+ All Categories
Home > Documents > Review Article - Hindawi Publishing CorporationA2 (PLA2) but can be further metabolized by...

Review Article - Hindawi Publishing CorporationA2 (PLA2) but can be further metabolized by...

Date post: 25-Feb-2021
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
8
The Scientific World Journal Volume 2012, Article ID 217518, 7 pages doi:10.1100/2012/217518 The cientificWorldJOURNAL Review Article Exhaled Breath Condensate: A Promising Source for Biomarkers of Lung Disease Yan Liang, 1 Samantha M. Yeligar, 1, 2 and Lou Ann S. Brown 1 1 Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University and Emory+Children’s Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA 30322, USA 2 Department of Medicine, Atlanta Veterans’ Aairs and Emory University Medical Centers, Decatur, GA 30033, USA Correspondence should be addressed to Lou Ann S. Brown, [email protected] Received 20 October 2012; Accepted 25 November 2012 Academic Editors: B. Balbi, T. A. Mori, and S. A. Papiris Copyright © 2012 Yan Liang et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Exhaled breath condensate (EBC) has been increasingly studied as a noninvasive research method for sampling the alveolar and airway space and is recognized as a promising source of biomarkers of lung diseases. Substances measured in EBC include oxidative stress and inflammatory mediators, such as arachidonic acid derivatives, reactive oxygen/nitrogen species, reduced and oxidized glutathione, and inflammatory cytokines. Although EBC has great potential as a source of biomarkers in many lung diseases, the low concentrations of compounds within the EBC present challenges in sample collection and analysis. Although EBC is viewed as a noninvasive method for sampling airway lining fluid (ALF), validation is necessary to confirm that EBC truly represents the ALF. Likewise, a dilution factor for the EBC is needed in order to compare across subjects and determine changes in the ALF. The aims of this paper are to address the characteristics of EBC; strategies to standardize EBC sample collection and review available analytical techniques for EBC analysis. 1. General Characteristics of EBC Exhaled breath condensate (EBC) is collected from exhaled breath, usually through a refrigerated device [14]. During exhalation, volatile molecules and water evaporation directly diuse as gases from the lining fluid covering airspaces (e.g., alveoli), airways (e.g., bronchi), and the mouth. These gases are then collected into the expiratory air flow. Laser particle counting revealed that micron- and submicron- sized droplet particles are formed in the exhaled breath. Such particles serve as the only evidence of nonvolatile components in the EBC [4]. Yet, the nature and source of exhaled particles/droplets in the EBC matrix are not fully understood. Droplet formation within the lungs during exhalation is largely in the airways where turbulence is encountered (Figure 1). In addition, energy to overcome surface tension during inspiration may also apply to the airway and alveoli, potentially creating exhalable particles. However, the major source for nonvolatile components in the EBC is believed to be the airway lining fluid (ALF) [5, 6]. The main components of EBC include condensed water vapor, volatile molecules (such as nitric oxide, carbon monoxide, and hydrocarbons), and nonvolatile molecules (such as urea, GSH, leukotrienes, prostanoids, and cytokines) [1, 3]. 2. Aspects of EBC Sample Collection EBC sampling has great advantages over bronchoalveolar lavage (BAL) because it is noninvasive. However, compo- nents of the ALF are highly diluted in the EBC and are mixed with compounds from the mucus layer of the airway. Reproducibility is a major concern in EBC sample collection [1]. During EBC collection, tidal breathing is recommended [7]. With tidal breathing, the volume of air that is inhaled or exhaled is included in a single breath. This normal resting breathing pattern generates a reproducible volume of EBC and sampling of the ALF. However, EBC volume is not the only variable but dierent mediators in EBC can contribute to greater variability than volume. The causes of this variability have yet to be fully investigated but include the varied dilution eects for dierent nonvolatiles and
Transcript
Page 1: Review Article - Hindawi Publishing CorporationA2 (PLA2) but can be further metabolized by cyclooxy-genases (COX), 5-lipoxygenases (5-LO) and cytochrome P450 (CYP) [19–22]. A detailed

The Scientific World JournalVolume 2012, Article ID 217518, 7 pagesdoi:10.1100/2012/217518

The cientificWorldJOURNAL

Review Article

Exhaled Breath Condensate: A Promising Source forBiomarkers of Lung Disease

Yan Liang,1 Samantha M. Yeligar,1, 2 and Lou Ann S. Brown1

1 Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University and Emory+Children’s Healthcare of AtlantaCenter for Developmental Lung Biology, Atlanta, GA 30322, USA

2 Department of Medicine, Atlanta Veterans’ Affairs and Emory University Medical Centers, Decatur, GA 30033, USA

Correspondence should be addressed to Lou Ann S. Brown, [email protected]

Received 20 October 2012; Accepted 25 November 2012

Academic Editors: B. Balbi, T. A. Mori, and S. A. Papiris

Copyright © 2012 Yan Liang et al. This is an open access article distributed under the Creative Commons Attribution License,which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Exhaled breath condensate (EBC) has been increasingly studied as a noninvasive research method for sampling the alveolar andairway space and is recognized as a promising source of biomarkers of lung diseases. Substances measured in EBC include oxidativestress and inflammatory mediators, such as arachidonic acid derivatives, reactive oxygen/nitrogen species, reduced and oxidizedglutathione, and inflammatory cytokines. Although EBC has great potential as a source of biomarkers in many lung diseases, thelow concentrations of compounds within the EBC present challenges in sample collection and analysis. Although EBC is viewedas a noninvasive method for sampling airway lining fluid (ALF), validation is necessary to confirm that EBC truly represents theALF. Likewise, a dilution factor for the EBC is needed in order to compare across subjects and determine changes in the ALF. Theaims of this paper are to address the characteristics of EBC; strategies to standardize EBC sample collection and review availableanalytical techniques for EBC analysis.

1. General Characteristics of EBC

Exhaled breath condensate (EBC) is collected from exhaledbreath, usually through a refrigerated device [1–4]. Duringexhalation, volatile molecules and water evaporation directlydiffuse as gases from the lining fluid covering airspaces(e.g., alveoli), airways (e.g., bronchi), and the mouth. Thesegases are then collected into the expiratory air flow. Laserparticle counting revealed that micron- and submicron-sized droplet particles are formed in the exhaled breath.Such particles serve as the only evidence of nonvolatilecomponents in the EBC [4]. Yet, the nature and sourceof exhaled particles/droplets in the EBC matrix are notfully understood. Droplet formation within the lungs duringexhalation is largely in the airways where turbulence isencountered (Figure 1). In addition, energy to overcomesurface tension during inspiration may also apply to theairway and alveoli, potentially creating exhalable particles.However, the major source for nonvolatile components in theEBC is believed to be the airway lining fluid (ALF) [5, 6]. Themain components of EBC include condensed water vapor,

volatile molecules (such as nitric oxide, carbon monoxide,and hydrocarbons), and nonvolatile molecules (such as urea,GSH, leukotrienes, prostanoids, and cytokines) [1, 3].

2. Aspects of EBC Sample Collection

EBC sampling has great advantages over bronchoalveolarlavage (BAL) because it is noninvasive. However, compo-nents of the ALF are highly diluted in the EBC and aremixed with compounds from the mucus layer of the airway.Reproducibility is a major concern in EBC sample collection[1]. During EBC collection, tidal breathing is recommended[7]. With tidal breathing, the volume of air that is inhaledor exhaled is included in a single breath. This normalresting breathing pattern generates a reproducible volumeof EBC and sampling of the ALF. However, EBC volume isnot the only variable but different mediators in EBC cancontribute to greater variability than volume. The causes ofthis variability have yet to be fully investigated but includethe varied dilution effects for different nonvolatiles and

Page 2: Review Article - Hindawi Publishing CorporationA2 (PLA2) but can be further metabolized by cyclooxy-genases (COX), 5-lipoxygenases (5-LO) and cytochrome P450 (CYP) [19–22]. A detailed

2 The Scientific World Journal

technique sensitivity needed for these nonvolatiles. AlthoughEBC dilution is relevant for nonvolatile constituents of EBC,this is not relevant for volatile components. Currently theestimated dilution factor is determined using nonvolatilemolecules which have similar concentrations in ALF andplasma. Dilution factors that have been used include urea,cations, total protein concentration, or the conductivity oflyophilized EBC [2, 6, 8]. The dilution (D) of an interestednonvolatile biomarker can be calculated as shown below,using urea as the standard indicator:

[nonvolatile]ALF = D × [nonvolatile]EBC,

where D = [Urea]ALF

[Urea]EBC= [Urea]plasma

[Urea]EBC.

(1)

However, it needs to point out that the dilution of thesenonvolatile biomarkers by water vapor can vary dramatically,and to date, there is no gold standard for assessing thedilution of ALF biomarkers in the EBC [1, 6, 9, 10].

Clearly, the interest in EBC relies on its ease in samplecollection. However, sufficient and reproducible techniquesare needed in EBC sampling. Recommendations for EBCsample collections are well documented and accepted amongEBC researchers. In brief, precondensation conditions, suchas ambient air and environment temperature, should berecorded; the condenser’s design, material, surface area andcooling temperature should be well adjusted; the subject’sconditions, such as medications, tobacco smoking, food anddrinks, exercise, et al., can have significant effects on EBCcollection and should be recoded and adjusted. Detailedrecommendation conditions for EBC collection have beendescribed in [1, 4, 11, 12].

3. Measurements of Mediators in EBC

As described above, EBC components are classified intotwo categories: volatile and nonvolatile. Guidelines andrecommendations to measure EBC substances are usefulin standardizing measurements and further develop newtechniques. The following issues are generally consideredwhen measuring substances in EBC [1, 4, 11, 13]. (1)The cooling temperature range should be specified. Coldercondensation temperature is usually better for unstable EBCcomponents; however, colder temperature may reduce theamount of volatiles because they are more readily absorbedinto the liquid phase. (2) Sublimation of the volatiles intothe airspace should be considered when using frozen storagefor EBC samples. (3) Each substance of interest shouldbe studied in detail to control for the potential effects ofduration of EBC collection, storage conditions, and assaymethods. (4) To validate the results with more than one assayusing different methodologies is usually necessary for EBCanalysis. (5) Because many of the nonvolatile componentsfound in EBC are identified by assays at their lower limitsof accuracy, the use of lyophilization, dehydration, or freezedrying of the EBC coupled with resuspension in smallvolumes of highly pure water can improve the detectionsensitivity. (6) EBC is highly dilute, assay controls must

be performed appropriately with similar EBC componentconcentration scales. The following sections describe themost studied mediators in EBC.

3.1. EBC pH. Respiratory symptoms such as cough, wheeze,dyspnea, and apnea are induced when acids are introducedinto the airways or when the endogenous airway pHhomeostasis is altered by diverse pulmonary diseases. Theregulation of airway pH is involved in innate host defensesbut also contributes to the pathophysiology of obstructivelung disease [14]. Therefore, it is important and beneficialto precisely and conveniently measure the airway pH inthe diagnosis of many pulmonary conditions. Measurementof EBC pH or airway acidification is very challenging andcomplicated by poor reproducibility [15, 16]. The pH ofraw EBC samples is unstable and is profoundly affected bycarbon dioxide, the major volatile component of EBC. Onestrategy is to deaerate EBC with an inert (carbon dioxidefree) gas such as argon or nitrogen to remove carbon dioxide.However, even after 20 min of deaeration, EBC samples maystill contain an unpredictable amount of carbon dioxide,which may bias pH readings. To improve the reproducibilityof pH readings and standardize the carbon dioxide effecton EBC pH, a carbon dioxide gas standardization methodwas developed [17, 18]. In this method, carbon dioxide isbubbled into an EBC sample for short intervals (1 s each)which cause a rapid but stepwise increase of the carbondioxide partial pressure in the EBC sample. After each bub-bling period, EBC pH and carbon dioxide partial pressureare measured simultaneously using a blood gas analyzer. Acorrelation plot between the EBC pH and carbon dioxidepartial pressure is then generated. This correlation allowsthe calculation of pH at a carbon dioxide partial pressureof 5.33 kPa, the physiological alveolar carbon dioxide partialpressure. Although more reliable and convenient methodsneed to be developed for EBC pH measurement, this methodcurrently provides the most reproducible EBC pH values.

3.2. Arachidonic Acid Derivatives in the EBC. Arachidonicacid (AA) is a polyunsaturated omega-6 fatty acid presentin the phospholipids of cell membranes. Arachidonic acidis released by the activation of the enzyme phospholipaseA2 (PLA2) but can be further metabolized by cyclooxy-genases (COX), 5-lipoxygenases (5-LO) and cytochromeP450 (CYP) [19–22]. A detailed scheme is presented inFigure 2 for arachidonic acid metabolism, where intracellularinteractions control arachidonic acid conversion and activity.Cyclooxygenases generate prostanoids which can be furthersubdivided into three main groups: the prostaglandins(PGs), prostacyclin (PGI2), and thromboxanes (TXs), eachof which is involved in some aspect of the inflammatoryresponse. Arachidonate 5-lipoxygenase converts AA to yieldleukotrienes (LTs). CYP epoxygenases (CYP-EO) convertarachidonic acid to epoxyeicosatrienoic acids (EETs). CYPhydroxylases (CYP-HO) metabolize arachidonic acid tohydroxyeicosatetraenoic acids (HETEs). Airway epithelialcells are sensitive to arachidonate metabolites and haveabundant arachidonic acid and novel cyclooxygenases and

Page 3: Review Article - Hindawi Publishing CorporationA2 (PLA2) but can be further metabolized by cyclooxy-genases (COX), 5-lipoxygenases (5-LO) and cytochrome P450 (CYP) [19–22]. A detailed

The Scientific World Journal 3

Expiratory flow

Airspace

Airway ling fluid

Epithelial ling fluid

Volatile biomarkers

Droplets Airway

H2O vapor

Figure 1: Nonvolatile and volatile components in EBC. Water vapor is rapidly diffused from the lining fluid on the surface of the airway(bronchi) and airspace (alveolar) into the expiratory flow. Droplets (nonvolatile biomarker) formation in the lung is largely from the liningfluid of the airway where turbulence is encountered. Respiratory gases (volatile biomarkers) are from both airspace and airway, and moresoluble vapors are typically greater in the airway [5, 67].

Phospholipids

Arachidonic acid

LTs

TXs PGs

PLA2

5-LOCOX

5-HPETECyclo-

endoperoxides

PGI2

CYP-EO

EETs

CYP-HO

HETEs

ROS 8-IP

Figure 2: Metabolism of arachidonic acid. Arachidonic acidis released from phospholipids by the action of phospholipaseA2 (PLA2). Arachidonic acid is metabolized by cyclooxygenases(COXs), lipoxygenases (LOXs), and cytochrome P450 (CYP). COXsmetabolize arachidonic acid to prostaglandins (PGs), prostacyclin(PGI2), and thromboxanes (TXs). Leukotrienes (LTs) are the finalarachidonic acid metabolites in the 5-lipoxygenase- (5-LO-) medi-ated pathway. CYP epoxygenases (CYP-EO) metabolize arachidonicacid to epoxyeicosatrienoic acid (EETs), and CYP hydroxylases(CYP-HO) metabolize arachidonic acid to hydroxyeicosatetraenoicacids (HETEs). 8-Isoprostane (8-IP) can be generated in vivo by thefree radical-catalyzed peroxidation of arachidonic acid.

lipoxygenases at increased levels relative to other cell types[23]. However, arachidonate metabolites can be synthesizedby and have potent biologic effects on other airway cellssuch as leukocytes, smooth muscle, nerves, mucus glands,and platelets. 8-Isoprostane (8-IP), a prostaglandin (PG)-F2-like compound, belongs to the F2 isoprostane class that isproduced in vivo by the free radical-catalyzed peroxidationof arachidonic acid [24]. Because of the transcellular featureof arachidonic acid metabolism and function, airway liningfluid is the critical medium for these actions. Significantamounts of arachidonic acid and its derivatives are present in

ALF. 8-Isoprostane, LTs, and prostanoids have been detectedin EBC and used as biomarkers for oxidative stress and res-piratory infection. Methods used to detect arachidonic acidderivatives in the EBC include gas chromatography/massspectrometry (GC/MS), liquid chromatography/mass spec-trometry (LC/MS), radioimmunoassay (RIA), and enzymeimmunoassay (EIA).

Most studies measuring 8-IP used commercial EIA kitswith a detection limit of 1 pg/mL [25]. GC/MS is a moresensitive method for 8-IP detection and has been used tovalidate EIA results [26]. 8-IP levels in the EBC of healthysubjects were reported in the range of 0–40 pg/mL. Increasedconcentrations of 8-IP in the EBC as a marker of oxidativestress has been demonstrated in multiple lung diseases,such as asthma [27, 28], COPD [29, 30], interstitial lungdisease [31], and cystic fibrosis [32, 33]. LTs can also bemeasured in the EBC by EIA with a detection limit of4 pg/mL. Other methods such as LC/MS/MS, GC/MS, andhigh-performance liquid chromatography (HPLC) are alsoused in LT detection [34]. LTs in EBC samples from healthysubjects range from 0 to 25 pg/mL. Elevated LTs were foundto be correlated with parameters of inflammation in thelungs [35]. Similarly, prostanoids can be measured by EIA,RIA, and chromatographic techniques and are present in therange of 0–200 pg/mL in EBC [26, 36].

3.3. Oxygen and Nitrogen Reactive Species and Redox-RelevantMolecules in EBC. Investigations of reactive oxygen species(ROS) and reactive nitrogen species (RNS) are among theinterests of EBC biomarkers in many lung disease studies.Multiple RNS formation starts with nitric oxide (NO). NOis a volatile component of the EBC [3, 13, 37, 38] andis synthesized from the amino acid L-arginine by nitricoxide synthase (NOS) (Figure 3). Different cell types withinthe respiratory tract have been identified to contain NOS,including airway and alveolar epithelial cells, macrophages,neutrophils, eosinophils, mast cells, and vascular endothelialand smooth muscle cells. Superoxide anion (•O2

−) is aROS that reacts quickly with NO, to form highly reactiveperoxynitrite (ONOO−). ONOO− can cause the nitrosation

Page 4: Review Article - Hindawi Publishing CorporationA2 (PLA2) but can be further metabolized by cyclooxy-genases (COX), 5-lipoxygenases (5-LO) and cytochrome P450 (CYP) [19–22]. A detailed

4 The Scientific World Journal

L-arginine

NO

NOS

RS-NOThiols

NO3−

NO2−ONOO−

3-NT

•O2−

O2

NOX

H2O2Fe2+

Tyr

H2O

Thiols•OH

H+

SOD

Thiols

Figure 3: Reactive oxygen and nitrogen species and redox relevantmolecules in EBC. Exhaled nitric oxide (NO) is derived fromL-arginine by enzyme nitric oxide synthase (NOS). NO cancombine with superoxide (•O2

−) to form peroxynitrite (ONOO−).ONOO− induces nitrosation of tyrosine (Tyr) residues and forms3-nitrotyrosine (3-NT). NO can also react with thiols to formS-nitroso thiols (RS-NO). The end-products of NO are nitrite(NO2

−) or nitrate (NO3−). •O2

− is one of major reactive oxygenspecies generated from NADPH oxidase (NOX) or mitochondrialelectron transfer chain. •O2

− is converted to hydrogen peroxide(H2O2) by superoxide dismutases (SOD). H2O2 can be convertedto the highly reactive hydroxyl radical (•OH), which is catalyzedby Fe2+ (Fenton reaction). H2O2 can be removed by thiol-specificantioxidant enzymes to form water.

of either tyrosine or tyrosine residues in proteins to form3-nitrotyrosine (3-NT). Nitrotyrosine can be measured byenzyme immune assays or HPLC and MS [39–41]. NO canalso react with thiols, such as cysteine, glutathione, or proteinthiol residues to produce S-nitrosothiols (RS-NO) whichcan be measured by the colorimetric assay [42]. The end-products of NO metabolism are nitrite (NO2

−) and nitrate(NO3

−). In EBC, nitrite and nitrate can be measured bycolorimetric, fluorometric, and chemiluminescent assays, orby ion, gas, and liquid chromatography [43, 44].

Hydrogen peroxide (H2O2) is another volatile moleculein EBC [3, 13]. In several cell types, H2O2 can be producedby superoxide dismutase (SOD) through conversion of thesuperoxide anion (•O2

−). H2O2 can be released from bothinflammatory and structural cells including neutrophils,eosinophils, macrophages, and epithelial cells. Since H2O2

is unstable in the EBC, samples should be freshly collectedor rapidly frozen after collection. Common methods usedto measure H2O2 include spectrophotometric, fluorometric,or chemiluminescent assays and indicate a concentrationof ∼200 nM in different pulmonary pathologies [45, 46].Reactive oxygen species can degrade polyunsaturated lipidsand form malondialdehyde (MDA), another biomarker ofoxidative stress [47, 48]. The MDA present in the EBC canbe measured by HPLC in the 10 nM concentration range[49, 50].

Increasing reactive oxygen and nitrogen species or theirderivatives in the EBC are used as indicators of oxidativestress or inflammation in the respiratory track. Compared

with healthy nonsmokers, concentrations of H2O2, MDA,RS-NO, 3-NT, NO2

−, and NO3− are increased in the EBC of

patients with asthma, COPD, idiopathic pulmonary fibrosis,and cystic fibrosis [1, 3, 13, 47]. In addition to ROS/RNS, theALF also contains significant antioxidant compounds suchas cysteine (Cys) and glutathione (GSH). Although the GSHconcentration in the bronchoalveolar lining fluid is in themagnitude of μM, the GSH concentration in the EBC is inthe magnitude of nM resulting in a 1000 dilution of GSH inthe EBC pool when compared to the bronchoalveolar lavagefluid [51–53]. When subjects with or without an alcoholuse disorder were compared, both the lavage fluid and theEBC demonstrated ∼80% decrease in GSH and oxidationof the thiol/disulfide redox potential by ∼40 mV [54]. Thissuggests that changes in the EBC can be representative ofphysiological changes in the ALF.

3.4. EBC Proteins. Playing central roles in both the immu-nity and inflammation aspects of the host defense system,cytokines can be classified by their ability to promote orinhibit inflammatory response: proinflammatory cytokines(IL-1β, IL-2, IL-6, IL-8, IL-12, IL-17, IFN-γ, and TNF-α), anti-inflammatory cytokines (IL-4, IL-5, IL-10, IL-13,and TGF-β), and chemokines (IL-8, MCP-1, and MIP-1ß).Cytokines can also be grouped based on the type of T-lymphocytes with which they are associated. T helper (Th)lymphocytes stem from T CD4+ lymphocytes precursors(Th0), and depending on the cytokine environment, helper Tcells can differentiate into three major different phenotypes:Th1, Th2, and Th17. The Th1 cytokine profile includesIFN-γ, TNF-α, IL-1, IL-2, and IL-12. The Th2 cytokinesare IL-4, IL-5, IL-6, IL-10, and IL-13. Th17 cytokines (IL-17, IL-21, IL-22, TNF-α, and TGF-β) include “regulatory”cytokines involved in the immune tolerance process. System-atic cytokine profiling is useful in diagnosis and therapeutictreatment for airway diseases. Identification of cytokines inthe EBC using ELISA assays has been reported. In the EBC,the cytokines IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-17, IFN-γ, TGF-β, and TNF-α have been reported to bein the ∼50 pg/mL range [55–57]. An EBC dilution factorof 10−3 is generally accepted relative to ALF [9, 58] givingan estimated ALF cytokine level in the order of 50 ng/mL.However, cytokine detection in EBC is often at the lowerlimits of detection for the assay, and these values are furthercomplicated by the absence of a gold standard for dilution ofthe EBC or the bronchoalveolar lavage.

Due to the complications from detection bias and cor-rection for dilution, the measurement of multiple substancesconcurrently and determination of their ratios would reducethe detection bias and avoid artifacts due to correctionfor dilution. For cytokine analysis, a shift in the Th1/Th2ratio usually accompanies with varied immune responsein pathological pulmonary conditions. Examples of suchapproach have been reported in determining the IFN-γ(Th1)/IL-4(Th2) ratio [56, 59]. Systematic approaches,such as proteomic analysis of EBC, have been previouslyused and may provide a more detailed overall view aboutcytokine profile in the EBC. However, EBC is challenging for

Page 5: Review Article - Hindawi Publishing CorporationA2 (PLA2) but can be further metabolized by cyclooxy-genases (COX), 5-lipoxygenases (5-LO) and cytochrome P450 (CYP) [19–22]. A detailed

The Scientific World Journal 5

proteomics studies because of low protein concentrations.Proteome analysis of low-abundance proteins depends onthe complexity of the protein mixture, the power of theresolution, and the sensitivity of the separation and iden-tification methods. Although proteomic analysis has beenused with EBC, the majority of the proteins detected werekeratins, a family of fibrous structural proteins present inthe outer layer of human skin [60–63]. Although keratincontent in EBC has shown significant differences betweendisease subjects and healthy control groups [61, 62, 64], theEBC cytokine profile is still a useful tool for monitoringlung inflammation. To detect low-abundance EBC cytokinespresent in the pg/mL range, advanced techniques such asimmunoaffinity depletion and selective target enrichmentare required for proteomic analysis [65, 66].

4. Conclusion

EBC is an exciting new approach for investigating lung dis-eases because it is noninvasive and contains many potentialbiomarkers. However, the key limitation for the EBC as adiagnostic tool is the low concentration range of differentEBC biomarkers. Currently, efforts to address methodolog-ical issues include standardization of sample collection andvalidation of analytical techniques. To establish the repro-ductively of EBC measurements, more sensitive assays andnew molecular detection techniques are necessary. Meta-bolomics and proteomics can provide systemic profile forEBC biomarkers and may prove to be useful in screening anddiagnosing lung diseases. In addition, systematic techniquesthat can concurrently measure multiple EBC substances maylimit detection bias and provide patterns of biomarkers thatare sensitive to disease and disease treatments.

References

[1] I. Horvath, J. Hunt, P. J. Barnes et al., “Exhaled breath con-densate: methodological recommendations and unresolvedquestions,” European Respiratory Journal, vol. 26, no. 3, pp.523–548, 2005.

[2] J. Hunt, “Exhaled breath condensate—an overview,” Immuno-logy And Allergy Clinics of North America, vol. 27, no. 4, pp.587–596, 2007.

[3] P. Montuschi, “Analysis of exhaled breath condensate in respi-ratory medicine: methodological aspects and potential clinicalapplications,” Therapeutic Advances in Respiratory Disease, vol.1, no. 1, pp. 5–23, 2007.

[4] M. D. Davis, A. Montpetit, and J. Hunt, “Exhaled breathcondensate: an overview,” Immunology and Allergy Clinics ofNorth America, vol. 32, no. 3, pp. 363–375, 2012.

[5] R. M. Effros, R. Casaburi, J. Porszasz, E. M. Morales, and V.Rehan, “Exhaled breath condensates: analyzing the expiratoryplume,” American Journal of Respiratory and Critical CareMedicine, vol. 185, no. 8, pp. 803–804, 2012.

[6] R. M. Effros, J. Biller, B. Foss et al., “A simple methodfor estimating respiratory solute dilution in exhaled breathcondensates,” American Journal of Respiratory and Critical CareMedicine, vol. 168, no. 12, pp. 1500–1505, 2003.

[7] E. M. H. Huttmann, T. Greulich, A. Hattesohl et al., “Com-parison of two devices and two breathing patterns for exhaled

breath condensate sampling,” PLoS ONE, vol. 6, no. 11, ArticleID e27467, 2011.

[8] R. M. Effros, B. Peterson, R. Casaburi et al., “Epithelial liningfluid solute concentrations in chronic obstructive lung diseasepatients and normal subjects,” Journal of Applied Physiology,vol. 99, no. 4, pp. 1286–1292, 2005.

[9] R. M. Effros, K. W. Hoagland, M. Bosbous et al., “Dilution ofrespiratory solutes in exhaled condensates,” American Journalof Respiratory and Critical Care Medicine, vol. 165, no. 5, pp.663–669, 2002.

[10] A. Zacharasiewicz, N. Wilson, and A. Bush, “Dilution ofrespiratory solutes in exhaled condensates,” American Journalof Respiratory and Critical Care Medicine, vol. 167, no. 5, p.802, 2003.

[11] N. M. Grob, M. Aytekin, and R. A. Dweik, “Biomarkers inexhaled breath condensate: a review of collection, processingand analysis,” Journal of Breath Research, vol. 2, no. 3, ArticleID 037004, 18 pages, 2008.

[12] P. Rosias, “Methodological aspects of exhaled breath conden-sate collection and analysis,” Journal of Breath Research, vol. 6,no. 2, Article ID 027102, 13 pages, 2012.

[13] P. Montuschi and P. J. Barnes, “Analysis of exhaled breathcondensate for monitoring airway inflammation,” Trends inPharmacological Sciences, vol. 23, no. 5, pp. 232–237, 2002.

[14] J. F. Hunt and B. Gaston, “Airway acidification and gastroe-sophageal reflux,” Current Allergy and Asthma Reports, vol. 8,no. 1, pp. 79–84, 2008.

[15] J. F. Hunt, K. Fang, R. Malik et al., “Endogenous airway acid-ification: implications for asthma pathophysiology,” AmericanJournal of Respiratory and Critical Care Medicine, vol. 161, no.3 I, pp. 694–699, 2000.

[16] R. M. Effros, “Endogenous airway acidification: implicationsfor asthma pathology,” American Journal of Respiratory andCritical Care Medicine, vol. 163, no. 1, pp. 293–294, 2001.

[17] J. L. Lin, M. H. Bonnichsen, and P. S. Thomas, “Standard-ization of exhaled breath condensate: effects of different de-aeration protocols on pH and H2O2 concentrations,” Journalof Breath Research, vol. 5, no. 1, Article ID 011001, 2011.

[18] B. Antus, I. Barta, T. Kullmann et al., “Assessment ofexhaled breath condensate pH in exacerbations of asthma andchronic obstructive pulmonary disease,” American Journal ofRespiratory and Critical Care Medicine, vol. 182, no. 12, pp.1492–1497, 2010.

[19] Y. Tesfaigzi, M. Kluger, and W. Kozak, “Clinical and cellulareffects of cytochrome P-450 modulators,” Respiration Physiol-ogy, vol. 128, no. 1, pp. 79–87, 2001.

[20] S. P. Farooque and T. H. Lee, “Aspirin-sensitive respiratorydisease,” Annual Review of Physiology, vol. 71, pp. 465–487,2009.

[21] A. A. Spector, “Arachidonic acid cytochrome P450 epoxyge-nase pathway,” Journal of Lipid Research, vol. 50, pp. S52–S56,2009.

[22] B. N. M. Zordoky and A. O. S. El-Kadi, “Effect of cytochromeP450 polymorphism on arachidonic acid metabolism andtheir impact on cardiovascular diseases,” Pharmacology andTherapeutics, vol. 125, no. 3, pp. 446–463, 2010.

[23] M. J. Holtzman, “Arachidonic acid metabolism in airwayepithelial cells,” Annual Review of Physiology, vol. 54, pp. 303–329, 1992.

[24] J. D. Morrow, K. E. Hill, R. F. Burk, T. M. Nammour, K.F. Badr, and L. J. Roberts, “A series of prostaglandin F2-like compounds are produced in vivo in humans by a non-cyclooxygenase, free radical-catalyzed mechanism,” Proceed-ings of the National Academy of Sciences of the United Statesof America, vol. 87, no. 23, pp. 9383–9387, 1990.

Page 6: Review Article - Hindawi Publishing CorporationA2 (PLA2) but can be further metabolized by cyclooxy-genases (COX), 5-lipoxygenases (5-LO) and cytochrome P450 (CYP) [19–22]. A detailed

6 The Scientific World Journal

[25] E. Baraldi, S. Carraro, R. Alinovi et al., “Cysteinyl leukotrienesand 8-isoprostane in exhaled breath condensate of childrenwith asthma exacerbations,” Thorax, vol. 58, no. 6, pp. 505–509, 2003.

[26] P. Montuschi, E. Ragazzoni, S. Valente et al., “Validation of8-isoprostane and prostaglandin E2 measurements in exhaledbreath condensate,” Inflammation Research, vol. 52, no. 12, pp.502–507, 2003.

[27] E. Baraldi, L. Ghiro, V. Piovan et al., “Increased exhaled 8-isoprostane in childhood asthma,” Chest, vol. 124, no. 1, pp.25–31, 2003.

[28] A. Antczak, P. Montuschi, S. Kharitonov, P. Gorski, and P.J. Barnes, “Increased exhaled cysteinyl-leukotrienes and 8-isoprostane in aspirin-induced asthma,” American Journal ofRespiratory and Critical Care Medicine, vol. 166, no. 3, pp. 301–306, 2002.

[29] D. Makris, E. Paraskakis, P. Korakas et al., “Exhaled breathcondensate 8-isoprostane, clinical parameters, radiologicalindices and airway inflammation in COPD,” Respiration, vol.75, no. 2, pp. 138–144, 2008.

[30] V. L. Kinnula, H. Ilumets, M. Myllarniemi, A. Sovijarvi, andP. Rytila, “8-Isoprostane as a marker of oxidative stress innonsymptomatic cigarette smokers and COPD,” EuropeanRespiratory Journal, vol. 29, no. 1, pp. 51–55, 2007.

[31] P. Montuschi, G. Ciabattoni, P. Pared et al., “8-Isoprostane asa biomarker of oxidative stress in interstitial lung diseases,”American Journal of Respiratory and Critical Care Medicine,vol. 158, no. 5, pp. 1524–1527, 1998.

[32] P. Latzin, J. Beck-Ripp, D. Hartl, A. Bartenstein, J. Noss, and M.Griese, “8-isoprostane in nasally exhaled breath condensate indifferent pediatric lung diseases,” European Journal of MedicalResearch, vol. 12, no. 1, pp. 21–25, 2007.

[33] P. Montuschi, S. A. Kharitonov, G. Ciabattoni et al., “Exhaled8-isoprostane as a new non-invasive biomarker of oxidativestress in cystic fibrosis,” Thorax, vol. 55, no. 3, pp. 205–209,2000.

[34] K. Syslova, P. Kacer, B. Vilhanova et al., “Determination ofcysteinyl leukotrienes in exhaled breath condensate: methodcombining immunoseparation with LC—ESI-MS/MS,” Jour-nal of Chromatography B, vol. 879, no. 23, pp. 2220–2228,2011.

[35] T. Tsuburai, H. Mita, N. Tsurikisawa et al., “The relationshipbetween cysteinyl leukotriene in exhaled breath condensateand the severity of asthma in adult asthmatics in Japan,”Japanese Journal of Allergology, vol. 57, no. 2, pp. 121–129,2008.

[36] P. Montuschi, C. Mondino, R. Pistelli, A. Trove, P. Koch,and G. Ciabattoni, “Leukotrienes and prostanoids in exhaledbreath condensate in childhood asthma,” Journal of Allergy andClinical Immunology, vol. 113, no. 2, article S187, 2004.

[37] J. C. Ojoo, S. A. Mulrennan, J. A. Kastelik, A. H. Morice, andA. E. Redington, “Exhaled breath condensate pH and exhalednitric oxide in allergic asthma and in cystic fibrosis,” Thorax,vol. 60, no. 1, pp. 22–26, 2005.

[38] L. E. Donnelly, “Exhaled breath condensate: nitric oxide-related compounds,” European Respiratory Monograph, vol. 49,pp. 207–216, 2010.

[39] B. Balint, S. A. Kharitonov, T. Hanazawa et al., “Increasednitrotyrosine in exhaled breath condensate in cystic fibrosis,”European Respiratory Journal, vol. 17, no. 6, pp. 1201–1207,2001.

[40] M. Larstad, A. S. Soderling, K. Caidahl, and A. C. Olin, “Selec-tive quantification of free 3-nitrotyrosine in exhaled breath

condensate in asthma using gas chromatography/tandem massspectrometry,” Nitric Oxide, vol. 13, no. 2, pp. 134–144, 2005.

[41] A. Conventz, A. Musiol, C. Brodowsky et al., “Simultaneousdetermination of 3-nitrotyrosine, tyrosine, hydroxyprolineand proline in exhaled breath condensate by hydrophilic inter-action liquid chromatography/electrospray ionization tandemmass spectrometry,” Journal of Chromatography B, vol. 860, no.1, pp. 78–85, 2007.

[42] M. Corradi, P. Montuschi, L. E. Donnelly, A. Pesci, S. A.Kharitonov, and P. J. Barnes, “Increased nitrosothiols inexhaled breath condensate in inflammatory airway diseases,”American Journal of Respiratory and Critical Care Medicine,vol. 163, no. 4, pp. 854–858, 2001.

[43] J. Chladkova, I. Krcmova, J. Chladek, P. Cap, S. Micuda, and Y.Hanzalkova, “Validation of nitrite and nitrate measurementsin exhaled breath condensate,” Respiration, vol. 73, no. 2, pp.173–179, 2006.

[44] H. Marteus, D. C. Tornberg, E. Weitzberg, U. Schedin, andK. Alving, “Origin of nitrite and nitrate in nasal and exhaledbreath condensate and relation to nitric oxide formation,”Thorax, vol. 60, no. 3, pp. 219–225, 2005.

[45] W. M. Brooks, H. Lash, A. J. Kettle, and M. J. Epton,“Optimising hydrogen peroxide measurement in exhaledbreath condensate,” Redox Report, vol. 11, no. 2, pp. 78–84,2006.

[46] S. Loukides, P. Bakakos, and K. Kostikas, “Exhaled breathcondensate: hydrogen peroxide,” European Respiratory SocietyMonograph, vol. 49, pp. 162–172, 2010.

[47] M. L. Bartoli, F. Novelli, F. Costa et al., “Malondialdehyde inexhaled breath condensate as a marker of oxidative stress indifferent pulmonary diseases,” Mediators of Inflammation, vol.2011, Article ID 891752, 7 pages, 2011.

[48] I. Romieu, A. Barraza-Villarreal, C. Escamilla-Nunez et al.,“Exhaled breath malondialdehyde as a marker of effect ofexposure to air pollution in children with asthma,” Journal ofAllergy and Clinical Immunology, vol. 121, no. 4, pp. 903.e6–909.e6, 2008.

[49] M. Celik, A. Tuncer, O. U. Soyer, C. Sackesen, H. T. Besler,and O. Kalayci, “Oxidative stress in the airways of childrenwith asthma and allergic rhinitis,” Pediatric Allergy andImmunology, vol. 23, no. 6, pp. 556–561, 2012.

[50] M. Larstad, G. Ljungkvist, A. C. Olin, and K. Toren,“Determination of malondialdehyde in breath condensate byhigh-performance liquid chromatography with fluorescencedetection,” Journal of Chromatography B, vol. 766, no. 1, pp.107–114, 2002.

[51] M. Y. Yeh, E. L. Burnham, M. Moss, and L. A. S. Brown,“Chronic alcoholism alters systemic and pulmonary glu-tathione redox status,” American Journal of Respiratory andCritical Care Medicine, vol. 176, no. 3, pp. 270–276, 2007.

[52] M. Y. Yeh, E. L. Burnham, M. Moss, and L. A. S. Brown, “Non-invasive evaluation of pulmonary glutathione in the exhaledbreath condensate of otherwise healthy alcoholics,” RespiratoryMedicine, vol. 102, no. 2, pp. 248–255, 2008.

[53] M. Moss, D. M. Guidot, M. Wong-Lambertina, T. Ten Hoor,R. L. Perez, and L. A. S. Brown, “The effects of chronic alcoholabuse on pulmonary glutathione homeostasis,” AmericanJournal of Respiratory and Critical Care Medicine, vol. 161, no.2, pp. 414–419, 2000.

[54] O. Gajic, R. Rana, J. L. Winters et al., “Transfusion-relatedacute lung injury in the critically Ill: prospective nested case-control study,” American Journal of Respiratory and CriticalCare Medicine, vol. 176, no. 9, pp. 886–891, 2007.

Page 7: Review Article - Hindawi Publishing CorporationA2 (PLA2) but can be further metabolized by cyclooxy-genases (COX), 5-lipoxygenases (5-LO) and cytochrome P450 (CYP) [19–22]. A detailed

The Scientific World Journal 7

[55] K. Matsunaga, S. Yanagisawa, T. Ichikawa et al., “Airwaycytokine expression measured by means of protein arrayin exhaled breath condensate: correlation with physiologicproperties in asthmatic patients,” Journal of Allergy andClinical Immunology, vol. 118, no. 1, pp. 84–90, 2006.

[56] C. M. H. H. T. Robroeks, Q. Jobsis, J. G. M. C. Damoiseauxet al., “Cytokines in exhaled breath condensate of childrenwith asthma and cystic fibrosis,” Annals of Allergy, Asthma andImmunology, vol. 96, no. 2, pp. 349–355, 2006.

[57] U. Sack, R. Scheibe, M. Wotzel et al., “Multiplex analysis ofcytokines in exhaled breath condensate,” Cytometry A, vol. 69,no. 3, pp. 169–172, 2006.

[58] C. Gessner, R. Scheibe, M. Wotzel et al., “Exhaled breath con-densate cytokine patterns in chronic obstructive pulmonarydisease,” Respiratory Medicine, vol. 99, no. 10, pp. 1229–1240,2005.

[59] S. K. Shahid, S. A. Kharitonov, N. M. Wilson, A. Bush, and P. J.Barnes, “Increased interleukin-4 and decreased interferon-γ inexhaled breath condensate of children with asthma,” AmericanJournal of Respiratory and Critical Care Medicine, vol. 165, no.9, pp. 1290–1293, 2002.

[60] D. H. Conrad, J. Goyette, and P. S. Thomas, “Proteomicsas a method for early detection of cancer: a review ofproteomics, exhaled breath condensate, and lung cancerscreening,” Journal of General Internal Medicine, vol. 23, no.1, pp. 78–84, 2008.

[61] Z. Cheng, C. R. Lewis, P. S. Thomas, and M. J. Raftery, “Com-parative proteomics analysis of exhaled breath condensate inlung cancer patients,” Journal of Cancer Therapy, vol. 2, no. 1,pp. 1–8, 2011.

[62] K. Bloemen, J. Hooyberghs, K. Desager, E. Witters, and G.Schoeters, “Non-invasive bioarker sampling and analysis ofthe exhaled breath proteome,” Proteomics, vol. 3, no. 4, pp.498–504, 2009.

[63] E. Dompeling and Q. Jobsis, “Proteomics of exhaled breathcondensate: a realistic approach in children with asthma?”Clinical and Experimental Allergy, vol. 41, no. 3, pp. 299–301,2011.

[64] E. Gianazza, L. Allegra, E. Bucchioni et al., “Increased keratincontent detected by proteomic analysis of exhaled breathcondensate from healthy persons who smoke,” AmericanJournal of Medicine, vol. 117, no. 1, pp. 51–54, 2004.

[65] T. Shi, D. Su, T. Liu et al., “Advancing the sensitivity of selectedreaction monitoring-based targeted quantitative proteomics,”Proteomics, vol. 12, no. 8, pp. 1074–1092, 2012.

[66] J. Brand, T. Haslberger, W. Zolg, G. Pestlin, and S. Palme,“Depletion efficiency and recovery of trace markers from amultiparameter immunodepletion column,” Proteomics, vol.6, no. 11, pp. 3236–3242, 2006.

[67] J. C. Anderson, W. J. E. Lamm, and M. P. Hlastala, “Measuringairway exchange of endogenous acetone using a single-exhalation breathing maneuver,” Journal of Applied Physiology,vol. 100, no. 3, pp. 880–889, 2006.

Page 8: Review Article - Hindawi Publishing CorporationA2 (PLA2) but can be further metabolized by cyclooxy-genases (COX), 5-lipoxygenases (5-LO) and cytochrome P450 (CYP) [19–22]. A detailed

Submit your manuscripts athttp://www.hindawi.com

Stem CellsInternational

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Disease Markers

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation http://www.hindawi.com Volume 2014

Immunology ResearchHindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Parkinson’s Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttp://www.hindawi.com


Recommended