxidation, which is one of the consequences of the constant and inevitable formation of oxygen radicals in the body. During the process of peroxidation of polyunsaturated fatty acids hydrocarbons are distributed in the body, partly metabolized, and excreted in the breath, making it possible to estimate the magnitude of in vivo lipid peroxidation. Numerous methods have been developed to measure lipid peroxidation products and lipid peroxidation damage in tissues, cells, and body fluids. For volatile organic compounds, sampling and analysis of breath is preferable to direct measurement from blood samples because it is noninvasive, and the measurements are much simpler in the gas phase than in a complex biologic fluid. Recently, in patients with abnormal chest radiographs, a combination of 22 volatile organic compounds discriminated patients with and without lung cancer (305), suggesting that exhaled breath profile of hydrocarbons may be more informative than single hydrocarbons.

Origin

In contrast to the predominantly airway source of exhaled NO, hydrocarbons are representative of blood-borne concentrations through gas exchange in the blood/breath interface in the lungs. The main source of exhaled hydrocarbons in the body is the liver (306), with contribution from red blood cells and other organs (307). The low molecular mass hydrocarbons ethane and pentane are among the numerous end-products of lipid peroxidation of peroxidized polyunsaturated fatty acids, and have been extensively studied in exhaled breath. However, the primary localization of their generation it is not yet clear. Hydrocarbons such as propane and butane (products of peroxidation of linoleic and arachidonic acid) are mainly derived from protein oxidation and fecal flora and their role as the markers of lipid peroxidation is doubtful. However, ethane and pentane excretion are increased during the first few days of life in premature newborns when the gut is not colonized and, therefore, supporting that the bacterial flora is not the major contributor of these exhaled hydrocarbons (308). The available evidence suggests that peroxidation of polyunsaturated fatty acids is the major, if not the only, endogenous source of the pentane and ethane in breath (307).

Measurement

The first reports of exhaled breath analysis using gas chromatography go back more than 30 yr (309), and since then lipid peroxidation has gained increasing interest as one of the more prominent features of free-radical-induced damage in clinical medicine. Lipid peroxidation is assessed by measuring its secondary reaction products such as chemiluminescent and fluorescent molecular products, lipid hydroperoxides, conjugated dienes, aldehydes, malonaldehyde or thiobarbituric acid-reactive substances, and aliphatic hydrocarbons (307). Exhaled hydrocarbons are measured by gas chromatography. There are several technical difficulties that should be overcome to obtain reliable measurements. Sample preparation and storage were major problems in earlier methods for breath analysis. Recently, avoidance of air contamination, adequate preinjection concentrations of the samples, and sensitive gas chromatographic techniques have enabled more accurate and reproducible measurements of hydrocarbons in human breath (310-312). New techniques have been also developed for analyzing small volumes of gas (ethane, pentane) from single-breath samples, in which no preconcentration is required (313) and exhaled air, collected during a single flow-controlled exhalation into a Teflon reservoir, is injected directly into gas chromatograph (294, 314, 315). Particular attention should be paid to the storage (no longer than 48 h in Tedlar bags and 24 h in capped desorption tubes). Measurement of exhaled pentane is more problematic than ethane, as it exists in the ambient air and is coeluted with isoprene, a prominent hydrocarbon that is affected by diet (316). The presence of specific compounds, for example, ethane and pentane in the breath, may also be an indicator of recent exposure to the gases. The contamination of ambient ethane can be eliminated by discarding the dead space during the first part of exhalation, and potential loss of organic vapors to condensed water is excluded by using silica gel granules in reservoirs (294, 314, 315, 317). A simple field method for sampling benzene in end-exhaled air of healthy subjects has been developed, where the sample is collected directly on an adsorbent tube while the subject exhales through the sampling device, which consists of a modified peak expiratory meter (318). Exhaled breath condensate is another source of the lipid fatty-acids, which can be analyzed by gas chromatography, as described in children with pneumonia (319, 320).

Factors Affecting Levels in Exhaled Air

Hydrocarbons are present in ambient air, inhaled and retained in steady state in the equilibrium between various body compartments (body fat) and ambient air. Although it takes a few minutes to wash out the lungs, it requires no less than 90 min to wash out the body stores of hydrocarbons, making this approach impractical (321). To make exhaled breath hydrocarbon tests usable in clinical medicine two approaches to deal with ambient contamination have a considerable advantage (307). First employment of a washout period (4 to 10 min), and second to record the local ambient levels of hydrocarbons and subtract them from the levels in exhaled breath (294).

Newborns excrete ethane 11-fold and pentane 12-fold more than healthy adult men (322). The elimination of hydrocarbons is mainly (85%) the results of metabolism by hepatic cytochrome P450 enzymes (323). Therefore, large doses of ethanol, or other liver toxic agents (acetone) may increase pentane in exhaled air because of P-450 inhibition. Exhaled pentane in infants receiving total parenteral nutrition, including intravenous lipid emulsion, is 70 times higher than in adults (322). However, no statistically significant changes in exhaled hydrocarbons are found relative to the fasting level, suggesting that diet does not alter ethane or pentane excretion in healthy subjects (324).

Although different minute volumes has no effect on ethane excretion in children (325), the diffusion rate of lipophilic substances such as ethane and pentane may be reduced and will require a longer exhalation, or collection of the last part of exhalation (294). Smoking increases exhaled ethane and pentane. This effect is possibly related to oxidative damage caused by smoking and to high concentrations of hydrocarbons in cigarette smoke (326-328). Both mental (329) and physical stress (330) also increases lipid peroxidation levels of ethane and pentane in exhaled air of normal subjects.

Oxidative stress causing cell damage and lipid peroxidation plays an important role in several inflammatory lung diseases such as COPD, asthma, CF, and interstitial lung disease. Exhaled hydrocarbons may help to estimate the magnitude of in vivo lipid peroxidation by measuring, for example, pentane and ethane exhaled in breath, and to monitor the effect of novel drugs with antioxidant properties in clinical practice.

Asthma

Exhaled pentane is elevated during acute asthma exacerbations and reduced to normal levels during recovery (331). Exhaled ethane levels are higher in patients with mild steroid-naı̈ve asthma compared with steroid-treated patients and normal subjects (332) (Figure 6). The measurements of two different exhaled markers, NO and pentane for example, might be helpful to distinguish severe nocturnal asthma from obstructive sleep apnea, which is associated with low levels of circulating nitrite/nitrate (333). Elevated levels of exhaled and nasal NO, but not pentane, have been found in patients with sleep apnea (334), suggesting the presence of predominantly upper airway inflammation in these patients.

COPD

Pentane (335) and isoprene (336) are increased in normal smokers (328), and ethane in patients with COPD who smoke (327) (Figure 6). Although vitamin E given for 3 wk failed to reduce exhaled ethane in cigarette smokers, those whose ethane values fell the most tended to have better-preserved lung function (337). Increased levels of volatile organic compounds in exhaled breath could be used as biochemical markers of exposure to cigarette smoke and oxidative damage caused by smoking. For example, levels of 2,5-dimethyl furan (338), or known carcinogen benzene (339), in smokers are sufficiently discriminative to differentiate smokers from nonsmokers. However, if transient elevation of ethane in exhaled air (returned to baseline within 3 h) in healthy smokers is due to ethane in cigarette smoke, chronically elevated ethane levels in current and, especially in ex-smokers, is more likely related to oxidative damage (326). In fact, there is a correlation between the ethane levels and the degree of airway obstruction in COPD (327), and current (packs per day) and lifelong (pack-years) tobacco consumption (328). Breath analysis, therefore, may also be employed to evaluate the elimination process of a variety of volatile organic compounds after microenvironmental exposures, and an improved portable breath measurement method has been successfully tested (340).

Cystic Fibrosis

Patients with CF have elevated levels of exhaled ethane, which is significantly correlated with exhaled CO and airway obstruction (294) (Figure 6), supporting the view that oxidative stress and lipid peroxidation are increased in the airways of patients with CF.

Other Lung Diseases

Exhaled breath profile of different hydrocarbons may be of diagnostic value in a variety of clinical conditions, as it has been shown in patients with lung cancer (305). Simultaneous pentane and isoprene measurements have been measured in critically ill mechanically ventilated patients (341). In patients who developed pulmonary infection, pentane elimination was increased, but isoprene elimination was reduced, resulting in a significant increase in their ratio when compared with patients without pulmonary infection. A significant increase of exhaled ethane, which is related to a lower cardiac index and a higher systemic vascular resistance, has been demonstrated in patients undergoing cardiopulmonary bypass operations (304), suggesting oxidative damage caused by reperfusion in these patients. A potentially important application for exhaled hydrocarbons analysis would be to differentiate patients with viral and bacterial infection to justify the use of antibiotics. Elevated levels of pentane are found in critically ill patients who develop chest infection compared with patients without pulmonary infection, and they might be an indication for antibiotic treatment (341). It might even be possible, in the future, to identify a specific pathogen, hence to apply the most appropriate antibiotic therapy by studying the patients' exhaled hydrocarbon profiles.

The detection of nonvolatile mediators and inflammatory markers from the respiratory tract involves invasive techniques such as bronchoalveolar lavage or induced sputum. They cannot be repeated within a short period of time because of their invasiveness, and because the procedures themselves may induce an inflammatory response (2, 342). Exhaled breath condensate is collected by cooling or freezing exhaled air and is totally noninvasive. The collection procedure has no influence on airway function or inflammation, and there is accumulating evidence that abnormalities in condensate composition may reflect biochemical changes of airway lining fluid. Several nonvolatile chemicals, including proteins, have now been detected in breath condensates. The first studies identifying surface-active properties, including pulmonary surfactant, of exhaled condensate were published in Russia in the 1980s (343, 344) and since then several inflammatory mediators, oxidants, and ions have been identified in exhaled breath condensates.

Origin

Potentially, condensate measurements reflect different markers and molecules derived from the mouth (oral cavity and oropharynx), tracheobronchial system, and alveoli, and their proportional contribution has not yet been sufficiently studied. It is assumed that airway surface liquid becomes aerosolized during turbulent airflow, so that the content of the condensate reflects the composition of airway surface liquid, although large molecules may not aerosolize as well as small soluble molecules. A strong correlation between the levels of CO2 and O2 in exhaled fluid and exhaled breath (345) suggests that aerosol particles exhaled in human breath reflect the composition of the bronchoalveolar extracellular lining fluid.

Factors Affecting Measurements

Several methods of condensate collection have been described. The most common approach is to ask the subject to breathe tidally via a mouthpiece through a non rebreathing valve in which inspiratory and expiratory air is separated (Figure 7). During expiration the exhaled air flows through a condenser, which is cooled to 0° C by melting ice (346), or to −20° C by a refrigerated circuit (347), and breath condensate is then collected into a cooled collection vessel. A low temperature may be important for preserving labile markers as lipid mediators during the collection period, which usually takes between 10 to 15 min to obtain 1 to 3 ml of condensate. Exhaled condensate may be stored at −70° C and is subsequently analyzed by gas chromatography and/or extraction spectrophotometry, or by immunoassays (ELISA).

Salivary contamination may influence the levels of several markers detectable in exhaled breath condensate. Thus, high concentrations of eicosanoids (thromboxane B2, LTB4, PGF), but low levels of PGE2 and prostacyclin have been found in saliva of children with acute asthma (348). The presence of high concentrations of nitrite/nitrate from the diet may affect NO-related markers in condensate (349). It is therefore important to minimize and monitor salivary contamination. Subjects should rinse their mouth before collection and to keep the mouth dry by periodically swallowing their saliva. Salivary contamination, measured by amylase concentration of condensate, should be routinely monitored. In most of the studies reported, amylase has been measured in condensate and no salivary contamination has been detected (347, 350, 351). Subjects should wear a noseclip in order to collect only mouth-conditioned exhaled air into the collection system. Flushing the nose with helium may help to reduce contamination of exhaled breath with nasal air that contains high levels of NO, which potentially may influence the results of NO-related markers (nitrite/nitrate, S-nitrosothiols) (352).

Another approach to exclude nasal contamination is to collect condensate during a series of exhalations against a resistance (352). However, it has not yet been shown that nasal NO affects measurements in exhaled condensate. The quantity of exhaled condensate is dependent on the ventilation volume per unit time (minute volume), but this does not affect the concentration of mediators (346, 353). It is also dependent on exhaled air temperature and humidity (Paredi P. et al.: unpublished observation).

Hydrogen Peroxide

Activation of inflammatory cells, including neutrophils, macrophages, and eosinophils, result in an increased production of O2 , which by undergoing spontaneous or enzyme-catalyzed dismutation lead to formation of H2O2. As H2O2 is less reactive than other reactive oxygen species, it has the propensity to cross biologic membranes and enter other compartments (354). Because it is soluble, increased H2O2 in the airway equilibrates with air (355). Compared with the cellular antioxidant scavenging systems, the extracellular space and airways have significantly less ability to scavenge reactive oxygen species (356, 357). Catalase is the major enzyme involved in removing H2O2 and is preset in low concentrations in the respiratory tract. Thus, exhaled H2O2 has potential as a marker of oxidative stress in the lungs.

Asthma. H2O2 has been detected in exhaled condensate in healthy adults and children with increased concentrations in asthma (350, 355, 358, 359). There is no correlation between the levels of exhaled H2O2 and age, sex, or lung function in healthy children (359). However, exhaled H2O2 concentration is related to the number of sputum eosinophils and airway hyperresponsiveness in asthma of different severity, and it is elevated in patients with severe unstable asthma, although exhaled NO is significantly reduced by treatment with corticosteroids (350). This may be related to the fact that neutrophils, prevalent in severe asthma (121), generate higher amounts of superoxide radicals and therefore H2O2 (360). Asthmatic patients also exhale significantly higher levels of thiobarbituric acid-reactive products (TBARs), which indirectly reflect increased oxidative stress (358).

COPD. Cigarette smoking causes an influx of neutrophils and other inflammatory cells into the lower airways, and fivefold higher levels of H2O2 have been found in exhaled breath condensate of smokers than in nonsmokers (361). Levels of exhaled H2O2 are increased compared with those in normal subjects in patients with stable COPD and are further increased during exacerbations (362, 363). Cigarette smoking is by far the commonest cause of COPD, but only 10 to 20% of smokers develop symptomatic COPD. No significant differences have been found between H2O2 levels in current smokers with COPD and subjects with COPD who have never smoked, and there is no correlation between expired H2O2 concentration and daily cigarette consumption (363). Thus, oxidative stress is a characteristic feature of COPD and presumably related to airway inflammation, and it cannot be explained entirely by the oxidants present in tobacco smoke.

Other lung diseases. Increased H2O2 levels in exhaled breath condensate have been found in ARDS (364, 365), bronchiectasis (351), and after lobectony/pneumonectomy in patients with lung carcinoma (366), indicating increased oxidative stress in these conditions, and are significantly reduced during antibiotic treatment in patients with infective exacerbations of CF (367).

Eicosanoids

Eicosanoids are potent mediators of inflammation responsible for vasodilatation/vasoconstriction, plasma exudation, mucus secretion, bronchoconstriction/bronchodilatation, cough, and inflammatory cell recruitment. They are derived from arachidonic acid and include prostaglandins, thromboxane, isoprostanes and leukotrienes. Noninvasive exhaled condensate analysis provides an opportunity to assess the eicosanoid profile in lung diseases directly, and it may be a better predictor of clinical efficacy of leukotriene antagonists or thromboxane inhibitors in lung disease than urine, serum, or invasive BAL.

Prostanoids. There is an increased expression of inducible cyclooxygenase (COX-2), which forms prostaglandins and thromboxane in asthma and COPD (368) and CF (369). Most prostaglandins and thromboxane have proinflammatory properties, but others, for example PGE2 and PGI2, are anti- inflammatory (370). For example, PGE2 inhibits induction of NOS2 in cell lines (371), and when inhaled reduces exhaled NO in asthma (155). Exhaled prostanoids are detectable in exhaled breath condensate. PGE2 and PGF are markedly increased in patients with COPD, whereas these prostaglandins are not significantly elevated in asthma (372). In contrast, TxB2 is increased in asthma but not detectable in normal subjects or in patients with COPD (Montuschi P. et al., unpublished observation). Exhaled thromboxanes may predict more accurately than urinary levels those patients who may benefit from a thromboxane receptor antagonist in asthma (373).

Leukotrienes. Leukotrienes (LTs), a family of lipid mediators derived from arachidonic acid via the 5-lipoxygenase pathways, are potent constrictor and proinflammatory mediators that contribute to the pathophysiology of asthma. The cysteinyl-leukotrienes (cys-LTs) LTC4, LTD4 and LTE4 are generated predominantly by mast cells and eosinophils and are able to contract airway smooth muscle, cause plasma exudation, and stimulate mucus secretion, as well as recruiting eosinophils (374). By contrast, LTB4 has potent chemotactic activity towards neutrophils (375). Detectable levels of LTB4, C4, D4, E4, and F4 have been reported in exhaled condensate of asthmatic and normal subjects (376, 377). Elevated exhaled condensate levels of LTB4 have been found in healthy calves during an experimental chest infection (353). There have been attempts to measure leukotrienes in urine, and increased levels of LTE4 have been reported in some asthmatic patients, but they are not consistently increased after allergen challenge (378). Allergen provocation increases LTC4 and LTE4 concentrations in BAL and in urine during early and late asthmatic responses (379). However, measurement of airway mediators in urine is problematic because of dilution of the lung-derived signal and delay in excretion. Increased levels of LTE4 have also been found in induced sputum during the late response to allergen in patients with mild asthma (380). In patients with mild asthma levels of LTE4, LTC4, LTD4 in exhaled condensate are increased during the late asthmatic response to allergen challenge (381). The levels of leukotrienes LTE4, C4, D4 in breath condensate are elevated significantly in patients with moderate or severe asthma (377), and steroid withdrawal in moderate asthma leads to worsening of asthma and further increase in exhaled NO and the concentration of LTB4, LTE4, LTC4, LTD4 in exhaled condensate (381) (Figure 7). LTB4 concentrations are increased in exhaled breath condensate of patients with COPD (Montuschi P. et al.: unpublished observations) and in moderate and severe asthma (377). This suggests that LTB4 may be involved in exacerbations of asthma and may contribute towards neutrophils recruitment.

Isoprostanes. Isoprostanes are a novel class of prostanoids formed by free radical-catalyzed lipid peroxidation of arachidonic acid (382). They are initially esterified in membrane phospholipids, from which they are cleaved by a phospholipase A2, circulate in plasma, and are excreted in urine, and can be detected in exhaled breath condensate and BAL. Their formation is largely independent of COX-1 and COX-2. They can be detected by ELISA (346, 383) and by GC/MS analysis (382). F2-isoprostanes are the major candidates for clinical measurement of oxidative stress in vivo. They are stable compounds, detectable in all normal biologic fluids and tissues (384), and their formation is increased by systemic oxidative stress, for example in patients with diabetes (385) or ARDS (386). F2-isoprostanes are reduced by antioxidants, for example by alpha-lipoic acid in normal subjects (387). They are not simply markers of lipid peroxidation but also possess biologic activity, and they could be mediators of the cellular effects of oxidant stress and a reflection of complex interactions between the RNS and ROS. Indeed, peroxynitrite is capable of activating biosynthesis of endoperoxide synthase and thromboxanes in inflammatory cells (388), and oxidizing arachidonic acid to form F2-isoprostanes. The most prevalent isoprostane in humans in 8-epi-PGF, also known as 8-isoprostane.

Asthma. F2-isoprostanes are increased in plasma (389) and BAL fluid of asthmatic patients and further increased after allergen challenge (390). 8-isoprostane levels are approximately doubled in patients with mild asthma compared with those in normal subjects, and increased by about 3-fold in those with severe asthma, irrespective of their treatment with corticosteroids (346) (Figure 7). The relationship to asthma severity is a useful aspect of this marker, in contrast to exhaled NO. The relative lack of effect of corticosteroids on exhaled 8-isoprostane has been confirmed in a placebo-controlled study with the two different doses of inhaled steroids (120). This provides evidence that inhaled corticosteroids may not be very effective in reducing oxidative stress. Exhaled isoprostanes may a better means of reflecting disease activity than exhaled NO.

COPD. Urinary levels of isoprostanes, in particular 8-isoprostane, are increased in COPD, but they decline in patients with acute exacerbation as their clinical condition improves (391). Aspirin treatment fails to decrease urinary levels of isoprostanes, whereas TxB2 were significantly reduced, confirming that cyclooxygenases are not involved in their formation. The concentration of 8-isoprostane in exhaled condensate is also increased in normal cigarette smokers, but to a much greater extent in patients with COPD (392). Interestingly, exhaled 8-isoprostane is increased to a similar extent in patients with COPD who are ex-smokers as in smoking patients with COPD, indicating that the exhaled isoprostanes in COPD are largely derived from oxidative stress from airway inflammation, rather than from cigarette smoking.

Cystic fibrosis. CF is characterized by marked oxidative stress in the airways (393), and elevated levels of 8-isoprostane have been detected in plasma (394). Concentrations of 8-isoprostane in the breath condensate of patients with stable CF are increased about threefold compared with those in normal subjects (293).

Interstitial lung disease. Interstitial lung diseases such as cryptogenic fibrosing alveolitis (CFA) and fibrosing alveolitis associated with systemic sclerosis (FASSc), are characterized by enhanced oxidative stress in both serum (395) and BAL fluid (396). The imbalance between the oxidants and antioxidants is also a prominent feature of sarcoidosis (397). 8-isoprostane is detectable in BAL fluid of normal subjects and is increased in patients with sarcoidosis, CFA, and FASSc, suggesting a higher level of oxidant stress and greater lung injury in these patients than in those with sarcoidosis (383).

Products of Lipid Peroxidation

There are several methods to measure lipid peroxidation products and lipid peroxidation damage in tissues, cells, and body fluids. The most simple, but nonspecific, method is measurement of thiobarbituric acid-reactive substances (TBARS). The specificity of colorimetric or fluorimetric assays can be significantly improved if combined with high pressure liquid chromatography. If levels of TBARS are increased, as for example in exhaled condensate in asthma and COPD (358), other more sophisticated assays may be performed for verification. Assays are available for phospholipid- and cholesterylester, hydroperoxides, aldehydic lipid peroxidation products, including 4-hydroxynonenal, fluorescent protein adducts (e.g., lipofuscin), conjugated dienes, and antioxidants (398).

Although there is a still a question whether lipid peroxidation contributes to organ dysfunction or simply reflects oxidative injury, tissue-specific lipid peroxidation has been confirmed. Thus, lung lipid conjugated dienes are increased after intravenous infusion of both endotoxin and H2O2 in rats (399). However, venous plasma-conjugated dienes are elevated only after H2O2. Significantly higher concentrations of primary (diene conjugates) and secondary (ketodienes) products of lipid peroxidation have been found in exhaled condensate and in bronchial biopsy samples from patients with COPD and chronic bronchitis compared with those in normal subjects (400, 401). Increased levels of free fatty acids, including linoleic and arachidonic acids, have been measured in exhaled condensate and sweat in children (320) and in adults (402) with acute pneumonia and lung edema (403). In contrast, the level of lipid peroxidation in patients with cancer was significantly reduced compare with that in healthy control subjects (404). Exhaled condensates may be used in prenatal diagnosis of fetal hypoxia, as significantly higher levels of diene conjugates and malonic dialdehydes have been found in pregnant women who gave birth to babies with severe fetal and neonatal hypoxia (405). Recent studies have suggested that the increased permeability in patients with interstitial lung disease results in an increase of alveolar-to-vascular leakage of surfactant proteins A and D (406). The clearance system of these proteins from the bloodstream is unknown at present, but if they are detectable in exhaled breath condensate, they may be the best practical examination for this disease.

Vasoactive Amines

Elevated levels of acetylcholine, serotonin, and histamine, which were related to the severity of airway inflammation, airway obstruction, and airway hyperresponsiveness, have been reported in exhaled breath condensate in asthma (407) and in acute bronchitis (408). High levels of acetylcholine, catecholamines, histamine, and serotonin and low levels of cortisol and thyroxine are reported in exhaled condensate in coal miners with early stages of silicosis (409).

NO-related Products

NO reacts with superoxide to yield peroxynitrite, and it can be trapped by thiol-containing biomolecules such as cysteine and glutathione, to form S-nitrosothiols or can be oxidized to nitrate and nitrite (410). Nitrogen intermediates, for example peroxynitrite, can induce a number of covalent modifications in various biomolecules such as nitrosoadducts and nitroadducts. One such modification yields 3-nitrotyrosine, and detection of this adduct in proteins is now commonly used as a diagnostic tool to identify involvement of NO-derived oxidants in many disease states (411). The balance between nitrite/nitrate, S-nitrosothiols, and nitrotyrosine in lung epithelial lining fluids, as reflected by exhaled breath condensate, gives insight into NO synthesis and short- and long-term changes in NO production. There are several methods, apart from the immunoassays, available for nitrite/nitrate and S-nitrosothiol quantification. They include an adsorptive stripping voltammetry (412) and electrochemical (413), fluorimetric, and colorimetric measurements (414, 415). There is also a method that allows the separation of the thiols from their S-nitrosylated derivatives using capillary zone electrophoresis (416).

Asthma. High levels of nitrite have been found in exhaled breath condensate (417) and sputum (418) of asthmatic patients, especially during acute exacerbations (417). The ratio of airway wall thickness to lumen diameter measured by high resolution computed tomography was significantly correlated with the sputum concentration of nitrite/nitrate (418). In fact, we have shown that nitrotyrosine, a stable product of peroxynitrite decomposition in exhaled breath condensate, is increased in mild steroid-naı̈ve asthma and is reduced in patients with severe asthma receiving steroid therapy (377). However, increased levels of nitrotyrosine in exhaled breath condensate are associated with worsening of asthma symptoms and deterioration of lung function during inhaled steroid withdrawal in moderate asthma (381), suggesting that nitrotyrosine may be not only a predictor of asthma deterioration, but may play a key role in the pathogenesis of airway remodeling.

A deficiency in S-nitrosothiols has been demonstrated in tracheal lining fluid in asthmatic children with respiratory failure (419), suggesting that the levels of S-nitrosothiols, which are endogenous bronchodilators, may normally counteract increased airway tone in asthma. The levels of S-nitrosothiols in exhaled breath condensate are reduced after 3 wk of treatment with a higher (400 μg daily) but not a lower dose (100 μg daily) of inhaled budesonide (120). In contrast, there is a rapid and dose-dependent reduction in nitrite/nitrate in exhaled breath condensate in the same patients with mild asthma, suggesting that nitrite/nitrate are more sensitive to anti-inflammatory treatment.

COPD. Habitual smokers have unusually high antioxidant concentrations in the epithelial lining fluid and higher resistance to oxidative pulmonary damage. NO can be trapped in the epithelial lining fluid of the respiratory tract in the form of S-nitrosothiols or peroxynitrite and released thereafter, leading to transient elevation of exhaled NO after smoking of a cigarette (420). Chronic oxidative stress presented to the lung by cigarette smoke may decrease the availability of thiol compounds and may increase decomposition of nitrosothiols, explaining elevated levels of S-nitrosothiols in exhaled condensate in healthy smokers, which are related to smoking history (421). Levels of exhaled nitrite/nitrate are increased in COPD (unpublished observation). A significant negative correlation between FEV1 and the amount of nitrotyrosine formation has been demonstrated in patients with COPD, but not in those with asthma and normal subjects (422), suggesting that NO produced in the airways is consumed by its reaction with superoxide anion and/or peroxidase-dependent mechanisms, and reactive nitrogen species play an important role in the pathobiology of the airway inflammatory and obstructive process in COPD.

Cystic fibrosis. Elevated levels of nitrite and nitrate (352, 423) and nitrotyrosine (424) have been found in exhaled condensate and sputum (425) of patients with CF during both the stable period and exacerbations. In children with CF and normal lung function, however, the nitrite/nitrate concentrations in BAL are normal and concentrations of S-nitrosothiols are reduced (426). In contrast, elevated levels of nitrite and S-nitrosothiols are found in exhaled breath condensate of adult patients with more severe CF (427).

Myeloperoxidase, a heme enzyme of neutrophils that uses H2O2 to oxidize chloride to hypochlorous acid, is capable of catalyzing nitration of tyrosine, providing an alternative to peroxynitrite in the formation of 3-nitrotyrosine (428). At sites of neutrophilic inflammation myeloperoxidase will nitrate proteins because the cosubstrate tyrosine will be available to facilitate the reaction (428). Patients with stable CF have significantly higher levels of nitrotyrosine in exhaled breath condensate than do normal subjects (424). This suggests that nitration of proteins by myeloperoxidase may be an additional source of nitrotyrosine in patients with CF who have a very low NO production. In fact, myeloperoxidase is elevated in CF sputum and correlates with nitrotyrosine concentrations (425), implying that an absence of an increase in exhaled NO does not exclude the possibility of NO participating in airway inflammation, including CF.

Other lung diseases. Nitrite and nitrate concentrations are increased in exhaled breath condensate of patients with active pulmonary sarcoidosis (214).

Ammonia

Ammonia (NH3), a product of urease hydrolysis of urea to ammonia and carbamate, is one of the key steps in the nitrogen cycle. Ammonia in the respiratory tract may be able to neutralize inhaled acid vapors and aerosols, mitigating the pulmonary effects of pollution (429) and has the potential to regulate NOS activity. Thus, plasma of patients with uremia has an inhibitory effect on NOS3 in a human endothelial cell line and NOS2 in murine macrophages (430).

The urea breath test has been in clinical practice for a considerable period of time as one of the most important noninvasive methods for detecting Helicobacter pylori infection (76). The test exploits the hydrolysis of orally administered urea by the enzyme urease, which H. pylori produces in large quantities. Urea is hydrolyzed to ammonia and carbon dioxide, which diffuses into the blood and is excreted by the lungs. The first measurements of exhaled NH3 were used to assess different food supplements given during the space flights in the 1970s (431). Recently, using selected ion flow tube mass spectrometric technique the levels of alveolar exhaled ammonia (in the range of 200 to 1,750 ppb) have been detected from single exhalations in healthy volunteers who have ingested a liquid protein meal (432).

Exhaled breath ammonia may be an important counteracting agent in a variety of respiratory conditions, as a low pH in exhaled breath condensate has recently been reported in asthma (32). Exposure to ammonia gas in the workplace is significantly associated with increase in respiratory symptoms and asthma (433). It has been shown that elevated levels of urea can be used to predict oxidative stress, as the levels of urea in saliva are significantly increased after chronic hyperbaric oxygen exposure (434). The fact that acidic rinsing results in a considerable (90%), fast and lasting for 1 h reduction in exhaled ammonia in normal subjects (429) should be considered when ammonia is measured in exhaled condensate.

Ammonia is an important pathogenic factor for certain bacteria, for example Cryptococcus neoformans, which is a significant human pathogenic fungus that produces large amounts of urease (435). Exhaled ammonia levels measured by chemiluminescence are not different between normal subjects and patients with stable CF, but are significantly higher in asthma and in normal subjects with upper respiratory tract infections (436). It is possible that measurements of exhaled ammonia might differentiate viral and bacterial infections in a variety of lung diseases.

Electrolytes

Increased airway fluid osmolality in the lower airways as a result of exercise, may activate mast cells and cause subsequent bronchoconstriction in a subset of asthmatics. A deficiency in magnesium and an elevation in calcium concentrations in exhaled breath condensate have been reported in atopic asthma (437), although a histamine-induced decrease in plasma magnesium levels occurs regardless of the diagnosis of asthma (438). We have recently demonstrated that exhaled Na+ and Cl are elevated in exhaled condensates of patients with CF and correlate with the sweat test and the disease severity (Balint et al., unpublished observation). Recently, a strong negative correlation between sputum Cl concentrations and exhaled NO has been demonstrated in patients with PCD (191), suggesting that airway mucociliary clearance impairment might be monitored by exhaled/nasal NO and exhaled Cl levels.

Hydrogen Ions

An acidic microenvironment up-regulates NOS2 in macrophages through the activation of NF-κB (439), making NO release moderately pH-dependent (30). Elevated levels of lactic acid have been found in exhaled condensate in patients with acute bronchitis (408), and a low pH of exhaled condensate is reported in patients with acute asthma (32). Exhaled pH is free of salivary, nasal, and gastric contamination and is not influenced by either airflow obstruction or inhaled albuterol, but it is increased by corticosteroid therapy.

Proteins and Cytokines

Measurement and identification of proteins in exhaled condensate is controversial. It has been reported that the amount of protein in the breath condensate of eight healthy subjects was from 4 to 1.4 mg, originating from the nasopharynx, oropharynx, and lower airways (347). The same group has also reported the presence of IL-1β, soluble IL-2 receptor protein, IL-6, and TNF-α in exhaled breath condensate of patients with a variety of respiratory conditions (347). Recently, higher concentrations of total protein in exhaled condensate have been found in young smokers when compared with nonsmokers, whereas the levels of IL-1β and TNF-α were not different (440). We have found that IL-8 levels in exhaled condensate are mildly elevated in stable CF but are more than doubled in patients with unstable CF compared with normal subjects (Balint B. et al., unpublished observations).

Exhaled Temperature

Airway cooling provokes an increase in bronchial blood flow and is manifested as a rapid resupply of heat in asthma (441). NO modulates temperature by regulating vascular tone and blood flow (71). Measurements of exhaled temperature and humidity have been used to assess the conditioning function of the respiratory apparatus in asthma, COPD, pneumonia, and pneumoconiosis (442, 443).

Asthma is characterized by inflammation-related vascular hyperperfusion (444), so airway mucosal blood flow and exhaled temperature may be an index of airway inflammation. Indeed, exhaled temperature measured under controlled conditions (standardized expiratory flow and pressure) (56), as breathing pattern may affect airway wall temperature (445), is low in CF and COPD (446), but elevated in asthma when compared with normal subjects (447, 448). Exhaled breath temperature may serve as a nonspecific, simple, and inexpensive method for home monitoring of several upper and lower respiratory conditions such as asthma, COPD, CF, and rhinitis and for assessing the effects of anti-inflammatory treatments.

Combined Gas Chromatography/Spectroscopy

A new analytical method of gas chromatography combined with UV spectroscopy has been used to measure isoprene and acetone in expired air in healthy newborns, preschool children, healthy and diabetic school children (449), or isoprene in healthy adult subjects (450). A new method for analysis of ethanol and acetone in exhaled air using a portable gas chromatograph with a photoionization detector has been developed and has demonstrated that ethanol levels are more than tenfold higher in patients with cardiorespiratory disorders than in normal subjects (451). Exhaled formaldehyde from women with breast cancer and in the tumor-bearing mice is significantly higher than in healthy subjects, suggesting that these carbonyl compounds may be used as a biomarker (452). Laser magnetic resonance spectroscopy (LMRS) is a sensitive and isotope-selective technique for determining low concentrations of gaseous free radicals with high time resolution, which has been successfully used to measure exhaled and nasal NO at the end of exhalation in normal subjects (453), or it can be a simple alternative to mass spectrometry in detection of exhaled 14C-urea in patients with H. pylori infection (454, 455).

The Selected Ion Flow Tube (SIFT) Technique

The selected ion flow tube (SIFT) technique for trace gas analysis of air and breath is based on soft chemical ionization exploiting the ion-molecule reactions that occur between the trace gases and the preselected precursor ions (H3O+, NO+, and O2 +) (456). This method is sensitive (detection limit is down to about 10 ppb) and fast (response time ∼ 20 ms) and can be used during a normal breathing cycle.

Polymer-coated Surface-acoustic-wave Resonators

Portable instruments based on microsensor arrays of polymer-coated surface-acoustic-wave resonators have been introduced and are capable of the analysis of organic vapors (457). Detection of the bound immunocomplex has been made possible via the silicon chip-based light-addressable potentiometric sensor. For example, in the presence of the urea, urease converts the substrate to ammonia and CO2 and this leads to a pH change at the silicon surface. The resultant pH change can be monitored with time and the signal output can be reported in real time.

Exhaled breath analysis has enormous potential as a noninvasive means of monitoring airway and inflammation, oxidative stress, and other conditions (for example, metabolic disorders, bacterial and viral infections). The technique is simple for patients to perform and may be applied in neonates and patients with severe disease. Because the techniques are noninvasive, it is possible to make repeated measurements without disturbing the system, in contrast to the invasive procedures currently used.

Standardization of Measurements

Precautions need to be taken to ensure uniformity of measurement between different centers, and physiologic and measurement factors are likely to differ between markers. This has been most carefully worked out for exhaled NO, and two International Taskforce meetings have defined standards and procedures for measurement of exhaled NO in adults and children (53, 61). Similar standardization methods are now needed for the other exhaled markers currently under investigation.

Clinical Application

There is a pressing need for the evaluation of these techniques in long-term clinical studies (3). Whether repeated measurements of exhaled markers will help in the clinical management of lung diseases needs to be determined by longitudinal studies relating exhaled markers to other measurements of asthma control. This is most advanced with measurement of exhaled NO (3), but it is still uncertain whether routine measurement of exhaled NO will improve the clinical control of asthma in a cost-effective way.

None of the exhaled markers are diagnostic for a particular lung disease, apart from the very low nasal and exhaled NO in primary ciliary dyskinesia. Nevertheless, measurement of these markers may aid differential diagnosis of lung diseases. For example, a normal level of exhaled NO in a patient with chronic cough makes the diagnosis of asthma very unlikely. A high level of exhaled NO in an asthmatic patient receiving inhaled corticosteroids most likely indicates poor compliance with therapy.

Exhaled markers may also be used to assess the response to therapies such as inhaled corticosteroids and novel anti-inflammatory treatments now in development. Some markers may even be used to predict responses to specific treatments. For example, high levels of LTE4 in exhaled breath condensates may predict a better clinical response to antileukotrienes, and a high level of markers of oxidative stress may indicate patients who might respond to antioxidant therapy.

Profiles of Mediators

We have reviewed a large body of data on exhaled volatile gases and exhaled breath condensate that demonstrate different patterns of change in different pulmonary diseases (summarized in Tables 3 and Table 4). At the moment single exhaled markers are usually evaluated in isolation, but, as indicated above, markers are affected differently in different diseases, and different markers vary in their sensitivity to certain maneuvers such as the effect of therapy. For example, asthma is characterized by a large increase in exhaled NO, a modest increase in CO, and a moderate increase in exhaled 8-isoprostane, whereas COPD is characterized by little or no increase in exhaled NO, and by larger increases in exhaled CO and 8-isoprostane. By contrast, patients with CF typically have low exhaled NO concentrations and high levels of exhaled CO and 8-isoprostane. Exhaled NO appears to be sensitive to inhibition by low doses of inhaled corticosteroids in asthma, whereas exhaled CO and 8-isoprostane are much less sensitive to inhibition by corticosteroids. These differences may be exploited in the future as more markers are characterized, so that each disease may have a characteristic profile or fingerprint of different markers that may be diagnostic. Treatments too may impose a characteristic effect on these markers, and this may improve the specificity of treatment in the future, particularly as more potent and specific treatments become available.

Table 3.  CHANGES IN EXHALED GASES IN LUNG DISEASE

AsthmaCOPDCFBronchILDPCD
StableUnstableStableUnstableStableUnstable
Nitric oxide↑ ↑ ↑ ↑ ↑ ↑ ↑ ↓ ↓
Carbon monoxide↑ ↑ ↑ ↑ ↑ ↑ ↑ ↑ ↑ ↑ ↑ ?
Ethane↑ ↑ ?↑ ↑ ?↑ ↑ ↑ ↑ ↑ ???

Definition of abbreviations: Bronch = bronchiectasis; CF = cystic fibrosis; ILD = interstitial lung disease; PCD = primary ciliary dyskinesia; ↑ = increase; ↓ = decrease; ↔ = no change; ? = not yet known.

Table 4.  CHANGES IN EXHALED CONDENSATE IN LUNG DISEASE

AsthmaCOPDCFBronchILDPCD
StableUnstableStableUnstableStableUnstable
Eicosanoids??
 8-isoprostanes↑ ↑ ↑ ?↑ ↑ ↑ ?↑ ↑
 LTE4, C4, D4 ↑ ↑ ????
 LTB4 ↑ ↑ ↑ ↑ ↑ ????
 PG???????
 Tx???????
NO-related products???
 Nitrotyrosine????
 NO2 /NO3 ↑ ↑ ?↑ ↑
 SNO??↑ ↑
H2O2 ↑ ↑ ↑ ↑ ????
Lipid peroxidation product?↑ ↑ ??????
Vasoactive amines????????
Ammonia????????
Hydrogen ions (pH)↑ ↑ ???????
Cytokines???????
 IL-1β, IL-2, IL-6,
 TNF-α
 IL-8
Electrolytes???????
 Na, Cl?↑ ↑
 Mg
 Ca

Definition of abbreviations: H2O2 = hydrogen peroxide; IL-1β, −2, −6 = interleukin 1β, −2, −6; IL8 = interleukin-8; LT = leukotriene (E4, C4, D4, B4); NO2 = nitrite; NO3 = nitrate; SNO = S-nitrosothiols; TNF-α = tumor necrosis factor α. For other definitions, see Table 3.

Measuring Devices

The value of particular markers will depend on the availability of reliable, fast, and inexpensive detector systems. NO chemiluminescence analyzers are currently relatively expensive and are mainly available in academic research laboratories. However, advances in technology have now resulted in smaller devices that are cheaper and easier to use. This will increase the availability of the measurement, which will further reduce the price as exhaled NO analyzers become routine lung function measurements. Eventually it may be possible to introduce such analyzers in family practice and even into patients' homes, so that patients themselves will be able to monitor their own markers and adjust their treatment accordingly.

Measurement of some of the other exhaled markers such as hydrocarbons is much more difficult using present technology, but it may also be possible to develop much smaller and cheaper detectors that would make this measurement more readily available. Although exhaled breath condensates is an attractive approach that could easily be adapted to home measurements, its value is limited by the fact that complex assays, including ELISAs, fluorimetric assays, and HPLC are needed to measure the individual chemical markers. In the future these assays may be simplified by the use of strip reagents that give rapid color changes, so that these measurements may be available for clinicians and for patients to use at home.

New Markers

It is likely that the possibilities for measurement of markers in exhaled breath are far greater than currently realized. It is clear that exhaled breath condensates contain many different molecules, including proteins. In fact, application of proteomics, with high resolution two-dimensional gel electrophoresis and microanalysis of protein spots may allow the recognition of particular protein patterns in different diseases and may result in the recognition of new diagnostic proteins or therapeutic targets. New and more sensitive assays may also allow the detection of many other markers of inflammation and even specific fingerprints of activation of particular cell types within the respiratory tract such as eosinophils, neutrophils, epithelial cells, and macrophages. This could have far-reaching potential for the diagnosis and treatment of many airway diseases.

1. Parameswaran K, Pizzichini E, Pizzichini MM, Hussack P, Efthimiadis A, Hargreave FEClinical judgement of airway inflammation versus sputum cell counts in patients with asthma. Eur Respir J152000486490
2. Nightingale JA, Rogers DF, Barnes PJEffect of repeated sputum induction on cell counts in normal volunteers. Thorax5319988790
3. Kharitonov SA, Barnes PJClinical aspects of exhaled nitric oxide. Eur Respir J162000781792
4. Kharitonov SAExhaled nitric oxide and carbon monoxide in asthma. Eur Respir J91999212218
5. Kharitonov SA, Barnes PJClinical aspects of exhaled nitric oxide. Eur Respir J162000781792
6. Gustafsson LEExhaled nitric oxide as a marker in asthma. Eur Respir J Suppl26199849S52S
7. Nathan C, Xie QWRegulation of biosynthesis of nitric oxide. J Biol Chem26919941372513728
8. Barnes PJ, Belvisi MGNitric oxide and lung disease. Thorax48199310341043
9. Gaston B, Drazen JM, Loscalzo J, Stamler JSThe biology of nitrogen oxides in the airways. Am J Respir Crit Care Med1491994538551
10. Barnes PJTranscription factors and inflammatory disease. Hosp Pract3119969396
11. Gao PS, Kawada H, Kasamatsu T, Mao XQ, Roberts MH, Miyamoto Y, Yoshimura M, Saitoh Y, Yasue H, Nakao K, Adra CN, Kun JF, Moro-oka S, Inoko H, Ho LP, Shirakawa T, Hopkin JMVariants of NOS1, NOS2, and NOS3 genes in asthmatics. Biochem Biophys Res Commun2672000761763
12. Grasemann H, Yandava CN, vonStorm G, Deykin A, Pillari A, Ma J, Sonna LA, Lilly C, Stampfer MJ, Israel E, Silverman EK, Drazen JMA neuronal NO synthase (NOS1) gene polymorphism is associated with asthma. Biochem Biophys Res Commun2722000391394
13. Wechsler ME, Grasemann H, Deykin A, Silverman EK, Yandava CN, Israel E, Wand M, Drazen JMExhaled nitric oxide in patients with asthma: association with NOS1 genotype. Am J Respir Crit Care Med162200020432047
14. Cremona G, Higenbottam TW, Takao M, Hall L, Bower EAExhaled nitric oxide in isolated pig lungs. J Appl Physiol7819955963
15. Persson MG, Midtvedt T, Leone AM, Gustafsson LECa2+-dependent and Ca2+-independent exhaled nitric oxide, presence in germ-free animals, and inhibition by arginine analogues. Eur J Pharm26419941320
16. Shaul PW, North AJ, Wu LC, Wells LB, Brannon TS, Lau KS, Michel T, Margraf LR, Star RAEndothelial nitric oxide synthase is expressed in cultured human bronchiolar epithelium. J Clin Invest94199422312236
17. Mannick JB, Asano K, Izumi K, Kieff E, Stamler JSNitric oxide produced by human B lymphocytes inhibits apoptosis and Epstein-Barr virus reactivation. Cell79199411371146
18. Guo FH, De Raeve HR, Rice TW, Stuehr DJ, Thunnissen FB, Erzurum SCContinuous nitric oxide synthesis by inducible nitric oxide synthase in normal human airway epithelium in vivo. Proc Natl Acad Sci USA92199578097813
19. Steudel W, Kirmse M, Weimann J, Ullrich R, Hromi J, Zapol WMExhaled nitric oxide production by nitric oxide synthase-deficient mice. Am J Respir Crit Care Med162200012621267
20. Belvisi MG, Barnes PJ, Larkin S, Yacoub M, Tadjkarimi S, Williams TJNitric oxide synthase activity is elevated in inflammatory lung diseases. Eur J Pharmacol2831995255258
21. Hamid Q, Springall DR, Riveros-Moreno V, Chanez P, Howarth PH, Redington A, Bousquet J, Godard P, Holgate S, Polak JMInduction of nitric oxide synthase in asthma. Lancet342199315101513
22. Guo FH, Comhair SA, Zheng S, Dweik RA, Eissa NT, Thomassen MJ, Calhoun W, Erzurum SCMolecular mechanisms of increased nitric oxide (NO) in asthma: evidence for transcriptional and post-translational regulation of NO synthesis. J Immunol164200059705980
23. Xie Q, Kashiwarbara Y, Nathan CRole of transcription factor NF-kB/ Rel in induction of nitric oxide synthase. J Biol Chem269199447054708
24. Asano K, Chee CB, Gaston B, Lilly CM, Gerard C, Drazen JM, Stamler JSConstitutive and inducible nitric oxide synthase gene expression, regulation, and activity in human lung epithelial cells. Proc Natl Acad Sci USA9119941008910093
25. Chartrain NA, Geller DA, Koty PP, Sitrin NF, Nussler AK, Hoffman EP, Billiar TR, Hutchinson NI, Mudgett JSMolecular cloning, structure, and chromosomal localization of the human inducible nitric oxide synthase gene. J Biol Chem269199467656772
26. Saleh D, Ernst P, Lim S, Barnes PJ, Giaid AIncreased formation of the potent oxidant peroxynitrite in the airways of asthmatic patients is associated with induction of nitric oxide synthase: effect of inhaled glucocorticoid. FASEB J121998929937
27. Robbins RA, Barnes PJ, Springall DR, Warren JB, Kwon OJ, Buttery LD, Wilson AJ, Geller DA, Polak JMExpression of inducible nitric oxide in human lung epithelial cells. Biochem Biophys Res Commun2031994209218
28. Kharitonov SA, Yates DH, Robbins RA, Logan-Sinclair R, Shinebourne EA, Barnes PJIncreased nitric oxide in exhaled air of asthmatic patients. Lancet3431994133135
29. Stamler JS, Simon DI, Osborne JA, Mullins ME, Jaraki O, Michel T, Singel DJ, Loscalzo JS-nitrosylation of proteins with nitric oxide: synthesis and characterization of biologically active compounds. Proc Natl Acad Sci USA891992444448
30. Sheu FS, Zhu W, Fung PCDirect observation of trapping and release of NO by glutathione and cysteine with electron paramagnetic resonance spectroscopy. Biophys J78200012161226
31. Klebanoff SJReactive nitrogen intermediates and antimicrobial activity: role of nitrite. Free Radic Biol Med141993351360
32. Hunt JF, Fang K, Malik R, Snyder A, Malhotra N, Platts-Mills TA, Gaston BEndogenous airway acidification: implications for asthma pathophysiology. Am J Respir Crit Care Med1612000694699
33. Kharitonov SA, Chung FK, Evans DJ, O'Connor BJ, Barnes PJThe elevated level of exhaled nitric oxide in asthmatic patients is mainly derived from the lower respiratory tract. Am J Respir Crit Care Med153199617731780
34. Baraldi E, Azzolin NM, Cracco A, Zacchello FReference values of exhaled nitric oxide for healthy children 6–15 years old. Pediatr Pulmonol2719995458
35. Thomas SR, Kharitonov SA, Scott SF, Hodson ME, Barnes PJNasal and exhaled nitric oxide is reduced in adult patients with cystic fibrosis and does not correlate with cystic fibrosis genotype. Chest117200010851089
36. Lundberg JO, Weitzberg ENasal nitric oxide in man. Thorax541999947952
37. Borland C, Cox Y, Higenbottam TMeasurement of exhaled nitric oxide in man. Thorax48199311601162
38. Gustafsson LE, Leone AM, Persson MG, Wiklund NP, Moncada SEndogenous nitric oxide is present in the exhaled air of rabbits, guinea pigs and humans. Biochem Biophys Res Commun1811991852857
39. Alving K, Weitzberg E, Lundberg JMIncreased amount of nitric oxide in exhaled air of asthmatics. Eur Respir J6199313681370
40. Massaro AF, Gaston B, Kita D, Fanta C, Stamler JS, Drazen JMExpired nitric oxide levels during treatment of acute asthma. Am J Respir Crit Care Med1521995800803
41. Schedin U, Frostell C, Persson MG, Jakobsson J, Andersson G, Gustafsson LEContribution from upper and lower airways to exhaled endogenous nitric oxide in humans. Acta Physiol Scand391995327332
42. Massaro AF, Mehta S, Lilly CM, Kobzik L, Reilly JJ, Drazen JMElevated nitric oxide concentrations in isolated lower airway gas of asthmatic subjects. Am J Respir Crit Care Med153199615101514
43. Lundberg JO, Farkas-Szallasi T, Weitzberg E, Rinder J, Lindholm J, Anggaro A, Hokfelt T, Lundberg JM, Alving KHigh nitric oxide production in human paranasal sinuses. Nat Med11995370373
44. Kondo T, Inokuchi T, Ohta K, Annoh H, Chang JDistribution, chemical coding and origin of nitric oxide synthase-containing nerve fibres in the guinea pig nasal mucosa. J Auton Nerv Syst8020007179
45. Cervin A, Onnerfalt J, Edvinsson L, Grundemar LFunctional effects of neuropeptide Y receptors on blood flow and nitric oxide levels in the human nose. Am J Respir Crit Care Med160199917241728
46. Kawamoto H, Takumida M, Takeno S, Watanabe H, Fukushima N, Yajin K. Localization of nitric oxide synthase in human nasal mucosa with nasal allergy. Acta Otolaryngol Suppl (Stockh) 1998;539:65–70.
47. Ramis I, Bioque G, Lorente J, Jares P, Quesada P, Rosello-Catafau J, Gelpi E, Bulbena OConstitutive nuclear factor-kappa B activity in human upper airway tissues and nasal epithelial cells. Eur Respir J152000582589
48. Qian W, Chatkin JM, Djupesland PG, McClean P, Zamel N, Irish JC, Haight JSUnilateral nasal nitric oxide measurement after nasal surgery. Ann Otol Rhinol Laryngol1092000952957
49. Sartori C, Lepori M, Busch T, Duplain H, Hildebrandt W, Bartsch P, Nicod P, Falke KJ, Scherrer UExhaled nitric oxide does not provide a marker of vascular endothelial function in healthy humans. Am J Respir Crit Care Med1601999879882
50. Yates DH, Kharitonov SA, Robbins RA, Thomas PS, Barnes PJEffect of a nitric oxide synthase inhibitor and a glucocorticosteroid on exhaled nitric oxide. Am J Respir Crit Care Med1521995892896
51. Yates DH, Kharitonov SA, Thomas PS, Barnes PJEndogenous nitric oxide is decreased in asthmatic patients by an inhibitor of inducible nitric oxide synthase. Am J Respir Crit Care Med1541996247250
52. Persson MG, Wiklund NP, Gustafsson LEEndogenous nitric oxide in single exhalations and the change during exercise. Am Rev Respir Dis148199312101214
53. Kharitonov SA, Alving K, Barnes PJExhaled and nasal nitric oxide measurements: recommendations. Eur Respir J10199716831693
54. Kharitonov SA, Barnes PJNasal contribution to exhaled nitric oxide during exhalation against resistance or during breath holding. Thorax521997540544
55. Silkoff PE, McClean PA, Slutsky AS, Furlott HG, Hoffstein E, Wakita S, Chapman KR, Szalai JP, Zamel NMarked flow-dependence of exhaled nitric oxide using a new technique to exclude nasal nitric oxide. Am J Respir Crit Care Med1551997260267
56. Paredi P, Loukides S, Ward S, Cramer D, Spicer M, Kharitonov SA, Barnes PJExhalation flow and pressure-controlled reservoir collection of exhaled nitric oxide for remote and delayed analysis. Thorax531998775779
57. Baraldi E, Scollo M, Zaramella C, Zanconato S, Zacchello FA simple flow-driven method for online measurement of exhaled NO starting at the age of 4 to 5 years. Am J Respir Crit Care Med162200018281832
58. Artlich A, Jonsson B, Bhiladvala M, Lonnqvist PA, Gustafsson LESingle breath analysis of endogenous nitric oxide in the newborn. Biol Neonate7920012126
59. Lehtimaki L, Turjanmaa V, Kankaanranta H, Saarelainen S, Hahtola P, Moilanen EIncreased bronchial nitric oxide production in patients with asthma measured with a novel method of different exhalation flow rates. Ann Med322000417423
60. Tsoukias NM, George SCA two-compartment model of pulmonary nitric oxide exchange dynamics. J Appl Physiol851998653666
61. Anonymous. Recommendations for standardized procedures for the online and offline measurement of exhaled lower respiratory nitric oxide and nasal nitric oxide in adults and children. Am J Respir Crit Care Med 1999;160:2104–2117.
62. Ekroos H, Tuominen J, Sovijarvi ARExhaled nitric oxide and its long-term variation in healthy non-smoking subjects. Clin Physiol202000434439
63. ten Hasken NHT, van der Vaart H, van der Mark TW, Koëter GH, Postma DS. Exhaled nitric oxide is higher both at day and night in subjects with nocturnal asthma. Am J Respir Crit Care Med 1998; 158:902–907.
64. Purokivi M, Randell J, Hirvonen MR, Tukiainen H. Reproducibility of measurements of exhaled NO, and cell count and cytokine concentrations in induced sputum. Eur Respir J 2000;16:242–246.
65. Bartley J, Fergusson W, Moody A, Wells AU, Kolbe J. Normal adult values, diurnal variation, and repeatability of nasal nitric oxide measurement. Am J Rhinol 1999;13:401–405.
66. Kharitonov SA, Logan-Sinclair RB, Busset CM, Shinebourne EAPeak expiratory nitric oxide differences in men and women: relation to the menstrual cycle. Br Heart J721994243245
67. Kirsch EA, Yuhanna IS, Chen Z, German Z, Sherman TS, Shaul PWEstrogen acutely stimulates endothelial nitric oxide synthase in H441 human airway epithelial cells. Am J Respir Crit Care Med201999658666
68. Kharitonov SA, Lubec G, Lubec B, Hjelm M, Barnes PJl-arginine increases exhaled nitric oxide in normal human subjects. Clin Sci881995135139
69. Sapienza MA, Kharitonov SA, Horvath I, Chung KF, Barnes PJEffect of inhaled l-arginine on exhaled nitric oxide in normal and asthmatic subjects. Thorax531998172175
70. McKnight GM, Smith LM, Drummond RS, Duncan CW, Golden M, Benjamin NChemical synthesis of nitric oxide in the stomach from dietary nitrate in humans. Gut401997211214
71. Holden WE, Wilkins JP, Harris M, Milczuk HA, Giraud GDTemperature conditioning of nasal air: effects of vasoactive agents and involvement of nitric oxide. J Appl Physiol87199912601265
72. Sippel JM, Giraud GD, Holden WENasal administration of the nitric oxide synthase inhibitor l-NAME induces daytime somnolence. Sleep221999786788
73. Deykin A, Massaro AF, Coulston E, Drazen JM, Israel EExhaled NO following repeated spirometry or repeated plethysmography in healthy individuals. Am J Respir Crit Care Med161200012371240
74. Silkoff PE, Wakita S, Chatkin J, Ansarin K, Gutierrez C, Caramori M, McClean P, Slutsky AS, Zamel N, Chapman KRExhaled nitric oxide after beta2-agonist inhalation and spirometry in asthma. Am J Respir Crit Care Med1591999940944
75. Phillips CR, Giraud GD, Holden WEExhaled nitric oxide during exercise: site of release and modulation by ventilation and blood flow. J Appl Physiol80199618651871
76. Piacentini GL, Bodini A, Costella S, Vicentini L, Suzuki Y, Boner ALExhaled nitric oxide is reduced after sputum induction in asthmatic children. Pediatr Pulmonol292000430433
77. Nightingale JA, Rogers DF, Barnes PJEffect of inhaled ozone on exhaled nitric oxide, pulmonary function, and induced sputum in normal and asthmatic subjects. Thorax54199910611069
78. Olin AC, Ljungkvist G, Bake B, Hagberg S, Henriksson L, Toren KExhaled nitric oxide among pulpmill workers reporting gassing incidents involving ozone and chlorine dioxide. Eur Respir J141999828831
79. van Amsterdam JG, Verlaan BP, van Loveren H, Elzakker BG, Vos SG, Opperhuizen A, Steerenberg PAAir pollution is associated with increased level of exhaled nitric oxide in nonsmoking healthy subjects. Arch Environ Health541999331335
80. Kharitonov SA, Robbins RA, Yates DH, Keatings V, Barnes PJAcute and chronic effects of cigarette smoking on exhaled nitric oxide. Am J Respir Crit Care Med1521995609612
81. Robbins RA, Floreani AA, Von Essen SG, Sisson JH, Hill GE, Rubinstein I, Townley RMeasurement of exhaled nitric oxide by three different techniques. Am J Respir Crit Care Med153199616311635
82. Yates DH, Kharitonov SA, Robbins RA, Thomas PS, Barnes PJThe effect of alcohol ingestion on exhaled nitric oxide. Eur Respir J9199611301133
83. Persson MG, Gustafsson LEEthanol can inhibit nitric oxide production. Eur Respir J224199299100
84. Kharitonov SA, Yates DH, Barnes PJ. Increased nitric oxide in exhaled air of normal human subjects with upper respiratory infections. Eur Respir J 1995;295–297.
85. Murphy AW, Platt-Mills TA, Lobo M, Hayden FRespiratory nitric oxide levels in experimental human influenza. Chest1141999452456
86. Ferguson EA, Eccles RChanges in nasal nitric oxide concentration associated with symptoms of common cold and treatment with a topical nasal decongestant. Acta Otolaryngol1171997614617
87. Persson MG, Zetterstrom O, Agrenius V, Ihre E, Gustafsson LESingle-breath nitric oxide measurements in asthmatic patients and smokers. Lancet3431994146147
88. Chatkin JM, Ansarin K, Silkoff PE, McClean P, Gutierrez C, Zamel N, Chapman KRExhaled nitric oxide as a noninvasive assessment of chronic cough. Am J Respir Crit Care Med159199918101813
89. Dupont LJ, Demedts MG, Verleden GMProspective evaluation of the accuracy of exhaled nitric oxide for the diagnosis of asthma [abstract]. Am J Respir Crit Care Med1591999A861
90. Ludviksdottir D, Janson C, Hogman M, Hedenstrom H, Bjornsson E, Boman GExhaled nitric oxide and its relationship to airway responsiveness and atopy in asthma: BHR-Study Group. Respir Med931999552556
91. Ho LP, Wood FT, Robson A, Innes JA, Greening APAtopy influences exhaled nitric oxide levels in adult asthmatics. Chest118200013271331
92. Silvestri M, Spallarossa D, Yourukova VF, Battistini E, Fregonese B, Rossi GAOrally exhaled nitric oxide levels are related to the degree of blood eosinophilia in atopic children with mild-intermitten asthma. Eur Respir J131999321326
93. Moody A, Fergusson W, Wells A, Bartley J, Kolbe JIncreased nitric oxide production in the respiratory tract in asymptomatic Pacific Islanders: an association with skin prick reactivity to house dust mite. J Allergy Clin Immunol1052000895899
94. Sovijärvi ARA, Saarinen A, Helin T, Malmberg P, Haahtela T, Linholm H, Laitinen LAIncreased nitric oxide in exhaled air in patients with asthmatic symptoms not fulfilling the functional criteria of asthma. Eur Respir J121998431S
95. Withers NJ, Bale KL, Laszlo GLevels of exhaled nitric oxide as a screening tool for undiagnosed asthma: results of a pilot study. Eur Respir J121998393S
96. van Den Toorn LM, Prins JB, Overbeek SE, Hoogsteden HC, de Jongste JCAdolescents in clinical remission of atopic asthma have elevated exhaled nitric oxide levels and bronchial hyperresponsiveness. Am J Respir Crit Care Med1622000953957
97. Adisesh LA, Kharitonov SA, Yates DH, Snashal DC, Newman-Taylor AJ, Barnes PJExhaled and nasal nitric oxide is increased in laboratory animal allergy. Clin Exp Allergy281998876880
98. Stirling RG, Kharitonov SA, Campbell D, Robinson D, Durham SR, Chung KF, Barnes PJExhaled NO is elevated in difficult asthma and correlates with symptoms and disease severity despite treatment with oral and inhaled corticosteroids. Thorax53199810301034
99. van Amsterdam JG, Verlaan AP, van Loveren H, Vos SG, Opperhuizen A, Steerenberg PAThe balloon technique: a convenient method to measure exhaled NO in epidemiological studies. Int Arch Occup Environ Health721999404407
100. Henriksen AH, Lingaas-Holmen T, Sue-Chu M, Bjermer LCombined use of exhaled nitric oxide and airway hyperresponsiveness in characterizing asthma in a large population survey. Eur Respir J152000849855
101. Horvath I, Barnes PJExhaled monoxides in asymptomatic atopic subjects. Clin Exp Allergy29199912761280
102. Frank TL, Adisesh A, Pickering AC, Morrison JFJ, Wright T, Francis H, Fletcher A, Frank PI, Hannaford PRelationship between exhaled nitric oxide and childhood asthma. Am J Respir Crit Care Med158199810321036
103. Kharitonov SA, O'Connor BJ, Evans DJ, Barnes PJAllergen-induced late asthmatic reactions are associated with elevation of exhaled nitric oxide. Am J Respir Crit Care Med151199518941899
104. Paredi P, Leckie MJ, Horvath I, Allegra L, Kharitonov SA, Barnes PJExhaled carbon monoxide is elevated following allergen challenge in patients with asthma. Eur Respir J1319994852
105. Baraldi E, Carra S, Dario C, Azzolin N, Ongarro R, Marcer G, Zacchello FEffect of natural grass pollen exposure on exhaled nitric oxide in asthmatic children. Am J Respir Crit Care Med1591999262266
106. Piacentini GL, Bodini A, Costella S, Vicentini L, Mazzi P, Suzuki Y, Peroni D, Boner ALExhaled nitric oxide in asthmatic children exposed to relevant allergens: effect of flunisolide. Eur Respir J152000730734
107. Simpson A, Custovic A, Pipis S, Adisesh A, Faragher B, Woodcock AExhaled nitric oxide, sensitization, and exposure to allergens in patients with asthma who are not taking inhaled steroids. Am J Respir Crit Care Med16019994549
108. Salome CM, Roberts AM, Brown NJ, Dermand J, Marks GB, Woolcock AJExhaled nitric oxide measurements in a population sample of young adults. Am J Respir Crit Care Med1591999911916
109. Steerenberg PA, Snelder JB, Fischer PH, Vos JG, van Loveren H, van Amsterdam JGCIncreased exhaled nitric oxide on days with high outdoor air pollution is of endogenous origin. Eur Respir J131999334337
110. Jenkins HS, Devalia JL, Mister RL, Bevan AM, Rusznak C, Davies RJThe effect of eposure to ozone and nitrogen dioxide on the airway response of atopic asthmatics to inhaled allergen. Dose- and time- dependent effects. Am J Respir Crit Care Med16019993339
111. Pizzichini E, Pizzichini MM, Kidney JC, Efthimiadis A, Hussack P, Popov T, Cox G, Dolovich J, O'Byrne P, Hargreave FEInduced sputum, bronchoalveolar lavage and blood from mild asthmatics: inflammatory cells, lymphocyte subsets and soluble markers compared. Eur Respir J111998828834
112. Kharitonov SA, Yates DH, Barnes PJInhaled glucocorticoids decrease nitric oxide in exhaled air of asthmatic patients. Am J Respir Crit Care Med1531996454457
113. Kharitonov SA, Yates DH, Chung KF, Barnes PJChanges in the dose of inhaled steroid affect exhaled nitric oxide levels in asthmatic patients. Eur Respir J91996196201
114. Jatakanon A, Lim S, Barnes PJChanges in sputum eosinophils predict loss of asthma control. Am J Respir Crit Care Med16120006472
115. Kharitonov SA, Barnes PJ, O'Connor BJReduction in exhaled nitric oxide after a single dose of nebulized budesonide in patients with asthma [abstract]. Am J Respir Crit Care Med1531996A799
116. Lim S, Jatakanon A, John M, Gilbey T, O'Connor BJ, Barnes PJEffect of inhaled budesonide on lung function and airway inflammation. Am J Respir Crit Care Med15919992230
117. Jatakanon A, Kharitonov SA, Lim S, Barnes PJEffect of differing doses of inhaled budesonide on markers of airway inflammation in patients with mild asthma. Thorax541999108114
118. van Rensen EL, Straathof KC, Veselic-Charvat MA, Zwinderman AH, Bel EH, Sterk PJEffect of inhaled steroids on airway hyperresponsiveness, sputum eosinophils, and exhaled nitric oxide levels in patients with asthma. Thorax541999403408
119. Silkoff PE, McClean PA, Slutsky AS, Caramori M, Chapman KR, Gutierrez C, Zamel NExhaled nitric oxide and bronchial reactivity during and after inhaled beclomethasone in mild asthma. J Asthma351998473479
120. Kharitonov SA, Donnelly LE, Corradi M, Montuschi P, Barnes PJDose-dependent onset and duration of action of 100/400 mcg budesonide on exhaled nitric oxide and related changes in other potential markers of airway inflammation in mild asthma [abstract]. Am J Respir Crit Care Med1612000A186
121. Jatakanon A, Uasuf CG, Maziak W, Lim S, Chung KF, Barnes PJNeutrophilic inflammation in severe persistent asthma. Am J Respir Crit Care Med160199915321539
122. Artlich A, Busch T, Lewandowski K, Jonas S, Gortner L, Falke KJChildhood asthma: exhaled nitric oxide in relation to clinical symptoms. Eur Respir J13199913961401
123. Massaro AF, Gaston B, Kita D, Fanta C, Stamler JS, Drazen JMExpired nitric oxide levels during treatment of acute asthma. Am J Respir Crit Care Med1521995800803
124. Baraldi E, Dario C, Ongaro R, Scollo M, Azzolin NM, Panza N, Paganini N, Zacchello FExhaled nitric oxide concentrations during treatment of wheezing exacerbation in infants and young children. Am J Respir Crit Care Med159199912841288
125. Baraldi E, Azzolin NM, Zanconato S, Dario C, Zacchello FCorticosteroids decrease exhaled nitric oxide in children with acute asthma. J Pediatr1311997381385
126. Lanz MJ, Leung DY, White CWComparison of exhaled nitric oxide to spirometry during emergency treatment of asthma exacerbations with glucocorticosteroids in children. Ann Allergy Asthma Immunol821999161164
127. Lanz MJ, Leung DY, McCormick DR, Harbeck R, Szefler SJ, White CWComparison of exhaled nitric oxide, serum eosinophilic cationic protein, and soluble interleukin-2 receptor in exacerbations of pediatric asthma. Pediatr Pulmonol241997305311
128. Sippel JM, Holden WE, Tilles SA, O'Holloren M, Cook J, Thukkani N, Priest J, Nelson B, Osbourne MLExhaled nitric oxide levels correlate with measures of disease control in asthma. J Allergy Clin Immunol1062000645650
129. Barnes PJ, Lim SInhibitory cytokines in asthma. Mol Med Today41998452458
130. Itano H, Zhang W, Ritter JH, McCarthy TJ, Mohanakumar T, Patterson GAAdenovirus-mediated gene transfer of human interleukin 10 ameliorates reperfusion injury of rat lung isografts. J Thorac Cardiovasc Surg1202000947956
131. Gibson PG, Henry RL, Thomas PNoninvasive assessment of airway inflammation in children: induced sputum, exhaled nitric oxide, and breath condensate. Eur Respir J16200010081015
132. Mattes J, von Storm G, Reining U, Alving K, Ihorst G, Henschen M, Kuehr JNO in exhaled air is correlated with markers of eosinophilic airway inflammation in corticosteroid-dependent childhood asthma. Eur Respir J13199913911395
133. Jatakanon A, Lim S, Kharitonov SA, Chung KF, Barnes PJCorrelation between exhaled nitric oxide, sputum eosinophils, and methacholine responsiveness in patients with mild asthma. Thorax5319989195
134. Dupont LJ, Rochette F, Demedts MG, Verleden GMExhaled nitric oxide correlates with airway hyperresponsiveness in steroid-naive patients with mild asthma. Am J Respir Crit Care Med1571998894898
135. Deykin A, Belostotsky O, Hong C, Massaro AF, Lilly CM, Israel EExhaled nitric oxide following leukotriene E(4) and methacholine inhalation in patients with asthma. Am J Respir Crit Care Med162200016851689
136. Lim S, Jatakanon A, Meah S, Oates T, Chung KF, Barnes PJRelationship between exhaled nitric oxide and mucosal eosinophilic inflammation in mild to moderately severe asthma. Thorax552000184188
137. Sato K, Sumino H, Sakamaki T, Sakamoto H, Nakamura T, Ono Z, Nagai RLack of inhibitory effect of dexamethasone on exhalation of nitric oxide by healthy humans. Intern Med351996356361
138. Little SA, Chalmers GW, MacLeod KJ, McSharry C, Thomson NCNon-invasive markers of airway inflammation as predictors of oral steroid responsiveness in asthma. Thorax552000232234
139. Aziz I, Wilson AM, Lipworth BJEffects of once-daily formoterol and budesonide given alone or in combination on surrogate inflammatory markers in asthmatic adults. Chest118200010491058
140. Wilson AM, Lipworth BJDose-response evaluation of the therapeutic index for inhaled budesonide in patients with mild-to-moderate asthma. Am J Med1082000269275
141. Griese M, Koch M, Latzin P, Beck JAsthma severity, recommended changes of inhaled therapy and exhaled nitric oxide in children: a prospective, blinded trial. Eur J Med Res52000334340
142. Yates DH, Kharitonov SA, Barnes PJEffect of short- and long-acting inhaled beta2-agonists on exhaled nitric oxide in asthmatic patients. Eur Respir J10199714831488
143. Lipworth BJ, Dempsey OJ, Aziz I, Wilson AMEffects of adding a leukotriene antagonist or a long-acting beta(2)-agonist in asthmatic patients with the glycine-16 beta(2)-adrenoceptor genotype. Am J Med1092000114121
144. Yates DH, Kharitonov SA, Barnes PJEffect of short- and long-acting inhaled beta2-agonists on exhaled nitric oxide in asthmatic patients. Eur Respir J10199714831488
145. Garnier P, Fajac I, Dessanges JF, Dall'ava-Santucci J, Lockhart A, Dinh-Xuan ATExhaled nitric oxide during acute changes of airways calibre in asthma. Eur Respir J9199611341138
146. Fuglsang G, Vikre JJ, Agertoft L, Pedersen SEffect of salmeterol treatment on nitric oxide level in exhaled air and dose-response to terbutaline in children with mild asthma. Pediatr Pulmonol251998314321
147. Wallin A, Sandstrom T, Soderberg M, Howarth P, Lundback B, Della-Cioppa G, Wilson S, Judd M, Djukanovic R, Holgate S, Lindberg A, Larssen L, Melander BThe effects of regular inhaled formoterol, budesonide, and placebo on mucosal inflammation and clinical indices in mild asthma. Am J Respir Crit Care Med15919997986
148. Ho LP, Wood FT, Robson A, Innes JA, Greening APThe current single exhalation method of measuring exhales nitric oxide is affected by airway calibre. Eur Respir J15200010091013
149. Kobayashi H, Takahashi Y, Mitsufuji H, Hataishi R, Cui T, Tanaka N, Kawakami T, Tomita TDecreased exhaled nitric oxide in mild persistent asthma patients treated with a leukotriene receptor antagonist, pranlukast. Jpn J Physiol491999541544
150. Bisgaard H, Loland L, Oj JANO in exhaled air of asthmatic children is reduced by the leukotriene receptor antagonist montelukast. Am J Respir Crit Care Med160199912271231
151. Wilson AM, Orr LC, Sims EJ, Dempsey OJ, Lipworth BJAntiasthmatic effects of mediator blockade versus topical corticosteroids in allergic rhinitis and asthma. Am J Respir Crit Care Med162200012971301
152. Bratton DL, Lanz MJ, Miyazawa N, White CW, Silkoff PEExhaled nitric oxide before and after montelukast sodium therapy in school-age children with chronic asthma: a preliminary study. Pediatr Pulmonol281999402407
153. Gomez FP, Barbera JA, Roca J, Iglesia R, Ribas J, Barnes PJ, Rodriguez-Roisin REffect of nitric oxide synthesis inhibition with nebulized l-NAME on ventilation-perfusion distributions in bronchial asthma. Eur Respir J121998865871
154. D'Acquisto F, Sautebin L, Iuvone T, Di Rosa M, Carnuccio RProstaglandins prevent inducible nitric oxide synthase protein expression by inhibiting nuclear factor-kappaB activation in J774 macrophages. FEBS Lett44019987680
155. Kharitonov SA, Sapienza MA, Barnes PJ, Chung KFProstaglandins E2 and F reduce exhaled nitric oxide in normal and asthmatic subjects irrespective of airway calibre changes. Am J Respir Crit Care Med158199813741378
156. Attur MG, Patel R, Thakker G, Vyas P, Levartovsky D, Patel P, Naqvi S, Raza R, Patel K, Abramson D, Bruno G, Abramson SB, Amin ARDifferential anti-inflammatory effects of immunosuppressive drugs: cyclosporin, rapamycin and FK-506 on inducible nitric oxide synthase, nitric oxide, cyclooxygenase-2 and PGE2 production. Inflamm Res4920002026
157. Vandivier RW, Eidsath A, Banks SM, Preas HL, Leighton SB, Godin PJ, Suffredini AF, Danner RLDown-regulation of nitric oxide production by ibuprofen in human volunteers. J Pharmacol Exp Ther289199913981403
158. Tamaoki J, Nakata J, Nishimura K, Kondo M, Aoshiba K, Kawatani K, Nagai AEffect of inhaled indomethacin in asthmatic patients taking high doses of inhaled corticosteroids. J Allergy Clin Immunol105200011341139
159. Oliver B, Tomita K, Meah S, Kelly C, Keller A, Ching KF, Barnes PJ, Lim SThe effect of low dose theophylline on cytokine production in alveolar macrophages in patients with mild asthma [abstract]. Am J Respir Crit Care Med1612000A614
160. Borish LC, Nelson HS, Lanz MJ, Claussen L, Whitmore JB, Agosti JM, Garrison LInterleukin-4 receptor in moderate atopic asthma. A phase i/ii randomized, placebo-controlled trial. Am J Respir Crit Care Med160199918161823
161. Rutgers SR, van der Mark TW, Coers W, Moshage H, Timens W, Kauffman HF, Koeter GH, Postma DSMarkers of nitric oxide metabolism in sputum and exhaled air are not increased in chronic obstructive pulmonary disease. Thorax541999576580
162. Von Essen SG, Scheppers LA, Robbins RA, Donham KJRespiratory tract inflammation in swine confinement workers studied using induced sputum and exhaled nitric oxide. J Clin Toxicol361998557565
163. Verleden GM, Dupont LJ, Verpeut AC, Demedts MGThe effect of cigarette smoking on exhaled nitric oxide in mild steroid-naive asthmatics. Chest11619995964
164. Su Y, Han W, Giraldo C, De Li Y, Block EREffect of cigarette smoke extract on nitric oxide synthase in pulmonary artery endothelial cells. Am J Respir Crit Care Med191998819825
165. Eiserich JP, Hristova M, Cross CE, Jones AD, Freeman BA, Halliwell B, van-der Vliet VAFormation of nitric oxide-derived inflammatory oxidants by myeloperoxidase in neutrophils. Nature3911998393397
166. Maziak W, Loukides S, Culpitt SV, Sullivan P, Kharitonov SA, Barnes PJExhaled nitric oxide in chronic obstructive pulmonary disease. Am J Respir Crit Care Med15719989981002
167. Saetta M, Di SA, Maestrelli P, Turato G, Ruggieri MP, Roggeri A, Calcagni P, Mapp CE, Ciaccia A, Fabbri LMAirway eosinophilia in chronic bronchitis during exacerbations. Am J Respir Crit Care Med150199416461652
168. Clini E, Cremona G, Campana M, Scotti C, Pagani M, Bianchi L, Giordano A, Ambrosino NProduction of endogenous nitric oxide in chronic obstructive pulmonary disease and patients with cor pulmonale: correlates with echo-Doppler assessment. Am J Respir Crit Care Med1622000446450
169. Fujimoto K, Kubo K, Yamamoto H, Yamaguchi S, Matsuzawa YEosinophilic inflammation in the airway is related to glucocorticoid reversibility in patients with pulmonary emphysema. Chest1151999697702
170. Papi A, Romagnoli M, Baraldo S, Braccioni F, Guzzinati I, Saetta M, Ciaccia A, Fabbri LMPartial reversibility of airflow limitation and increased exhaled NO and sputum eosinophilia in chronic obstructive pulmonary disease. Am J Respir Crit Care Med162200017731777
171. Balfour-Lynn IM, Laverty A, Dinwiddie RReduced upper airway nitric oxide in cystic fibrosis. Arch Dis Child751996319322
172. Jones KL, Bryan TW, Jinkins PA, Grisham PA, Owens SA, Milligan SA, Markewitz BA, Robbins RASuperoxide causes a reduction in nitric oxide gas and an increase in nitrate. Am J Physiol2751998L1120L1126
173. Yu H, Nasr SZ, Deretic VInnate lung defenses and compromised Pseudomonas aeruginosa clearance in the malnourished mouse model of respiratory infections in cystic fibrosis. Infect Immun68200021422147
174. Grasemann H, Michler E, Wallot M, Ratjen FDecreased concentration of exhaled nitric oxide (NO) in patients with cystic fibrosis. Pediatr Pulmonol241997173177
175. Thomas SR, Kharitonov SA, Scott SF, Hodson ME, Barnes PJNasal and exhaled nitric oxide is reduced in adult patients with cystic fibrosis and does not correlate with cystic fibrosis genotype. Chest117200010851089
176. Antuni JD, Kharitonov SA, Hughes D, Hodson ME, Barnes PJIncrease in exhaled carbon monoxide during exacerbations of cystic fibrosis. Thorax552000138142
177. Downey D, Elborn JSNitric oxide, iNOS, and inflammation in cystic fibrosis. J Pathol1902000115116
178. Kelley TJ, Drumm MLInducible nitric oxide synthase expression is reduced in cystic fibrosis murine and human airway epithelial cells. J Clin Invest102199812001207
179. Meng QH, Polak JM, Edgar AJ, Chacon MR, Evans TJ, Gruenert DC, Bishop AENeutrophils enhance expression of inducible nitric oxide synthase in human normal but not cystic fibrosis bronchial epithelial cells. J Pathol1902000126132
180. Grasemann H, Knauer N, Buscher R, Hubner K, Drazen JM, Ratjen FAirway nitric oxide levels in cystic fibrosis patients are related to a polymorphism in the neuronal nitric oxide synthase gene. Am J Respir Crit Care Med162200021722176
181. Davis PB, Drumm M, Konstan MWCystic fibrosis. Am J Respir Crit Care Med154199612291256
182. Johannesson M, Ludviksdottir D, Janson CLung function changes in relation to menstrual cycle in females with cystic fibrosis. Respir Med94200010431046
183. Kharitonov SA, Wells AU, O'Connor BJ, Cole PJ, Hansell DM, Logan-Sinclair RB, Barnes PJElevated levels of exhaled nitric oxide in bronchiectasis. Am J Respir Crit Care Med151199518891893
184. Tracey WR, Xue C, Klinghofer V, Barlow J, Pollock JS, Forstermann U, Johns RAImmunocytochemical detection of inducible NO synthase in human lung. Am J Physiol2661994L722L727
185. Ho LP, Innes JA, Greening APExhaled nitric oxide is not elevated in the inflammatory airways diseases of cystic fibrosis and bronchiectasis. Eur Respir J12199812901294
186. Loukides S, Kharitonov SA, Wodehouse T, Cole PJ, Barnes PJEffect of l-arginine on mucociliary function in primary ciliary dyskinesia. Lancet3521998371372
187. Horvath I, Loukides S, Wodehouse T, Cole P, Barnes PJExhaled monoxides in patients with primary ciliary dyskinesia [abstract]. Am J Respir Crit Care Med1571998A585
188. Karadag B, James AJ, Gultekin E, Wilson NM, Bush ANasal and lower airway level of nitric oxide in children with primary ciliary dyskinesia. Eur Respir J13199914021405
189. Bush APrimary ciliary dyskinesia. Acta Otorhinolaryngol Belg542000317324
190. Jain B, Rubinstein I, Robbins RA, Leise KL, Sisson JHModulation of airway epithelial cell ciliary beat frequency by nitric oxide. Biochem Biophys Res Commun19119938388
191. Tamaoki J, Taira M, Nishimura K, Nakata J, Nagai AImpairment of airway mucociliary transport in patients with sinobronchial syndrome: Role of nitric oxide. J Aerosol Med132000239244
192. Sawada T, Nishimura T, Saki M, Nagatsu IImmunohistochemical examination of NOS and SOD in nasal mucosa. Acta Otolaryngol Suppl (Stockh)53919988386
193. Turner PJ, Maggs JR, Foreman JCInduction by inhibitors of nitric oxide synthase of hyperresponsiveness in the human nasal airway. Br J Pharmacol1312000363369
194. Sato M, Fukuyama N, Sakai M, Nakazawa HIncreased nitric oxide in nasal lavage fluid and nitrotyrosine formation in nasal mucosa: indices for severe perennial nasal allergy. Clin Exp Allergy281998597605
195. Kang BH, Chen SS, Jou LS, Weng PK, Wang HWImmunolocalization of inducible nitric oxide synthase and 3-nitrotyrosine in the nasal mucosa of patients with rhinitis. Eur Arch Otorhinolaryngol2572000242246
196. Hanazawa T, Antuni JD, Kharitonov SA, Barnes PJIntranasal administration of eotaxin increases nasal eosinophils and nitric oxide in patients with allergic rhinitis. J Allergy Clin Immunol10520005864
197. Cardell LO, Agusti C, Nadel JANitric oxide-dependent neutrophil recruitment: role in nasal secretion. Clin Exp Allergy30200017991803
198. Westerveld GJ, Voss HP, van der Hee RM, de Haan-Koelewijn GJ, den Hartog GJ, Scheeren RA, Bast AInhibition of nitric oxide synthase by nasal decongestants. Eur Respir J162000437444
199. Martin U, Bryden K, Devoy M, Howarth PIncreased levels of exhaled nitric oxide during nasal and oral breathing in subjects with seasonal rhinitis. J Allergy Clin Immunol971996768772
200. Kharitonov SA, Rajakulasingam K, O'Connor B, Durham SR, Barnes PJNasal nitric oxide is increased in patients with asthma and allergic rhinitis and may be modulated by nasal glucocorticoids. J Allergy Clin Immunol9919975864
201. Baraldi E, Azzolin NM, Carra S, Dario C, Marchesini L, Zacchello FEffect of topical steroids on nasal nitric oxide production in children with perennial allergic rhinitis: a pilot study. Respir Med921998558561
202. Henriksen AH, Sue-Chu M, Lingaas Holmen T, Langhammer A, Bjermer LExhaled and nasal NO levels in allergic rhinitis: relation to sensitization, pollen season, and bonchial hyperresponsiveness. Eur Respir J131999301306
203. Kharitonov SA, Cailes JB, Black CM, Du Bois RM, Barnes PJDecreased nitric oxide in the exhaled air of systemic sclerosis patients with pulmonary hypertension. Thorax52199710511055
204. Rolla G, Colagrande P, Scappaticci E, Chiavassa G, Dutto L, Cannizzo S, Bucca C, Morello M, Bergerone S, Bardini D, Zaccagna A, Puiatti P, Fava C, Cortese GExhaled nitric oxide in systemic sclerosis: relationships with lung involvement and pulmonary hypertension. J Rheumatol27200016931698
205. Giaid A, Saleh DReduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N Engl J Med3331995214221
206. Hislop AA, Springall DR, Oliveira H, Pollock JS, Polak JM, Haworth SGEndothelial nitric oxide synthase in hypoxic newborn porcine pulmonary vessels. Arch Dis Child Fetal Neonatal Ed771997F16F22
207. Giaid ANitric oxide and endothelin-1 in pulmonary hypertension. Chest1141998208S212S
208. Black SM, Fineman JR, Steinhorn RH, Bristow J, Soifer SJIncreased endothelial NOS in lambs with increased pulmonary blood flow and pulmonary hypertension. Am J Physiol2751998H1643H1651
209. Tyler RC, Muramatsu M, Abman SH, Stelzner TJ, Rodman DM, Bloch KD, McMurtry IFVariable expression of endothelial NO synthase in three forms of rat pulmonary hypertension. Am J Physiol2761999L297L303
210. Everett AD, Le CT, Xue C, Johns RAeNOS expression is not altered in pulmonary vascular remodeling due to increased pulmonary blood flow. Am J Physiol2741998L1058L1065
211. Saleh D, Barnes PJ, Giaid AIncreased production of the potent oxidant peroxynitrite in the lungs of patients with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med155199717631769
212. Paredi P, Kharitonov SA, Loukides S, Pantelidis P, Du Bois RM, Barnes PJExhaled nitric oxide is increased in active fibrosing alveolitis. Chest115199913521356
213. Moodley YP, Chetty R, Lalloo UGNitric oxide levels in exhaled air and inducible nitric oxide synthase immunolocalization in pulmonary sarcoidosis. Eur Respir J141999822827
214. O'Donnell DM, Moynihan J, Finlay GA, Keatings VM, O'Connor CM, McLoughlin P, Fitzgerald MXExhaled nitric oxide and bronchoalveolar lavage nitrite/nitrate in active pulmonary sarcoidosis. Am J Respir Crit Care Med156199718921896
215. Forrest IA, Small T, Corris PAEffect of nebulized epoprostenol (prostacyclin) on exhaled nitric oxide in patients with pulmonary hypertension due to congenital heart disease and in normal controls. Clin Sci97199999102
216. Sumino H, Nakamura T, Kanda T, Sato K, Sakamaki T, Takahashi T, Saito Y, Hoshino J, Kurashina T, Nagai REffect of enalapril on exhaled nitric oxide in normotensive and hypertensive subjects. Hypertension362000934940
217. Kaneko FT, Arroliga AC, Dweik RA, Comhair SA, Laskowski D, Oppedisano R, Thomassen MJ, Erzurum SCBiochemical reaction products of nitric oxide as quantitative markers of primary pulmonary hypertension. Am J Respir Crit Care Med1581998917923
218. Budts W, Pokreisz P, Nong Z, Van Pelt N, Gillijns H, Gerard R, Lyons R, Collen D, Bloch KD, Janssens SAerosol gene transfer with inducible nitric oxide synthase reduces hypoxic pulmonary hypertension and pulmonary vascular remodeling in rats. Circulation102200028802885
219. Berg JT, Deem S, Kerr ME, Swenson ERHemoglobin and red blood cells alter the response of expired nitric oxide to mechanical forces. Am J Physiol Heart Circ Physiol2792000H2947H2953
220. van Amsterdam JG, Nierkens S, Vos SG, Opperhuizen A, van Loveren H, Steerenberg PAExhaled nitric oxide: a novel biomarker of adverse respiratory health effects in epidemiological studies. Arch Environ Health552000418423
221. Lund MB, Oksne PI, Hamre R, Kongerud JIncreased nitric oxide in exhaled air: an early marker of asthma in non-smoking aluminium potroom workers? Occup Environ Med572000274278
222. Allmers H, Chen Z, Barbinova L, Marczynski B, Kirschmann V, Baur XChallenge from methacholine, natural rubber latex, or 4,4-diphenylmethane diisocyanate in workers with suspected sensitization affects exhaled nitric oxide [change in exhaled NO levels after allergen challenges]. Int Arch Occup Environ Health732000181186
223. Haubitz M, Busch T, Gerlach M, Schafer S, Brunkhorst R, Falke K, Koch KM, Gerlach HExhaled nitric oxide in patients with Wegener's granulomatosis. Eur Respir J141999113117
224. Murphy AW, Platts MT, Lobo M, Hayden FRespiratory nitric oxide levels in experimental human influenza. Chest1141998452456
225. de Gouw HW, Grunberg K, Schot R, Kroes AC, Dick EC, Sterk PJRelationship between exhaled nitric oxide and airway hyperresponsiveness following experimental rhinovirus infection in asthmatic subjects. Eur Respir J111998126132
226. Saura M, Zaragoza C, McMillan A, Quick RA, Hohenadl C, Lowenstein JM, Lowenstein CJAn antiviral mechanism of nitric oxide: inhibition of a viral protease. Immunity1019992128
227. Zhu Z, Tang W, Ray A, Wu Y, Einarsson O, Landry ML, Gwaltney JJ, Elias JARhinovirus stimulation of interleukin-6 in vivo and in vitro. Evidence for nuclear factor kappa B-dependent transcriptional activation. J Clin Invest971996421430
228. Loveless MO, Phillips CR, Giraud GD, Holden WEDecreased exhaled nitric oxide in subjects with HIV infection. Thorax521997185186
229. Palm J, Lidman C, Graf P, Alving K, Lundberg J. Nasal nitric oxide is reduced in patients with HIV. Acta Otolaryngol 2000; 120:420–423.
230. Evans TG, Rasmussen K, Wiebke G, Hibbs JBJNitric oxide synthesis in patients with advanced HIV infection. Clin Exp Immunol9719948386
231. Barton CH, Biggs TE, Mee TR, Mann DAThe human immunodeficiency virus type 1 regulatory protein Tat inhibits interferon-induced iNos activity in a murine macrophage cell line. J Gen Virol77199616431647
232. Long R, Light B, Talbot JAMycobacteriocidal action of exogenous nitric oxide. Antimicrob Agents Chemother431999403405
233. Wang CH, Liu CY, Lin HC, Yu CT, Chung KF, Kuo HPIncreased exhaled nitric oxide in active pulmonary tuberculosis due to inducible NO synthase upregulation in alveolar macrophages. Eur Respir J111998809815
234. Grasemann H, Ioannidis I, de Groot H, Ratjen FMetabolites of nitric oxide in the lower respiratory tract of children. Eur J Pediatr1561997575578
235. Parameswaran K, Kamada D, Borm A, Efthimiadis A, Allen C, Anvari M, Hargreave FESputum cell counts and exhaled nitric oxide in patients with non-asthmatic cough and gastro-esophageal reflux. Eur Respir J121998248S
236. Liu CY, Wang CH, Chen TC, Lin HC, Yu CT, Kuo HPIncreased level of exhaled nitric oxide and up-regulation of inducible nitric oxide synthase in patients with primary lung cancer. Br J Cancer781998534541
237. Marczin N, Riedel B, Gal J, Polak J, Yacoub MExhaled nitric oxide during lung transplantation. Lancet350199716811682
238. Pearl JM, Nelson DP, Wellmann SA, Raake JL, Wagner CJ, McNamara JL, Duffy JYAcute hypoxia and reoxygenation impairs exhaled nitric oxide release and pulmonary mechanics. J Thorac Cardiovasc Surg1192000931938
239. Fisher AJ, Gabbay E, Small T, Doig S, Dark JH, Corris PACross sectional study of exhaled nitric oxide levels following lung transplantation. Thorax531998454458
240. Gabbay E, Haydn WE, Orsida B, Whitford H, Ward C, Kotsimbos TC, Snell GI, Williams TJIn stable lung transplant recipients, exhaled nitric oxide levels positively correlate with airway neutrophilia and bronchial epithelial iNOS. Am J Respir Crit Care Med160199920932099
241. Silkoff PE, Caramori M, Tremblay L, McClean P, Chaparro C, Kesten S, Hutcheon M, Slutsky AS, Zamel N, Keshavjee SExhaled nitric oxide in human lung transplantation: a noninvasive marker of acute rejection. Am J Respir Crit Care Med157199818221828
242. Stewart TE, Valenza F, Ribeiro SP, Wener AD, Volgyesi G, Mullen JB, Slutsky ASIncreased nitric oxide in exhaled gas as an early marker of lung inflammation in a model of sepsis. Am J Respir Crit Care Med1511995713718
243. Brett SJ, Evans TWMeasurement of endogenous nitric oxide in the lungs of patients with the acute respiratory distress syndrome. Am J Respir Crit Care Med1571998993997
244. Ishibe Y, Liu R, Hirosawa J, Kawamura K, Yamasaki K, Saito NExhaled nitric oxide level decreases after cardiopulmonary bypass in adult patients. Crit Care Med28200038233827
245. Nakano H, Ide H, Imada M, Osanai S, Takahashi T, Kikuchi K, Iwamoto JReduced nasal nitric oxide in diffuse panbronchiolitis. Am J Respir Crit Care Med162200022182220
246. Berk PD, Rodkey FL, Blaschke TF, Collison HA, Waggoner JGComparison of plasma bilirubin turnover and carbon monoxide production in man. J Lab Clin Med8319742937
247. Levine AS, Bond JH, Prentiss RA, Levitt MDMetabolism of carbon monoxide by the colonic flora of humans. Gastroenterology831982633637
248. Coburn RFEndogenous carbon monoxide production. N Engl J Med2821970207209
249. Vreman HJ, Baxter LM, Stone RT, Stevenson DKEvaluation of a fully automated end-tidal carbon monoxide instrument for breath analysis. Clin Chem4219965056
250. Coburn RFEndogenous carbon monoxide metabolism. Annu Rev Med241973241250
251. Kharitonov SA, Lim S, Hanazawa T, Chung FK, Barnes PJExhaled carbon monoxide derives predominantly from alveoli in healthy non-smokers, smokers and mild stable asthmatics, but also from asthmatic airways after allergen challenge [abstract]. Am J Respir Crit Care Med1612000A584
252. Kharitonov SA, Paredi P, Barnes PJMethodological aspects of exhaled carbon monoxide measurements as a possible non-invasive marker of oxidative stress: influence of exhalation flow, breathholding and ambient air. Eur Respir J121998128s
253. Andersson JA, Uddman R, Cardell LOCarbon monoxide is endogenously produced in the human nose and paranasal sinuses. J Allergy Clin Immunol1052000269273
254. McCoubrey WKJ, Huang TJ, Maines MDIsolation and characterization of a cDNA from the rat brain that encodes hemoprotein heme oxygenase-3. Eur J Biochem2471997725732
255. Choi AM, Alam JHeme oxygenase-1: function, regulation, and implication of a novel stress-inducible protein in oxidant-induced lung injury. Am J Respir Crit Care Med151996919
256. Lim S, Groneberg D, Fischer A, Oates T, Caramori G, Mattos W, Adcock I, Barnes PJ, Chung KFExpression of heme oxygenase isoenzymes 1 and 2 in normal and asthmatic airways: effect of inhaled corticosteroids. Am J Respir Crit Care Med162200019121918
257. Chakder S, Rathi S, Ma XL, Rattan SHeme oxygenase inhibitor zinc protoporphyrin IX causes an activation of nitric oxide synthase in the rabbit internal anal sphincter. J Pharmacol Exp Ther277199613761382
258. Datta PK, Lianos EANitric oxide induces heme oxygenase-1 gene expression in mesangial cells. Kidney Int55199917341739
259. Klatt P, Schmidt K, Mayer BBrain nitric oxide synthase is a haemoprotein. Biochem J28819921517
260. Patel RPBiochemical aspects of the reaction of hemoglobin and NO: implications for Hb-based blood substitutes. Free Radic Biol Med28200015181525
261. Privalle C, Talarico T, Keng T, DeAngelo JPyridoxalated hemoglobin polyoxyethylene: a nitric oxide scavenger with antioxidant activity for the treatment of nitric oxide-induced shock. Free Radic Biol Med28200015071517
262. Skrupskii VA, Stepanov VE, Shulagin IuA. Monitoring of endogenous carbon monoxide elimination in exhaled air of rats in hyperoxia. Aviakosm Ekolog Med2919954952
263. Motterlini R, Kerger H, Green CJ, Winslow RM, Intaglietta MDepression of endothelial and smooth muscle cell oxygen consumption by endotoxin. Am J Physiol2751998H776H782
264. Foresti R, Clark JE, Green CJ, Motterlini RThiol compounds interact with nitric oxide in regulating heme oxygenase-1 induction in endothelial cells. Involvement of superoxide and peroxynitrite anions. J Biol Chem27219971841118417
265. Foresti R, Sarathchandra P, Clark JE, Green CJ, Motterlini RPeroxynitrite induces heme oxygenase-1 in vascular endothelial cells: a link to apoptosis. Biochem J3391999729736
266. Otterbein LE, Mantell LL, Choi AMCarbon monoxide provides protection against hyperoxic lung injury. Am J Physiol2761999L688L694
267. Camhi SL, Lee P, Choi AMThe oxidative stress response. New Horiz31995170182
268. Dailly E, Urien S, Barre J, Reinert P, Tillement JPRole of bilirubin in the regulation of the total peroxyl radical trapping antioxidant activity of plasma in sickle cell disease. Biochem Biophys Res Commun2481998303306
269. Suttner DMidhar K, Lee CS, Tomura T, Hansen TN, Dennery PA. Protective effects of transient HO-1 overexpression on susceptibility to oxygen toxicity in lung cells. Am J Physiol2761999L443L451
270. Dennery PA, Spitz DR, Yang G, Tatarov A, Lee CS, Shegog ML, Poss KDOxygen toxicity and iron accumulation in the lungs of mice lacking heme oxygenase-2. J Clin Invest101199810011011
271. Nikberg II, Murashko VA, Leonenko INCarbon monoxide concentration in the air exhaled by the healthy and the ill. Vrach Delo121972112114
272. Kharitonov SA, Paredi P, Barnes PJReproducibility of exhaled carbon monoxide measurements and its circadian variation in normal subjects [abstract]. Am J Respir Crit Care Med1571998A613
273. Chuchalin AG, Voznesenskiy N, Dulin K, Sakharova S, Soodaeva E, Stepanov EExhaled nitric oxide and exhaled carbon monoxide in pulmonary diseases [abstract]. Am J Respir Crit Care Med1591999A410
274. Alving K, Zetterquist W, Wennerholm P, Lundberg JONLow levels of exhaled carbon monoxide in asthmatics using infrared technique [abstract]. Am J Respir Crit Care Med1591999A841
275. Rodgers PA, Vreman HJ, Dennery PA, Stevenson DKSources of carbon monoxide (CO) in biological systems and applications of CO detection technologies. Semin Perinatol181994210
276. Uasuf CG, Jatakanon A, James A, Kharitonov SA, Wilson NM, Barnes PJExhaled carbon monoxide in childhood asthma. J Pediatr1351999569574
277. Middleton ET, Morice AHBreath carbon monoxide as an indication of smoking habit. Chest1172000758763
278. Wald NJ, Idle M, Boreham J, Bailey ACarbon monoxide in breath in relation to smoking and carboxyhaemoglobin levels. Thorax361981366369
279. Tonnesen P, Norregaard J, Mikkelsen K, Jorgensen S, Nilsson FA double-blind trial of a nicotine inhaler for smoking cessation. JAMA269199312681271
280. Stewart RD, Fisher TN, Hosko MJ, Peterson JE, Baretta ED, Dodd HCCarboxyhemoglobin elevation after exposure to dichloromethane. Science1761972295296
281. Zayasu K, Sekizawa K, Okinaga S, Yamaya M, Sasaki HIncreased carbon monoxide in exhaled air of asthmatic patients. Am J Respir Crit Care Med156199711401143
282. Horvath I, Donnelly LE, Kiss A, Paredi P, Kharitonov SA, Barnes PJElevated levels of exhaled carbon monoxide are associated with an increased expression of heme oxygenase-1 in airway macrophages in asthma: a new marker of oxidative stress. Thorax531998668672
283. Kharitonov SAExhaled nitric oxide and carbon monoxide in respiratory diseases other than asthma. Eur Respir J91999223226
284. Yamara M, Sekizawa K, Ishizuka M, Monma M, Sasaki HExhaled carbon monoxide levels during treatment of acute asthma. Eur Respir J131999757760
285. Stirling RG, Lim S, Kharitonov SA, Chung FK, Barnes PJExhaled breath carbon monoxide is minimally elevated in severe but not mild atopic asthma [abstract]. Am J Respir Crit Care Med1612000A922
286. Biernacki W, Kharitonov SA, Barnes PJExhaled carbon monoxide measurements can be used in general practice to predict the response to oral steroid treatment in patients with asthma [abstract]. Am J Respir Crit Care Med1591999A631
287. Delen FM, Sippel JM, Osborne ML, Law S, Thukkani N, Holden WEIncreased exhaled nitric oxide in chronic bronchitis. Comparison with asthma and COPD. Chest1172000695701
288. Culpitt SV, Paredi P, Kharitonov SA, Barnes PJExhaled carbon monoxide is increased in COPD patients regardless of their smoking habit [abstract]. Am J Respir Crit Care Med1571998A787
289. Muller T, Gebel SThe cellular stress response induced by aqueous extracts of cigarette smoke is critically dependent on the intracellular glutathione concentration. Carcinogenesis191998797801
290. Biernacki W, Kharitonov SA, Barnes PJCarbon monoxide in exhaled air in patients with lower respiratory tract infection. Eur Respir J121998345S
291. Horvath I, Loukides S, Wodehouse T, Kharitonov SA, Cole PJ, Barnes PJElevated levels of exhaled carbon monoxide in bronchiectasis: a new marker of oxidative stress. Thorax531998867870
292. Paredi P, Shah PL, Montuschi P, Sullivan P, Hodson ME, Kharitonov SA, Barnes PJIncreased carbon monoxide in exhaled air of cystic fibrosis patients. Thorax541999917920
293. Montuschi P, Kharitonov SA, Ciabattoni G, Corradi M, van Rensen L, Geddes DM, Hodson ME, Barnes PJExhaled 8-isoprostane as a new non-invasive biomarker of oxidative stress in cystic fibrosis. Thorax552000205209
294. Paredi P, Kharitonov SA, Leak D, Shah PL, Cramer D, Hodson ME, Barnes PJExhaled ethane is elevated in cystic fibrosis and correlates with CO levels and airway obstruction. Am J Respir Crit Care Med161200012471251
295. Antuni JD, Du Bois AB, Ward S, Cramer DS, Kharitonov SA, Barnes PJExhaled carbon monoxide may be a marker of deterioration of lung function in cryptogenic fibrosing alveolitis and scleroderma [abstract]. Am J Respir Crit Care Med1591999A51
296. Antuni JD, Ward S, Cramer DS, Kharitonov SA, Barnes PJUptake and elimination of exhaled carbon monoxide in patients with interstitial lung disease is related to the degree of impairment of carbon monoxide diffusion capacity [abstract]. Am J Respir Crit Care Med1591999A86
297. Monma M, Yamaya M, Sekizawa K, Ikeda K, Suzuki N, Kikuchi T, Takasaka T, Sasaki HIncreased carbon monoxide in exhaled air of patients with seasonal allergic rhinitis. Clin Exp Allergy29199915371541
298. Yamaya M, Sekizawa K, Ishizuka S, Monma M, Mizuta K, Sasaki HIncreased carbon monoxide in exhaled air of subjects with upper respiratory tract infections. Am J Respir Crit Care Med1581998311314
299. Scharte M, Bone HG, Van Aken H, Meyer JIncreased CO in exhaled air of critically ill patients. Biochem Biophys Res Commun2672000423426
300. Zegdi R, Caid R, Van De Louw A, Perrin D, Burdin M, Boiteau R, Tenaillon AExhaled carbon monoxide in mechanically ventilated critically ill patients: influence of inspired oxygen fraction. Intensive Care Med26200012281231
301. Paredi P, Biernacki W, Invernizzi G, Kharitonov SA, Barnes PJExhaled carbon monoxide levels elevated in diabetes and correlated with glucose concentration in blood: a new test for monitoring the disease? Chest116199910071011
302. Pauling L, Robinson AB, Teranishi R, Cary PQuantitative analysis of urine vapor and breath by gas-liquid partition chromatography. Proc Natl Acad Sci USA68197123742376
303. Poliakov VV, Ivanova SM, Noskov VB, Labetskaia OI, Iarlykova IV, Karashtin VV, Legenkov VI, Sarycheva TG, Shishkanova ZG, Kozinets GIHematological investigations in conditions of long-term space flights. Aviakosm Ekolog Med321998918
304. Andreoni KA, Kazui M, Cameron DE, Nyhan D, Sehnert SS, Rohde CA, Bulkley GB, Risby THEthane: a marker of lipid peroxidation during cardiopulmonary bypass in humans. Free Radic Biol Med261999439445
305. Phillips M, Gleeson K, Hughes JMB, Greenberg J, Cataneo RN, Baker L, McVay WPVolatile organic compounds in breath as markers of lung cancer: a cross-sectional study. Lancet353199919301933
306. Muller A, Sies HAssay of ethane and pentane from isolated organs and cells. Methods Enzymol1051984311319
307. Kneepkens CM, Lepage G, Roy CCThe potential of the hydrocarbon breath test as a measure of lipid peroxidation. Free Radic Biol Med171994127160
308. Pitkanen OM, Hallman M, Andersson SMCorrelation of free oxygen radical-induced lipid peroxidation with outcome in very low birth weight infants. J Pediatr1161990760764
309. Aulik IVGas chromatographic analysis of exhaled air and acetylene mixture. Biull Eksp Biol Med621966115117
310. Hotz P, Hoet P, Lauwerys R, Buchet JPDevelopment of a method to monitor low molecular mass hydrocarbons in exhaled breath of man: preliminary evaluation of its interest for detecting a lipoperoxidation process in vivo. Clin Exp Allergy1621987303310
311. Kneepkens CM, Ferreira C, Lepage G, Roy CCThe hydrocarbon breath test in the study of lipid peroxidation: principles and practice. Clin Invest Med151992163186
312. Shilov VN, Iakovchenko VA, Sergienko VIDiagnostic value of gas chromatographic study of exhaled air. Klin Lab Diagn51994910
313. Zarling EJ, Clapper MTechnique for gas-chromatographic measurement of volatile alkanes from single-breath samples. Clin Chem331987140141
314. Paredi P, Kharitonov SA, Leak D, Ward S, Cramer D, Barnes PJExhaled ethane, a marker of lipid peroxidation, is elevated in chronic obstructive pulmonary disease. Am J Respir Crit Care Med1622000369373
315. Paredi P, Kharitonov SA, Barnes PJElevation of exhaled ethane concentration in asthma. Am J Respir Crit Care Med162200014501454
316. Nycyk JA, Drury JA, Cooke RWBreath pentane as a marker for lipid peroxidation and adverse outcome in preterm infants. Arch Dis Child Fetal Neonatal Ed791998F67F69
317. Paredi P, Ward S, Cramer D, Kharitonov SA, Barnes PJSingle breath measurement of exhaled ethane [abstract]. Am J Respir Crit Care Med1591999A887
318. Ljungkvist GM, Nordlinder RGA field method for sampling benzene in end-exhaled air. Am Ind Hyg Assoc J561995693697
319. Iatsenko VP, Briuzgina TS, Khomenko VE, Reva SN. Gas chromatographic analysis of lipids in exhaled air condensate in children with bronchopulmonary diseases. Klin Lab Diagn 1997;16–17.
320. Prokhorova MN, Briuzgina TS, Umanets TR, Sokolova IV, Reva SN. The use of noninvasive biological means in assessing lipids in children. Klin Lab Diagn 1998;13–15.
321. Morita S, Snider MT, Inada YIncreased N-pentane excretion in humans: a consequence of pulmonary oxygen exposure. Anesthesiology641986730733
322. Wispe JR, Bell EF, Roberts RJAssessment of lipid peroxidation in newborn infants and rabbits by measurements of expired ethane and pentane: influence of parenteral lipid infusion. Pediatr Res191985374379
323. Allerheiligen SR, Ludden TM, Burk RFThe pharmacokinetics of pentane, a by-product of lipid peroxidation. Drug Metab Dispos151987794800
324. Zarling EJ, Mobarhan S, Bowen P, Sugerman SOral diet does not alter pulmonary pentane or ethane excretion in healthy subjects. J Am Coll Nutr111992349352
325. Refat M, Moore TJ, Kazui M, Risby TH, Perman JA, Schwarz KBUtility of breath ethane as a noninvasive biomarker of vitamin E status in children. Pediatr Res301991396403
326. Habib MP, Clements NC, Garewal HSCigarette smoking and ethane exhalation in humans. Am J Respir Crit Care Med151199513681372
327. Paredi P, Kharitonov SA, Leak D, Ward S, Cramer D, Barnes PJExhaled ethane, a marker of lipid peroxidation, is elevated in chronic obstructive pulmonary disease. Am J Respir Crit Care Med1622000369373
328. Do BK, Garewal HS, Clements-NC J, Peng YM, Habib MPExhaled ethane and antioxidant vitamin supplements in active smokers. Chest1101996159164
329. Ivanova SM, Orlov ON, Brantova SS, Labetskaia OI, Davydova NAEffect of intensive operator activity on lipid peroxidation processes in the human body. Kosm Biol Aviakosm Med2019862022
330. Leaf DA, Kleinman MT, Hamilton M, Barstow TJThe effect of exercise intensity on lipid peroxidation. Med Sci Sports Exerc29199710361039
331. Olopade CO, Zakkar M, Swedler WI, Rubinstein IExhaled pentane levels in acute asthma. Chest1111997862865
332. Paredi P, Kharitonov SA, Barnes PJElevation of exhaled ethane concentration in asthma. Am J Respir Crit Care Med162200014501454
333. Ip MS, Lam B, Chan LY, Zheng L, Tsang KW, Fung PC, Lam WKCirculating nitric oxide is suppressed in obstructive sleep apnea and is reversed by nasal continuous positive airway pressure. Am J Respir Crit Care Med162200021662171
334. Olopade CO, Christon JA, Zakkar M, Hua C, Swedler WI, Scheff PA, Rubinstein IExhaled pentane and nitric oxide levels in patients with obstructive sleep apnea. Chest111199715001504
335. Jeejeebhoy KNIn vivo breath alkane as an index of lipid peroxidation. Free Radic Biol Med101991191193
336. Foster WM, Jiang L, Stetkiewicz PT, Risby THBreath isoprene: temporal changes in respiratory output after exposure to ozone. J Appl Physiol801996706710
337. Habib MP, Tank LJ, Lane LC, Garewal HSEffect of vitamin E on exhaled ethane in cigarette smokers. Chest1151999684690
338. Gordon SMIdentification of exposure markers in smokers' breath. J Chromatogr5111990291302
339. Jo WK, Pack KWUtilization of breath analysis for exposure estimates of benzene associated with active smoking. Environ Res832000180187
340. Wallace LA, Pellizzari ED. Recent advances in measuring exhaled breath and estimating exposure and body burden for volatile organic compounds (VOCs). Environ Health Perspect 1995;103(Suppl 3:95–98).
341. Schubert JK, Muller WP, Benzing A, Geiger KApplication of a new method for analysis of exhaled gas in critically ill patients. Intensive Care Med241998415421
342. Holz O, Richter K, Jorres RA, Speckin P, Mucke M, Magnussen HChanges in sputum composition between two inductions performed on consecutive days. Thorax5319988386
343. Sidorenko GI, Zborovskii EI, Levina DISurface-active properties of the exhaled air condensate (a new method of studying lung function). Ter Arkh5219806568
344. Kurik MV, Rolik LV, Parkhomenko NV, Tarakhan LI, Savitskaia NV. Physical properties of a condensate of exhaled air in chronic bronchitis patients. Vrach Delo 1987;37–39.
345. von Pohle WR, Anholm JD, McMillan JCarbon dioxide and oxygen partial pressure in expiratory water condensate are equivalent to mixed expired carbon dioxide and oxygen. Chest101199216011604
346. Montuschi P, Corradi M, Ciabattoni G, Nightingale J, Kharitonov SA, Barnes PJIncreased 8-isoprostane, a marker of oxidative stress, in exhaled condensate of asthma patients. Am J Respir Crit Care Med1601999216220
347. Scheideler L, Manke HG, Schwulera U, Inacker O, Hammerle HDetection of nonvolatile macromolecules in breath. A possible diagnostic tool? Am Rev Respir Dis1481993778784
348. Mozalevskii AF, Travianko TD, Iakovlev AA, Smirnova EA, Novikova NP, Sapa IIContent of arachidonic acid metabolites in blood and saliva of children with bronchial asthma. Ukr Biokhim Zh691997162168
349. Zetterquist W, Pedroletti C, Lundberg JON, Alving KSalivary contribution to exhaled nitric oxide. Eur Respir J131999327333
350. Horvath I, Donnelly LE, Kiss A, Kharitonov SA, Lim S, Chung FK, Barnes PJCombined use of exhaled hydrogen peroxide and nitric oxide in monitoring asthma. Am J Respir Crit Care Med158199810421046
351. Loukides S, Horvath I, Wodehouse T, Cole PJ, Barnes PJElevated levels of expired breath hydrogen peroxide in bronchiectasis. Am J Respir Crit Care Med1581998991994
352. Ho LP, Innes JA, Greening APNitrite levels in breath condensate of patients with cystic fibrosis is elevated in contrast to exhaled nitric oxide. Thorax531998680684
353. Reinhold P, Langenberg A, Becher G, Rothe MBreath condensate: a medium obtained by a noninvasive method for the detection of inflammation mediators of the lung. Berl Munch Tierarztl Wochenschr1121999254259
354. Freeman BA, Crapo JDBiology of disease: free radicals and tissue injury. Lab Invest471982412426
355. Dohlman AW, Black HR, Royall JAExpired breath hydrogen peroxide is a marker of acute airway inflammation in pediatric patients with asthma. Am Rev Respir Dis1481993955960
356. Heffner JE, Repine JEPulmonary strategies of antioxidant defense. Am Rev Respir Dis1401989531554
357. Godwin JE, Heffner JEPlatelet prevention of oxidant lung oedema is not mediated through scavenging of hydrogen peroxide. Blood Coagul Fibrinolysis31992531539
358. Antczak A, Nowak D, Shariati B, Krol M, Piasecka G, Kurmanowska ZIncreased hydrogen peroxide and thiobarbituric acid-reactive products in expired breath condensate of asthmatic patients. Eur Respir J10199712351241
359. Jöbsis Q, Raatgeep HC, Schellekens SL, Hop WCJ, Hermans PWM, de Jongste JCHydrogen peroxide in exhaled air of healthy children: reference values. Eur Respir J121998483485
360. Antczak A, Nowak D, Bialasiewicz P, Kasielski M. Hydrogen peroxide in expired air condensate correlates positively with early steps of peripheral neutrophil activation in asthmatic patients. Arch Immunol Ther Exp (Warsz) 1999;47:119–126.
361. Nowak D, Antczak A, Krol M, Pietras T, Shariati B, Bialasiewicz P, Jeczkowski K, Kula PIncreased content of hydrogen peroxide in the expired breath of cigarette smokers. Eur Respir J91996652657
362. Dekhuijzen PN, Aben KK, Dekker I, Aarts LP, Wielders PL, van Herwaarden CL, HC, Bast A. Increased exhalation of hydrogen peroxide in patients with stable and unstable chronic obstructive pulmonary disease. Am J Respir Crit Care Med 1996;154:813–816.
363. Nowak D, Kasielski M, Pietras T, Bialasiewicz P, Antczak ACigarette smoking does not increase hydrogen peroxide levels in expired breath condensate of patients with stable COPD. Monaldi Arch Chest Dis531998268273
364. Baldwin SR, Simon RH, Grum CM, Ketai LH, Boxer LA, Devall LJOxidant activity in expired breath of patients with adult respiratory distress syndrome. Lancet119861114
365. Heard SO, Longtine K, Toth I, Puyana JC, Potenza B, Smyrnios NThe influence of liposome-encapsulated prostaglandin E1 on hydrogen peroxide concentrations in the exhaled breath of patients with the acute respiratory distress syndrome. Anesth Analg891999353357
366. Lases EC, Duurkens VA, Gerritsen WB, Haas FJOxidative stress after lung resection therapy: A pilot study. Chest11720009991003
367. Jobsis Q, Raatgeep HC, Schellekens SL, Kroesbergen A, Hop WC, de Jongste JCHydrogen peroxide and nitric oxide in exhaled air of children with cystic fibrosis during antibiotic treatment. Eur Respir J16200095100
368. Taha R, Olivenstein R, Utsumi T, Ernst P, Barnes PJ, Rodger IW, Giaid AProstaglandin H synthase 2 expression in airway cells from patients with asthma and COPD. Am J Respir Crit Care Med1612000636640
369. Kuitert LM, Newton R, Barnes NC, Adcock IM, Barnes PJEicosanoid mediator expression in mononuclear and polymorphonuclear cells in normal subjects and patients with atopic asthma and cystic fibrosis. Thorax51199612231228
370. Pavord ID, Tattersfield AEBronchoprotective role for endogenous prostaglandin E2. Lancet3441994436438
371. Tetsuka T, Morrison ARTyrosine kinase activation is necessary for inducible nitric oxide synthase expression by interleukin-1 beta. Am J Physiol2691995C55C99
372. Montuschi P, Kharitonov SA, Carpagnano E, Culpitt SV, Russell R, Collins JV, Barnes PJExhaled prostaglandin E2: a new biomarker of airway inflammation in COPD [abstract]. Am J Respir Crit Care Med1612000A821
373. Tanaka H, Saito T, Kurokawa K, Teramoto S, Miyazaki N, Kaneko S, Hashimoto M, Abe SLeukotriene (LT)-receptor antagonist is more effective in asthmatic patients with a low baseline ratio of urinary LTE4 to 2,3-dinor-6-keto-prostaglandin (PG)F1alpha. Allergy541999489494
374. Leff ARRole of leukotrienes in bronchial hyperresponsiveness and cellular responses in airways. Am J Respir Crit Care Med1612000S125S132
375. Larfars G, Lantoine F, Devynck MA, Palmblad J, Gyllenhammar HActivation of nitric oxide release and oxidative metabolism by leukotrienes B4, C4, and D4 in human polymorphonuclear leukocytes. Blood93199913991405
376. Becher G, Winsel K, Beck E, Neubauer G, Stresemann E. Breath condensate as a method of noninvasive assessment of inflammation mediators from the lower airways. Pneumologie 1997; 51(Suppl 2):456–459.
377. Hanazawa T, Kharitonov SA, Barnes PJIncreased nitrotyrosine in exhaled breath condensate of patients with asthma. Am J Respir Crit Care Med162200012731276
378. Dworski R, Sheller JRUrinary mediators and asthma. Clin Exp Allergy28199813091312
379. O'Sullivan S, Roquet A, Dahlén B, Dahlén SE, Kumlin MUrinary excretion of inflammatory mediators during allergen-induced early and late phase asthmatic reactions. Clin Exp Allergy28199813321339
380. Macfarlane AJ, Dworski R, Sheller JR, Pavord ID, Barry KA, Barnes NCSputum cysteinyl leukotrienes increase 24 hours after allergen inhalation in atopic asthmatics. Am J Respir Crit Care Med161200015531558
381. Hanazawa T, Kharitonov SA, Oldfield W, Kay AB, Barnes PJNitrotyrosine and cystenyl leukotrienes in breath condensates are increased after withdrawal of steroid treatment in patients with asthma [abstract]. Am J Respir Crit Care Med1612000A919
382. Morrow JD, Roberts LJThe isoprostanes: unique bioactive products of lipid peroxidation. Prog Lipid Res361997121
383. Montuschi P, Ciabattoni G, Paredi P, Pantelidis P, Du Bois RM, Kharitonov SA, Barnes PJ8-isoprostane as a biomarker of oxidative stress in interstitial lung diseases. Am J Respir Crit Care Med158199815241527
384. Roberts LJ, Morrow JDMeasurement of F(2)-isoprostanes as an index of oxidative stress in vivo. Free Radic Biol Med282000505513
385. Mori TA, Dunstan DW, Burke V, Croft KD, Rivera JH, Beilin LJ, Puddey IBEffect of dietary fish and exercise training on urinary F2-isoprostane excretion in non-insulin-dependent diabetic patients. Metabolism48199914021408
386. Carpenter CT, Price PV, Christman BWExhaled breath condensate isoprostanes are elevated in patients with acute lung injury or ARDS. Chest114199816531659
387. Marangon K, Devaraj S, Tirosh O, Packer L, Jialal IComparison of the effect of alpha-lipoic acid and alpha-tocopherol supplementation on measures of oxidative stress. Free Radic Biol Med27199911141121
388. Landino LM, Crews BC, Timmons MD, Morrow JD, Marnett LJPeroxynitrite, the coupling product of nitric oxide and superoxide, activates prostaglandin biosynthesis. Proc Natl Acad Sci USA9319961506915074
389. Wood LG, Fitzgerald DA, Gibson PG, Cooper DM, Garg MLLipid peroxidation as determined by plasma isoprostanes is related to disease severity in mild asthma. Lipids352000967974
390. Dworski R, Murray JJ, Jacksonroberts L, Oates JA, Morrow JD, Fisher L, Sheller JRAllergen-induced synthesis of F(2)-isoprostanes in atopic asthmatics. Evidence for oxidant stress. Am J Respir Crit Care Med160199919471951
391. Pratico D, Basili S, Vieri M, Cordova C, Violi F, Fitzgerald GAChronic obstructive pulmonary disease is associated with an increase in urinary levels of isoprostane F2alpha-III, an index of oxidant stress. Am J Respir Crit Care Med158199817091714
392. Montuschi P, Collins JV, Ciabattoni G, Lazzeri N, Corradi M, Kharitonov SA, Barnes PJExhaled 8-isoprostane as an in vivo biomarker of lung oxidative stress in patients with COPD and healthy smokers. Am J Respir Crit Care Med162200011751177
393. Hull J, Vervaart P, Grimwood K, Phelan PPulmonary oxidative stress response in young children with cystic fibrosis. Thorax521997557560
394. Collins CE, Quaggiotto P, Wood L, O'Loughlin EV, Henry RL, Garg MLElevated plasma levels of F2 alpha isoprostane in cystic fibrosis. Lipids341999551556
395. Jack CI, Jackson MJ, Johnston ID, Hind CRSerum indicators of free radical activity in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med153199619181923
396. Lenz AG, Costabel U, Maier KLOxidized BAL fluid proteins in patients with interstitial lung diseases. Eur Respir J91996307312
397. Schaberg T, Rau M, Stephan H, Lode HIncreased number of alveolar macrophages expressing surface molecules of the CD11/CD18 family in sarcoidosis and idiopathic pulmonary fibrosis is related to the production of superoxide anions by these cells. Am Rev Respir Dis147199315071513
398. Esterbauer H. Estimation of peroxidative damage. A critical review. Pathol Biol (Paris) 1996;44:25–28.
399. Clements NCJ, Habib MPThe early pattern of conjugated dienes in liver and lung after endotoxin exposure. Am J Respir Crit Care Med1511995780784
400. Ignatova GL, Volchegorskii IA, Volkova EG, Kazachkov EL, Kolesnikov OLLipid peroxidation processes in chronic bronchitis. Ter Arkh7019983637
401. Khyshiktuev BS, Khyshiktueva NA, Ivanov VN. Methods of measuring lipid peroxidation products in exhaled air condensate and their clinical significance. Klin Lab Diagn 1996;13–15.
402. Komar SI, Korobeinikova EN, Evdokimova EV. Lipids in the exhaled air condensate of pneumonia patients. Klin Lab Diagn 1996; 24–27.
403. Gichka SG, Briuzgina TS, Reva SN. The gas chromatographic analysis of the fatty acids in the expired air in ischemic heart disease. Klin Lab Diagn 1998;5–6.
404. Khyshiktyev BS, Khyshiktueva NA, Ivanov VN, Darenskaia SD, Novikov SVDiagnostic value of investigating exhaled air condensate in lung cancer. Vopr Onkol401994161164
405. Khyshiktueva NA, Khyshiktuev BS. Prenatal diagnosis of fetal hypoxia based on lipid peroxidation values in exhaled air condensate. Klin Lab Diagn 1998;21–22.
406. Takahashi H, Kuroki Y, Tanaka H, Saito T, Kurokawa K, Chiba H, Sagawa A, Nagae H, Abe SSerum levels of surfactant proteins A and D are useful biomarkers for interstitial lung disease in patients with progressive systemic sclerosis. Am J Respir Crit Care Med1622000258263
407. Goncharova VA, Mamedov DT, Dotsenko EK. Biologically active substance levels in exhaled air from patients with pre-asthma and bronchial asthma. Sov Med 1989;22–24.
408. Goncharova VA, Borisenko LV, Dotsenko EK, Pokhaznikova MAKallikrein-kinin indices and biological composition of exhaled condensate in acute bronchitis patients with varying disease course. Klin Med7419964648
409. Dzhangozina DM, Kulkybaev GA, Salimbaeva BMParameters of oxidative metabolism, neuro-humoral and hormonal regulation in the condensed exhaled air in early stages of anthracosilicosis. Med Tr Prom Ekol819991316
410. Stamler JSS-nitrosothiols and the bioregulatory actions of nitrogen oxides through reactions with thiol groups. Curr Topics Microbiol Immunol19619951936
411. van Der VA, Eiserich JP, Shigenaga MK, Cross CE. Reactive nitrogen species and tyrosine nitration in the respiratory tract. Epiphenomena or a pathobiologic mechanism of disease? Am J Respir Crit Care Med 1999;160:1–9.
412. Vukomanovic DV, Hussain A, Zoutman DE, Marks GS, Brien JF, Nakatsu KAnalysis of nanomolar S-nitrosothiol concentrations in physiological media. J Pharmacol Toxicol Method391998235240
413. Hou Y, Wang J, Arias F, Echegoyen L, Wang PGElectrochemical studies of S-nitrosothiols. Bioorg Med Chem Lett8199830653070
414. Wink DA, Kim S, Coffin D, Cook JC, Vodovotz Y, Chistodoulou D, Jourd'heuil D, Grisham MBDetection of S-nitrosothiols by fluorometric and colorimetric methods. Methods Enzymol3011999201211
415. Kostka P, Park JKFluorometric detection of S-nitrosothiols. Methods Enzymol3011999227235
416. Stamler JS, Loscalzo JCapillary zone electrophoretic detection of biological thiols and their S-nitrosated derivates. Anal Chem641992779785
417. Hunt J, Byrns RE, Ignarro LJ, Gaston BCondensed expirate nitrite as a home marker for acute asthma. Lancet346199512351236
418. Gabazza EC, Taguchi O, Tamaki S, Murashima S, Kobayashi H, Yasui H, Kobayashi T, Hataji O, Adachi Y. Role of nitric oxide in airway remodelling. Clin Sci (Colch) 2000;98:291–294.
419. Gaston B, Sears S, Woods J, Hunt J, Ponaman M, McMahon T, Stamler JSBronchodilator S-nitrosothiol deficiency in asthmatic respiratory failure. Lancet351199813171319
420. Chambers DC, Tunnicliffe WS, Ayres JGAcute inhalation of cigarette smoke increases lower respiratory tract nitric oxide concentrations. Thorax531998677679
421. Corradi M, Kharitonov SA, Donnelly LE, Montuschi P, Pesci A, Barnes PJElevated levels of nitrosothiols in breath condensate of healthy smokers [abstract]. Am J Respir Crit Care Med1612000A857
422. Ichinose M, Sugiura H, Yamagata S, Koarai A, Shirato KIncrease in reactive nitrogen species production in chronic obstructive pulmonary disease airways. Am J Respir Crit Care Med1622000701706
423. Linnane SJ, Keatings VM, Costello CM, Moynihan JB, O'Connor CM, Fitzgerakd MX, McLoughlin PTotal sputum nitrate plus nitrite is raised during acute pulmonary infection in cystic fibrosis. Am J Respir Crit Care Med1581998207212
424. Balint B, Donnelly LE, Hanazawa T, Kharitonov SA, Barnes PJNitric oxide metabolites in exhaled breath condensate and exhaled monoxides in cystic fibrosis [abstract]. Am J Respir Crit Care Med1612000A288
425. Jones KL, Hegab AH, Hillman BC, Simpson KL, Jinkins PA, Grisham MB, Owens MW, Sato E, Robbins RAElevation of nitrotyrosine and nitrate concentrations in cystic fibrosis sputum. Pediatr Pulmonol3020007985
426. Grasemann H, Gaston B, Fang K, Paul K, Ratjen FDecreased levels of nitrosothiols in the lower airways of patients with cystic fibrosis and normal pulmonary function. J Pediatr1351999770772
427. Corradi M, Montuschi P, Donnelly LE, Hodson ME, Kharitonov SA, Barnes PJNitrosothiols and nitrite in exhaled breath condensate of patients with cystic fibrosis [abstract]. Am J Respir Crit Care Med1591999A682
428. van Dalen CJ, Winterbourn CC, Senthilmohan R, Kettle AJNitrite as a substrate and inhibitor of myeloperoxidase: implications for nitration and hypochlorous acid production at sites of inflammation. J Biol Chem27520001163811644
429. Norwood DM, Wainman T, Lioy PJ, Waldman JMBreath ammonia depletion and its relevance to acidic aerosol exposure studies. Arch Environ Health471992309313
430. Arese M, Strasly M, Ruva C, Costamagna C, Ghigo D, MacAllister R, Verzetti G, Tetta C, Bosia A, Bussolino FRegulation of nitric oxide synthesis in uraemia. Nephrol Dial Transplant10199513861397
431. Vysotskii VGComparative characteristics of poly- and monomeric protein nutrition in relation to space flight. Kosm Biol Aviakosm Med919752328
432. Spanel P, Davies S, Smith DQuantification of ammonia in human breath by the selected ion flow tube analytical method using H30+ and 02+ precursor ions. Rapid Commun Mass Spectrom121998763766
433. Ballal SG, Ali BA, Albar AA, Ahmed HO, al-Hasan AYBronchial asthma in two chemical fertilizer producing factories in eastern Saudi Arabia. Int J Tuberc Lung Dis21998330335
434. Volozhin AI, Panin MG, Gnativ TV, Sel'tsovskaia GD, Sidel'nikova GM, Perova LA. The effect of hyperbaric oxygenation on the urea content of the saliva in acute and chronic soft-tissue inflammation in the maxillofacial area. Patol Fiziol Eksp Ter 1998;20–22.
435. Cox GM, Mukherjee J, Cole GT, Casadevall A, Perfect JRUrease as a virulence factor in experimental cryptococcosis. Infect Immun682000443448
436. Kharitonov SA, Barnes PJExhaled ammonia in asthma, cystic fibrosis and upper respiratory tract infection [abstract]. Am J Respir Crit Care Med1612000A307
437. Emel'ianov AV, Petrova MA, Lavrova OV, Guleva LI, Dolgodvorov AF, Fedoseev GBDisorders in mineral metabolism at different stages of the development of bronchial asthma. Ter Arkh6719954547
438. Zervas E, Loukides S, Papatheodorou G, Psathakis K, Tsindiris K, Panagou P, Kalogeropoulos NMagnesium levels in plasma and erythrocytes before and after histamine challenge. Eur Respir J162000621625
439. Bellocq A, Suberville S, Philippe C, Bertrand F, Perez J, Fouqueray B, Cherqui G, Baud LLow environmental pH is responsible for the induction of nitric-oxide synthase in macrophages: evidence for involvement of nuclear factor-kappa B activation. J Biol Chem273199850865092
440. Garey KW, Neuhauser MM, Rafice AL, Robbins RA, Danziger LH, Rubinstein IProtein, nitrite/nitrate, and cytokine concentration in exhaled breath condensate of young smokers [abstract]. Am J Respir Crit Care Med1612000A175
441. Gilbert IA, Fouke JM, McFadden ERJHeat and water flux in the intrathoracic airways and exercise-induced asthma. J Appl Physiol63198716811691
442. Agarkov FT, Agarkova SVThe temperature of exhaled air and the conditioning function of the respiratory apparatus in healthy miners and those with pneumoconiosis. Gig Tr Prof Zabol1419703134
443. Agarkov FTConditioning potentials of the respiratory tract. Fiziol Cheloveka101984981987
444. Brieva JL, Danta I, Wanner AEffect of an inhaled glucocorticosteroid on airway mucosal blood flow in mild asthma. Am J Respir Crit Care Med1612000293296
445. Solway J, Pichurko BM, Ingenito EP, McFadden ERJ, Fanta CH, Ingram RHJ, Drazen JMBreathing pattern affects airway wall temperature during cold air hyperpnea in humans. Am Rev Respir Dis1321985853857
446. Paredi P, Balint B, Barnes PJ, Kharitonov SASlower rise in exhaled breath temperature in cystic fibrosis: a novel marker of airway inflammation? Eur Respir J162000512S
447. Paredi P, Ward S, Cramer D, Barnes PJFaster rise in exhaled breath temperature in asthma: a novel marker of airway inflammation? Eur Respir J16200040S
448. Paredi P, Kharitonov SA, Willson K, Barnes PJSingle breath measurement of exhaled breath temperature. Eur Respir J16200040S
449. Nelson N, Lagesson V, Nosratabadi AR, Ludvigsson J, Tagesson CExhaled isoprene and acetone in newborn infants and in children with diabetes mellitus. Pediatr Res441998363367
450. Jones AW, Lagesson V, Tagesson CDetermination of isoprene in human breath by thermal desorption gas chromatography with ultraviolet detection. J Chromatogr B Biomed Sci Appl672199516
451. Skrupskii VA. Gas chromatographic analysis of ethanol and acetone in the air exhaled by patients. Klin Lab Diagn 1995;35–38.
452. Ebeler SE, Clifford AJ, Shibamoto TQuantitative analysis by gas chromatography of volatile carbonyl compounds in expired air from mice and human. J Chromatogr B Biomed Sci Appl7021997211215
453. Murtz P, Menzel L, Bloch W, Hess A, Michel O, Urban WLMR spectroscopy: a new sensitive method for on-line recording of nitric oxide in breath. J Appl Physiol86199910751080
454. Tanahashi T, Kodama T, Yamaoka Y, Sawai N, Tatsumi Y, Kashima K, Higashi Y, Sasaki YAnalysis of the 13C-urea breath test for detection of Helicobacter pylori infection based on the kinetics of delta-13CO2 using laser spectroscopy. J Gastroenterol Hepatol131998732737
455. Kaul A, Bhasin DK, Pathak CM, Ray P, Vaiphei K, Sharma BC, Singh KNormal limits of 14C-urea breath test. Trop Gastroenterol191998110113
456. Spanel P, Smith DSelected ion flow tube: a technique for quantitative trace gas analysis of air and breath. Med Biol Eng Comput341996409419
457. Groves WA, Zellers ETPrototype instrument employing a microsensor array for the analysis of organic vapors in exhaled breath. Am Ind Hyg Assoc J57199611031108
458. Runer T, Cervin A, Lindberg S, Uddman RNitric oxide is a regulator of mucociliary activity in the upper respiratory tract. Otolaryngol Head Neck Surg1191998278287
459. Pendergast DR, Krasney JA, DeRoberts DEffects of immersion in cool water on lung-exhaled nitric oxide at rest and during exercise. Respir Physiol11519997381
460. Franklin P, Dingle P, Stick SRaised exhaled nitric oxide in healthy children is associated with domestic formaldehyde levels. Am J Respir Crit Care Med161200017571759
461. Paredi P, Kharitonov SA, Hanazawa T, Barnes PJLocal vasodilator response to mobile phones. Eur Respir J16200040S
462. Binding N, Muller W, Czeschinski PA, Witting UNO chemiluminescence in exhaled air: interference of compounds from endogenous or exogenous sources. Eur Respir J162000499503
463. Tsuchiya M, Tokai H, Takehara Y, Haraguchi Y, Asada A, Utsumi K, Inoue MInterrelation between oxygen tension and nitric oxide in the respiratory system. Am J Respir Crit Care Med162200012571261
464. Guzel NA, Sayan H, Erbas DEffects of moderate altitude on exhaled nitric oxide, erythrocytes lipid peroxidation and superoxide dismutase levels. Jpn J Physiol502000187190
465. Jarvis MJ, Russell MA, Saloojee YExpired air carbon monoxide: a simple breath test of tobacco smoke intake. Br Med J2811980484485
466. Hewat VN, Foster EV, O'Brien GD, Town GIAmbient and exhaled carbon monoxide levels in a high traffic density area in Christchurch. N Z Med J1111998343344
467. Nightingale JA, Maggs R, Cullinan P, Donnelly LE, Rogers DF, Kinnersley R, Fan CK, Barnes PJ, Ashmore M, Newman-Taylor AAirway inflammation after controlled exposure to diesel exhaust particulates. Am J Respir Crit Care Med1622000161166
468. Togores B, Bosch M, Agusti AGThe measurement of exhaled carbon monoxide is influenced by airflow obstruction. Eur Respir J152000177180
469. Stevenson DK, Vreman HJCarbon monoxide and bilirubin production in neonates. Pediatrics1001997252254
470. Delivoria-Papadopoulos M, Coburn RF, Forster RECyclic variation of rate of carbon monoxide production in normal women. J Appl Physiol3619744951
471. Fischer AF, Nakamura H, Uetani Y, Vreman HJ, Stevenson DKComparison of bilirubin production in Japanese and Caucasian infants. J Pediatr Gastroenterol Nutr719882729
Correspondence and requests for reprints should be addressed to Professor P. J. Barnes, Department of Thoracic Medicine, National Heart & Lung Institute, Imperial College, Dovehouse Street, London SW3 6LY, UK. Email:

Related

No related items
American Journal of Respiratory and Critical Care Medicine
163
7

Click to see any corrections or updates and to confirm this is the authentic version of record