Nitrosative events in atopic asthma pathogenesis

The correlation between high exhaled nitric oxide levels and eosinophilic-mediated airway inflammation in patients with atopic asthma has been well documented. This generates prerequisites that a regulatory feedback mechanism exists between them. Therefore, the paper briefly describes evidence imple...

Full description

Saved in:
Bibliographic Details
Published in:Вiopolymers and Cell
Date:2015
Main Authors: Parilova, O.O., Volodina, T.T., Shandrenko, S.G.
Format: Article
Language:English
Published: Інститут молекулярної біології і генетики НАН України 2015
Subjects:
Online Access:https://nasplib.isofts.kiev.ua/handle/123456789/152698
Tags: Add Tag
No Tags, Be the first to tag this record!
Journal Title:Digital Library of Periodicals of National Academy of Sciences of Ukraine
Cite this:Nitrosative events in atopic asthma pathogenesis / O.O. Parilova, T.T. Volodina, S.G. Shandrenko // Вiopolymers and Cell. — 2015. — Т. 31, № 6. — С. 405-416. — Бібліогр.: 98 назв. — англ.

Institution

Digital Library of Periodicals of National Academy of Sciences of Ukraine
id nasplib_isofts_kiev_ua-123456789-152698
record_format dspace
spelling Parilova, O.O.
Volodina, T.T.
Shandrenko, S.G.
2019-06-12T16:13:43Z
2019-06-12T16:13:43Z
2015
Nitrosative events in atopic asthma pathogenesis / O.O. Parilova, T.T. Volodina, S.G. Shandrenko // Вiopolymers and Cell. — 2015. — Т. 31, № 6. — С. 405-416. — Бібліогр.: 98 назв. — англ.
0233-7657
1993-6842
DOI: http://dx.doi.org/10.7124/bc.0008FD
https://nasplib.isofts.kiev.ua/handle/123456789/152698
616.248
The correlation between high exhaled nitric oxide levels and eosinophilic-mediated airway inflammation in patients with atopic asthma has been well documented. This generates prerequisites that a regulatory feedback mechanism exists between them. Therefore, the paper briefly describes evidence implementing biosynthesis, enzyme structural features, expression regulation of its isoforms and effects of nitric oxide, which have helped elucidate molecular mechanisms by which nitric oxide selectively promotes asthma exacerbation. In previous study we have demonstrated that airway infiltrate of immune cells contributes to NO synthesis in the respiratory tract during allergic inflammation under guinea pig model of acute asthma with multiple challenges. On the basis of these findings the authors posits that nitric oxide represents an additional signal of the induction of Th2 subset response and be considerably involved in the complex network of immune regulation distinctive for atopic asthma phenotype.
Кореляцію між високим рівнем оксиду азоту, що видихається та еозинофіл-опосередкованим запаленням дихальних шляхів у пацієнтів з атопічною бронхіальною астмою добре доведено. Це створює передумови існування регуляторного механізму зворотнього зв’язку між ними. Тому стаття стисло наводить свідчення стосовно біосинтезу, структурних особливостей ензиму, регуляції експресії його ізоформ та дії оксиду азоту, що допомагає з’ясувати молекулярні механізми завдяки яким оксид азоту сприяє загостренню перебігу захворювання. В попередній експериментальній роботі ми продемонстрували, що інфільтрат імунних клітин, які персистують в дихальних шляхах, робить внесок у синтез оксиду азоту в респіраторному тракті під час пізньої алергічної реакції за моделі гострої бронхіальної астми мурчаків з множинними провокаціями алергену. На основі цих даних автори констатують, що оксид азоту є додатковим сигналом індукції відповіді Th2 ланки та залучений в складну мережу імунної регуляції, характерної для фенотипу атопічної астми.
Корреляция между высоким уровнем оксида азота в выдыхаемом воздухе и эозинофил-опосредованным воспалением дыхательных путей у пациентов с атопической бронхиальной астмой хорошо обоснована. Это порождает предпосылку, что существует механизм регулирования обратной связи между ними. Таким образом, статья кратко описывает данные касающиеся биосинтеза, структурных особенностей энзима, регулирования экспрессии его изоформ и воздействия оксида азота, которые помогают выяснить молекулярные механизмы, благодаря чему оксид азота селективно содействует обострению астмы. В предыдущей экспериментальной работе мы показали, что инфильтрат иммунных клеток в дыхательных путях дополняет синтез оксида азота в респираторном тракте во время аллергического воспаления на модели острой бронхиальной астмы морских свинок с множественными провокациями аллергена. На основании этих данных авторы констатируют, что оксид азота представляет собой дополнительный сигнал индукции ответа Th2 звена и значительно вовлечен в сложную сеть иммунной регуляции, отличительной для фенотипа атопической астмы.
en
Інститут молекулярної біології і генетики НАН України
Вiopolymers and Cell
Reviews
Nitrosative events in atopic asthma pathogenesis
Нітрозативні події в патогенезі атопічної астми
Нитрозативные события в патогенезе атопической астмы
Article
published earlier
institution Digital Library of Periodicals of National Academy of Sciences of Ukraine
collection DSpace DC
title Nitrosative events in atopic asthma pathogenesis
spellingShingle Nitrosative events in atopic asthma pathogenesis
Parilova, O.O.
Volodina, T.T.
Shandrenko, S.G.
Reviews
title_short Nitrosative events in atopic asthma pathogenesis
title_full Nitrosative events in atopic asthma pathogenesis
title_fullStr Nitrosative events in atopic asthma pathogenesis
title_full_unstemmed Nitrosative events in atopic asthma pathogenesis
title_sort nitrosative events in atopic asthma pathogenesis
author Parilova, O.O.
Volodina, T.T.
Shandrenko, S.G.
author_facet Parilova, O.O.
Volodina, T.T.
Shandrenko, S.G.
topic Reviews
topic_facet Reviews
publishDate 2015
language English
container_title Вiopolymers and Cell
publisher Інститут молекулярної біології і генетики НАН України
format Article
title_alt Нітрозативні події в патогенезі атопічної астми
Нитрозативные события в патогенезе атопической астмы
description The correlation between high exhaled nitric oxide levels and eosinophilic-mediated airway inflammation in patients with atopic asthma has been well documented. This generates prerequisites that a regulatory feedback mechanism exists between them. Therefore, the paper briefly describes evidence implementing biosynthesis, enzyme structural features, expression regulation of its isoforms and effects of nitric oxide, which have helped elucidate molecular mechanisms by which nitric oxide selectively promotes asthma exacerbation. In previous study we have demonstrated that airway infiltrate of immune cells contributes to NO synthesis in the respiratory tract during allergic inflammation under guinea pig model of acute asthma with multiple challenges. On the basis of these findings the authors posits that nitric oxide represents an additional signal of the induction of Th2 subset response and be considerably involved in the complex network of immune regulation distinctive for atopic asthma phenotype. Кореляцію між високим рівнем оксиду азоту, що видихається та еозинофіл-опосередкованим запаленням дихальних шляхів у пацієнтів з атопічною бронхіальною астмою добре доведено. Це створює передумови існування регуляторного механізму зворотнього зв’язку між ними. Тому стаття стисло наводить свідчення стосовно біосинтезу, структурних особливостей ензиму, регуляції експресії його ізоформ та дії оксиду азоту, що допомагає з’ясувати молекулярні механізми завдяки яким оксид азоту сприяє загостренню перебігу захворювання. В попередній експериментальній роботі ми продемонстрували, що інфільтрат імунних клітин, які персистують в дихальних шляхах, робить внесок у синтез оксиду азоту в респіраторному тракті під час пізньої алергічної реакції за моделі гострої бронхіальної астми мурчаків з множинними провокаціями алергену. На основі цих даних автори констатують, що оксид азоту є додатковим сигналом індукції відповіді Th2 ланки та залучений в складну мережу імунної регуляції, характерної для фенотипу атопічної астми. Корреляция между высоким уровнем оксида азота в выдыхаемом воздухе и эозинофил-опосредованным воспалением дыхательных путей у пациентов с атопической бронхиальной астмой хорошо обоснована. Это порождает предпосылку, что существует механизм регулирования обратной связи между ними. Таким образом, статья кратко описывает данные касающиеся биосинтеза, структурных особенностей энзима, регулирования экспрессии его изоформ и воздействия оксида азота, которые помогают выяснить молекулярные механизмы, благодаря чему оксид азота селективно содействует обострению астмы. В предыдущей экспериментальной работе мы показали, что инфильтрат иммунных клеток в дыхательных путях дополняет синтез оксида азота в респираторном тракте во время аллергического воспаления на модели острой бронхиальной астмы морских свинок с множественными провокациями аллергена. На основании этих данных авторы констатируют, что оксид азота представляет собой дополнительный сигнал индукции ответа Th2 звена и значительно вовлечен в сложную сеть иммунной регуляции, отличительной для фенотипа атопической астмы.
issn 0233-7657
url https://nasplib.isofts.kiev.ua/handle/123456789/152698
citation_txt Nitrosative events in atopic asthma pathogenesis / O.O. Parilova, T.T. Volodina, S.G. Shandrenko // Вiopolymers and Cell. — 2015. — Т. 31, № 6. — С. 405-416. — Бібліогр.: 98 назв. — англ.
work_keys_str_mv AT parilovaoo nitrosativeeventsinatopicasthmapathogenesis
AT volodinatt nitrosativeeventsinatopicasthmapathogenesis
AT shandrenkosg nitrosativeeventsinatopicasthmapathogenesis
AT parilovaoo nítrozativnípodíívpatogenezíatopíčnoíastmi
AT volodinatt nítrozativnípodíívpatogenezíatopíčnoíastmi
AT shandrenkosg nítrozativnípodíívpatogenezíatopíčnoíastmi
AT parilovaoo nitrozativnyesobytiâvpatogenezeatopičeskoiastmy
AT volodinatt nitrozativnyesobytiâvpatogenezeatopičeskoiastmy
AT shandrenkosg nitrozativnyesobytiâvpatogenezeatopičeskoiastmy
first_indexed 2025-11-24T20:41:41Z
last_indexed 2025-11-24T20:41:41Z
_version_ 1850495768606015488
fulltext 405 O. O. Parilova, T. T. Volodina, S. G. Shandrenko © 2015 O. O. Parilova et al.; Published by the Institute of Molecular Biology and Genetics, NAS of Ukraine on behalf of Biopolymers and Cell. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited UDC 616.248 Nitrosative events in atopic asthma pathogenesis O. O. Parilova, T. T. Volodina, S. G. Shandrenko Palladin Institute of Biochemistry, NAS of Ukraine 9, Leontovycha Str., Kyiv, Ukraine, 01030 elenparil@gmail.com The correlation between high exhaled nitric oxide levels and eosinophilic-mediated airway inflammation in patients with atopic asthma has been well documented. This generates prerequisites that a regulatory feedback mechanism exists between them. Therefore, the paper briefly describes evidence implementing biosynthesis, enzyme structural features, expression regulation of its isoforms and effects of nitric oxide, which have helped elucidate molecular mechanisms by which nitric oxide selectively promotes asthma exacerbation. In previous study we have demonstrated that airway infiltrate of immune cells contributes to NO synthesis in the respira- tory tract during allergic inflammation under guinea pig model of acute asthma with multiple challenges. On the basis of these findings the authors posits that nitric oxide represents an additional signal of the induction of Th2 subset response and be considerably involved in the complex network of immune regulation distinctive for atopic asthma phenotype. K e y w o r d s: nitric oxide, atopic asthma, nitric oxide synthases, allergic airway inflammation, eosinophilia. Introduction Asthma is a chronic inflammation of the airways that involves airflow obstruction and increased airway responsiveness to a variety of stimuli, which leads to the symptoms of recurrent coughing and dyspnea [1]. Albeit the understanding of disease pathogenesis has progressed enormously in the last several decades [1–3], asthma is still a major public health problem, affecting 300 million people worldwide and rapidly increasing in all age groups [4, 5]. Clinically, several distinct phenotypes are recognized, but the therapy and research have been basically focused on allergic (atopic) asthma [1–3]. Currently, the routine asthma treatment relies on suppressing the inflammation (glucocorticoids, leu- kotriene inhibitors as additional medication) as well as removal of bronchoconstriction (β agonists, anti- cholinergics) [6, 7]. The symptom-based guidance of treatment could predispose to an increased num- ber of exacerbations. Thus, asthmatics might benefit from the inflammatory marker-addressed therapy [1, 8]. Although many physiologic processes have been implicated in asthma pathogenesis, the range of evi- dence supports the key roles of endogenous nitric oxide (NO) and NO-derived reactive nitrogen spe- cies (RNS) in modulating either a normal airway function [8, 9] or inflammation in asthma [10, 11]. Therefore, NO has received a tremendous amount of attention in the medical community as a selective pulmonary disorder marker [8, 12, 13]. NO is an ubiquitous messenger that regulates var- ious biological functions — either at low concentra- tions as a signal molecule in many physiological processes, or at high concentrations as cytotoxic and cytostatic defensive mechanisms against tumors and pathogens [9, 11]. Also it is thought to be a marker of lung inflammation. NO is detected in exhaled breath [8, 12, 13]. Fractional exhaled NO (FeNO) levels were shown to correlate with sputum eosinophil Reviews ISSN 1993-6842 (on-line); ISSN 0233-7657 (print) Biopolymers and Cell. 2015. Vol. 31. N 6. P 405–416 doi: http://dx.doi.org/10.7124/bc.0008FD 406 O. O. Parilova, T. T. Volodina, S. G. Shandrenko count, airway hyperresponsiveness, bronchodilator response, serum IgE levels, allergen skin prick test- ing, asthma symptoms, and lung function. This led to the general belief that atopy is associated with higher levels of exhaled NO (eNO) [11]. From this background, the current article deals with the consideration of the source of NO formation and its triggers in asthma pathogenesis. We aim to discuss the cases, when NO becomes a marker of al- lergic inflammation. Simultaneously, we present our data that revealed the contribution of immune cells settling down the airways to the respiratory NO gen- eration. 1. Biosynthesis and enzyme structure basis The role of NO in the human body is under the scope of intensive research. NO is generated by oxidation of L-arginine; this reaction is typically catalyzed by the nitric oxide synthases (NOS) [9] in the presence of several co-factors: flavones (FAD, FMN), tetrahy- drobiopterin (BH4) and NADPH [14]. NOS are ste- reospecific and active as homodimers [14]. The en- zyme exists in three isoforms: neuronal NOS (nNOS/ NOS1), inducible NOS (iNOS/NOS2) and endothe- lial NOS (eNOS/NOS3) [15–18]. nNOS and eNOS are constitutively expressed iso- forms (cNOS) in cells as the preformed proteins that are activated in response to the cell-specific stimuli through the elevation of intracellular Ca2+ concentra- tions and the binding of calmodulin. iNOS form is generally associated with the immune system and produces NO for prolonged periods of time in a cal- cium-independent manner [9–11, 14–22]. iNOS is activated by bacterial lipopolysaccharides and pro- inflammatory cytokines such as inteleukin-1, TNF-α, IFN-γ [15]. However, IFN-γ is the most potent and prevailing inducer of iNOS in vivo [23]. As T-lymphocytes and NK-cells are the main source of IFN-γ production, the iNOS induction is mediated by them. The endothelial cell derived factor, endo- thelin-1 (ET-1), has been shown to promote iNOS expression [24, 25]. Ultimately, iNOS incorporates a calmodulin binding site to which the calmodulin is tightly bound independently of a calcium signal – this is thought to be responsible for the continuous activity of the enzyme [15–17]. Up to 1000 times higher levels of NO can be generated by iNOS com- pared to cNOS [11, 19]. The NOS enzyme functions as a dimer consisting of two identical monomers, which can be function- ally and structurally divided into two major domains: a C-terminal reductase domain and an N-terminal oxygenase (catalytic) domain. All NOS isoforms are members of the cytochrome P450 enzyme group owing to the presence of a haem complex [20]. Thus the mammalian NOS share similar domain architec- ture, with an N-terminal catalytic domain containing a haem active site, a nearby cofactor site for BH4 and a C-terminal reductase domain consisting of FMN, FAD and NADPH binding sites. We visual- ized catalytic domain of human NOS enzymes in three-dimensional form using PDB files via Cn3D viewer from MMDB (Fig. 1). The macromolecular 3D structures display the evidence, that eNOS and iNOS isoforms are very similar in the overall mo- lecular shape, relative orientation of cofactors and stereochemistry within the catalytic centre. As the catalytic sites of iNOS and eNOS are so similar it is not obvious from a structural starting point how the selective inhibitors may be designed [26]. 2. Localization of nitric oxide synthase iso- forms in respiratory tract and its biological role In the respiratory tract all isoforms have been identi- fied [20, 21]. cNOS-derived NO along with other NO adduct molecules are involved in maintaining normal metabolic functions, such as airway and pul- monary vascular tone, intracellular signaling, immu- nity (platelet aggregation, leukocyte adhesion) and neurotransmission [7, 22]. By contrast to this, iNOS- derived NO seems to play a pro-inflammatory pri- mary role in the cytokine dependent processes [19]. Albeit, agonists such as sheer stress, bradykinin, acetylcholine and histamine still may activate cNOS, resulting in the release of NO within seconds [20]. eNOS is constitutively expressed in both the en- dothelial cells of the bronchial and pulmonary circu- 407 Nitrosative events in atopic asthma pathogenesis lation and the alveolar endothelial cells, and airway epithelial cells throughout the respiratory tract [27]. The traces of NO from eNOS activity have been iso- lated in the airway epithelium and found to partici- pate in ciliary movement and mucus ejection [28, 29]. Although eNOS is present in airway epithelium, iNOS is considered to be a predominant form in it [19]. The recent evidence has demonstrated a link between certain eNOS gene polymorphisms and in- creased serum IgE levels in patients with allergic asthma [30]. Functionally, nNOS mediates neuronal bronchodilation in the skeletal muscles and choliner- gic airway nerves [27]. When allergic asthma was being launched, the increased arginase activity at- tenuates the nNOS-catalyzed synthesis of NO and induces neural bronchoconstriction [31]. In this way cNOS isoforms reveal an indirect involvement in asthma pathophysiology. In general, iNOS regulates approximately 200 genes that are relevant to inflammation, infection or apoptosis [21]. In the airways iNOS is expressed by epithelial and endothelial cells [23, 32], smooth mus- cle cells [18, 32, 33], lung fibroblasts [34]. In contrast to other cell types that do not express NOS2 unless induced by cytokines, the NOS2 mRNA expression and NOS2 protein were detected in epithelial cells of normal, noninflamed upper and lower airways and in pulmonary myocytes of large hilar veins by a variety of techniques, including Northern blotting, in situ hy- bridization, Western blotting, and immunohisto- chemistry [23, 35]. Additionally, iNOS-derived NO is produced by the genuine immune-system cells in- cluding monocytes and macrophages [16, 17, 36], eosinophils [37, 38], neutrophils [39, 40], dendritic cells [41]. The reports about the NOS expression in the mast cell population appear to be ambiguous. The data presented in numerous papers clearly indicate distinct differences in diversity and localization of NOS in a variety of mast cell populations expressed in human and rodents [42–44]. Notably, the iNOS ex- pression and NO generation are up-regulated by the antigen stimulation FcɛRI engagement in the differ- ent subsets of mast cells [44]. Additionally, the NO production prevents mitochondrial integrity collapse, thereby protecting mast cells against antigen-induced apoptosis [45]. Previously several reports have con- firmed that the rat mast cells produce iNOS mRNA and the protein upon treatment [46–48]. However subsequent studies have demonstrated that some types of human mast cell were positive for eNOS but showed variable expression of nNOS and no detect- able iNOS [42, 43]. Together these findings suggest the important differences between the rat and human mast cells in the NO production and NO-mediated regulation of mast cell function [42]. NK cells ex- press constitutive eNOS mRNA and protein, howev- er the levels of iNOS are not detectable. The endog- enous NO production is involved in protection of NK cells from activation of apoptosis induced by CD16 cross-linking, thereby maintaining NK activity [49]. This is the further evidence that, unlike NO produced by iNOS, cNOS-derived NO is critical for normal physiology. Farther, the biological effect of NO ultimately de- pends on its concentration and interaction with other bioreactive molecules and proteins [50]. NO can be converted to NO2, NO2–, NO3– and other reactive nitrogen intermediates (RNI), among which the most important are S-nitrosothiols (S-NO), peroxynitrite (ONOO–) and nitrosyl-metal complexes, which are directly implicated in the RNI-mediated free radical reactions (S-nitrosylation of cysteines, nitration of tyrosines, and nitrosylation of prosthetic groups, re- spectively) [51]. In summary, the excessive NO- adduct molecules namely ONOO–, nitrogen dioxide (NO2), dinitrogen trioxide (N2O3), and higher ox- ides of nitrogen can provoke detrimental post-trans- lational modifications [13, 50, 52]. 3. Nitric oxide formation as a marker of Th2-mediated infllammation There is rising evidence for an important immuno- regulatory role of NO in the development of the adaptive immune responses associated with autoim- mune and allergic diseases. Increased levels of ex- haled NO [53–56] along with an up-regulation of iNOS in asthmatic airways [57] confirm that this radical should be involved in asthma. It has been 408 O. O. Parilova, T. T. Volodina, S. G. Shandrenko demonstrated, that, in humans, a higher than normal NO concentration in exhaled breath is closely asso- ciated with the enlarged transcriptional activation of NOS2 gene [58]. Nevertheless, the previous studies, using murine and guinea pig models of asthma and iNOS inhibitor treatment or iNOS-deficient animals, have generated controversial results. In two different studies, iNOS deficiency [59] or iNOS inhibitor treatment [60] had no effect on the lung inflammation, whereas in other study the airway inflammation was inhibited in the iNOS deficient mice [61]. Such a discrepancy was attributed to significant differences in the immuniza- tion and challenge protocols [59]. Further investigations have shown that inhibited iNOS activity correlates with a decrease of nitroty- rosine and, more importantly, ameliorates FEV1 and airway responsiveness to histamine [14]. However, the different subgroups analysis of atopic individu- als now suggests that it is the inflammation in atopic individuals with clinical manifestations of airway disease, rather than atopy itself, that accounts for the increased production of NO by iNOS [62]. eNO was commonly regarded qua a marker of eosinophilic in- flammation [63–65]. In addition, the 2011 American Thoracic Society (ATS) guideline for the use of FeNO in clinical practice characterized FeNO as an indicator of eosinophilic airway inflammation [12]. However, the recent studies have indicated that FeNO is more representative of a Th2-driven local inflammation, specifically of the bronchial mucosa, rather than general eosinophilic inflammation, as measured by blood or induced sputum [66]. The dis- connect between FeNO and eosinophilic inflamma- tion has been highlighted by two separate studies with monoclonal antibodies (mAb) against IL-5 and IL-13, which indicate that treatment with mepoli- zumab, an anti-IL-5 mAb, significantly reduces blood and sputum eosinophils without affecting FeNO levels [67] whereas treatment with lebriki- zumab, an anti-IL-13 mAb, significantly reduces FeNO levels without reducing blood eosinophils [68]. Another research has shown that, in asthmatic children but not in controls, eNO levels and BAL fluid eosinophil percentages correlate well regard- less of the methods used to measure eNO [69]. Thus, eNO levels interconnect better with bronchial eo- sinophils than with sputum eosinophils. Based on the findings of the bronchoalveolar la- vage fluid (BALF) studies and the lung tissue histo- logical analysis, the mechanisms for late allergic re- sponsiveness (LAR) are considered to be causally related to the infiltration of eosinophils and other inflammatory cells into the bronchial mucosa [70]. It has been suggested that NO play an important role in LAR [10, 71]. In the airways of asthmatic patients [57] or in rodent lung after allergen challenge [61, 72], iNOS expression and/or enzymatic activity are increased. To give insight into the role of nitrosative stress established by immune cells infiltrating the airways we examined the NO formation in BAL cells under the guinea pig model of acute ovalbumin (OVA)- induced asthma with repeated challenges following 32 weeks after sensitization [73]. This model mimics those seen in humans exposed to an allergen a long period after acquired hypersensitivity. A suggested protocol reflects the allergen-driven pathway of asthma reproducing several characteristic features, such as airways infiltration by inflammatory cells, early allergic responsiveness (EAR) and LAR, AHR. Flow cytometry analysis using DAF-2DA dye showed, that multiple allergen challenge exposures of sensitized guinea pigs were associated with an ex- cessive level of the intracellular NO generation in BAL cells (Figure 2). Hence, OVA aerosol provoca- tions of guinea pigs with sensitization resulted in a substantial growth of BAL cells containing NO in comparison with sensitized group (33.50[27.80– 45.80] % versus 6.30[2.20–7.82] %; p=0.0001). Our results are in line with the data confirming that al- lergic inflammation is accompanied by NO. Moreover, there is apparent evidence that iNOS in- hibition, during the challenge period, markedly re- duced the development of the inflammatory process in the OVA-induced murine model after allergen challenge through down-regulation of chemokine expression [74]. 409 Nitrosative events in atopic asthma pathogenesis Taking into account that BAL explores large ar- eas of the alveolar compartment providing cellular constituents from the lower respiratory tract, we identified the alterations in cellular composition of BAL as indicated in FACS density graphs (Fig. 3). OVA provocations in sensitized animals were shown to cause a strong inflammation in lungs. This led to the redistribution of immune subpopula- Fig. 1. The structures of oxygenase do- main of human NOS isoforms. A – dimeric unit of eNOS haem domain with L-arginine bound (PBD ID:4D1O). B – tetrameric unit of nNOS haem domain with L-arginine bound (PBD ID: 4D1N). C – tetrameric unit of iNOS with Zn-bound and L-arginine complex (PBD ID: 1NSI). The enzyme structures show a ball and stick backbone in a trace shape, no side chains to alignment protein views, and sol- id objects – condensed lines and massive cylinders with arrows – to represent strands and helices. The arrows on helix cylinders point in the N-to-C direction. The extent of each monomer element in macromolecule is designated by color (blue, violet, green and brown). Haem, BH4 and arginine, glycerol are shown in a tube representation. The association into a dimer involves a large interface, which includes the binding site for BH4 and helps to structure the ac- tive-site pocket containing the haem and the L-arginine binding site Fig. 2. NO content in BAL cells 18–20 h after induction of allergic lung inflammation: morphological flow cytometric parameters of cell types concerning size (FS) and granularity (SS). Panel A: BAL cells derived from sensitized guinea pigs with allergen chal- lenges (II coumn) display augment of NO generation compared to sensitized animals (I column). Panel B: fractions of BAL cells incorporating NO from two compared groups (I and II columns) are also demonstrated in the individual dot plots. Green-colored cells include NO molecules (adopted from [73] and supplemented in this paper) 410 O. O. Parilova, T. T. Volodina, S. G. Shandrenko tions in the general pool of cells infiltrating respira- tory tract. In addition to quantitative assessment of NO for- mation, the allocation of intracellular NO content in BAL suspension was analyzed depending on the size and granularity (Fig. 4, Fig. 5) [73]. In view of these findings we infer that in asthma, due to increased iNOS protein expression and ac- tivity, the excessive NO generation is activated in lung tissue cells (mainly airway epithelial cells), but also in multi-cellular airway infiltrate recruited to trigger organ during allergic inflammation. High BAL levels of NO may reflect circulating nitrosa- tive stress in respiratory tract, when exacerbation in asthma is present. Indeed inhibition of iNOS-de- rived NO attenuates antigen-induced airway con- striction, inflammatory, and reduces collagen and elastic fiber deposition in a guinea pig model of al- lergic asthma [37]. 4. Immunoregulatory cross-talk of nitric oxide in asthma pathogenesis The network of molecules involved in the allergic inflammatory processes becomes more complex since NO derivatives have been demonstrated to play a role in these reactions. The allergic disorder pathogenesis is modulated by NO at the level of im- mune system [20, 32]. In turn, iNOS transcription is regulated by a number of pathways, including the JNK, JAK-STAT, and p38 MAPK pathways, which are largely activated through the cytokine induction Fig. 3. Distinction of BAL cellular compostion of sensitized guinea pigs (I column) compared to sensitized animals, that re- ceived allergen challenges (II column). Subpopulation content and ratio in BAL suspensions based on the morphological flow cytometric parameters side scatter (SS) versus forward scatter (FS) are illustrated in density plots Fig. 4. NO formation as a function of cellular size in BALF sub- populations derived from OVA sensitized guinea pigs (I column) and OVA sensitized animals treated with OVA aerosol exposures (II column). DAF-2DA stained cells are colored in green (ad- opted from [73] and supplemented here) Fig. 5. NO generation in accordance with the complexity of BALF cell subpopulations of OVA sensitized guinea pigs (I col- umn) and of OVA sensitized animals treated with OVA aerosol exposures (II column). DAF-2DA stained cells are colored in green (adopted from [73] and supplemented here) 411 Nitrosative events in atopic asthma pathogenesis [75]. In allergic asthma the immune response to in- haled antigens results from the activation of mast cells and antigen-specific Th2 cells, followed by the production of cytokines, including interleukin IL-4, IL-5 and IL-13 [3]. Further, IL-4 and IL-13 cause lung epithelial expression of iNOS to be upregulated via STAT-6, a process which is corticosteroid sensi- tive [76]. Moreover, the group of researchers pro- vided evidence, that IL-13 robustly induces the ex- pression of an active dimeric iNOS enzyme in pri- mary HAEC (human airway epithelial cells) main- tained in air–liquid interface culture, consistent with its expression in relation to Th2 inflammation [77]. Therefore, eNO is a direct signal of the Th2- mediated, pro-inflammatory cytokine mechanisms of central importance in the pathophysiology of al- lergic airway inflammation. At present, there is open debate about whether NO exacerbates or reduces au- toimmune and allergic chronic inflammation. The detrimental impact of NO is implemented due to its ability to launch a hyperinflammatory response, leading to tissue damage. Two mechanisms are pri- marily responsible for inducing NO-mediated dam- age: action of NO-induced inflammatory mediators and apoptosis of NO-targeted cells. NO modulates the production and function of cytokines released by immune cells, chemokines, and growth factors [78]. NO-mediated apoptosis represents one of the key factors contributing to enhanced inflammation and tissue damage during noninfectious respiratory dis- eases, such as asthma [79]. The recent study has re- vealed that NO induces eosinophil apoptosis in a mechanism mediated via ROS, JNK, and later mito- chondrial permeability transition [80]. iNOS-derived NO plays an immunomodulatory role, as it may shift the Th1/Th2 balance in favor of Th2, thus promoting IgE-mediated allergy [75, 81]. Th1 cells secrete IFN-γ, TNF-α, and IL-2 and pro- mote cellular immune responses against intracellular antigens, whereas Th2 cells secrete IL-4, IL-5, IL- 10, and IL-13, induce IgG1- and IgE-mediated hu- moral responses, and are important for the elimina- tion of large extracellular parasites such as helmin- thes and nematodes [82]. The molecular mechanisms of Th1 versus Th2 regulation rely on the counterbal- ance between expression of the IL-12 receptor β2- chain (IL-12Rβ2) 3 on Th1 cells and GATA-3 tran- scription in Th2 cells. IL-4 induced GATA-3 allows the stable commitment to the Th2 phenotype through promotion of Th2 cytokine production, such as IL-4, IL-5, IL-10 and IL-13 [83, 84]. IL-10 produced by Th2 cells further suppresses the Th1 development by inhibiting the secretion of IFN-γ and IL-12 and may also directly inhibit the induction of iNOS [81]. Moreover, NO markedly inhibits the induction of IL-2 promoter, which could account for most of the reduction in IL-2 production, and increases the pro- duction of IL-4 by effector T cells [85]. To conclude, NO can inhibit Th1 lymphokines but has no direct impact on the Th2 lymphokines production [78, 81]. As the continuous expression of iNOS in the normal epithelium is maintained mainly by IFN-γ [19, 21, 23], the present observations indicated that NO se- lectively inhibits the expansion of Th1 cells by a negative feedback mechanism. In contrast, low con- centrations of NO stimulate T cells to express IL- 12R and promote Th1 differentiation [86]. The counter-evidence of the absence of a direct effect of NO on Th2 cells was found since the NO donors inhibit the proliferation of Th1 and Th2 pop- ulations. The NO donors SIN-1 (3-morpholino-syd- nonimine) and SNAP (S-nitroso-N-acetyl-DL- penicillamine) suppress the proliferation of anti- CD3 or mitogen activated human peripheral Th1 and Th2 cell clones, and prevent the release of IFN-γ, IL-2, IL-4, IL-5 and IL-10 [87]. Additionally it has also been reported that NO inclusion to human bron- chial epithelial cells reversibly inhibits the prolifera- tion of activated Th1 and Th2 CD4+ T cells in atopic asthma [88]. IgE-dependent synthesis of NO by various cell populations bearing low-affinity IgE receptors (FceRII; CD23) implicates in local allergic and in- flammatory reactions, including macrophages, eo- sinophils and keratinocytes [89]. NO is also an im- portant agent in eosinophil migration and infiltra- tion. Several reports demonstrated that the acute treatment with the non-selective inhibitors of NO 412 O. O. Parilova, T. T. Volodina, S. G. Shandrenko reduced allergen-induced eosinophilia, showing that NO is involved in the inflammatory cell recruitment [44, 90, 91]. Furthermore, it has been found that the l-NAME (N ω-nitro-l-arginine methyl ester) treat- ment decreases the number of eosinophils positive for both nNOS and iNOS, whereas the treatment with 1400W, a highly selective iNOS inhibitor, de- creases only the iNOS-positive eosinophils, reduc- ing the eosinophil density in alveolar septa of aller- gen-sensitized animals [92]. In addition to these findings, NO is crucial to the numerous mast cell functions, including degranulation, adhesion, and protease release, as well as the production and secre- tion of chemokines and cytokines [42]. NO also con- trols the processes in immune system through regu- lation of the chemokine expression. Among the asthma pathophysiology processes, involving NO, are modulation of the co-stimulatory and adhesion molecules, such as VCAM-1 (vascular cell adhesion molecule-1), ICAM-1 (intercellular adhesion mole- cule-1), CD62E (E-selectin) and CD62P (P-selectin), synthesis and deposition of the extracellular material components via TGF-β [93]. The anti-inflammatory effects of NO may be me- diated by several mechanisms including the inhibi- tion of gene expression and the secretion of proin- flammatory cytokines [94, 95], reduction of the pro- inflammatory effects of cytokines such as IL-8 through the covalent modification by nitration of ty- rosine [96], or by protection against the programmed cell death through inactivation of the proteolytic en- zymes responsible for apoptosis [97, 98]. Thus, the endogenous NO synthesis in asthma, may be an im- portant early physiological defense mechanism against the injury and inflammation. Nevertheless, persistently elevated levels of the NO formation can deepen the tissue injury over time. Conclusions The presented evidence posits the nitrosative stress as a relevant pathogenic mechanism underlying atopic asthma. The diversity of NOS enzymes pro- duces different profiles of NO activities as well as of its derivatives facilitating and broadening the func- tional versatility of nitrosative events. Many studies and clinical observations are bringing to light a mul- titude of different mechanisms through which NO operates in respiratory tract and mediates dysfunc- tion in the disease. Mounting evidence of a link be- tween the NO synthesis and the allergic inflamma- tion raises the possibilities for therapeutic interven- tion. Two major challenges currently hinder the progress towards this aim; on one hand the multi- plicity of asthma nature with a complex immunoge- netic basis and a strong environmental contribution; on the other hand the likelihood of mimicking the full spectrum of responses to the allergen exposures and the tested pharmaceuticals by in vivo and in vitro models seems fragile. Hence, despite the significant advances in our understanding, the effective marker- addressed therapy strategies and reliable approaches for the precise diagnostics and monitoring are cur- rently still limited. REFERENCES 1. Busse WW, Lemanske RF Jr. Asthma. N Engl J Med. 2001;344(5):350–62. 2. Schröder NW, Maurer M. The role of innate immunity in asthma: leads and lessons from mouse models. Allergy. 2007;62(6):579–90. 3. Gaston B. The biochemistry of asthma. Biochim Biophys Acta. 2011;1810(11):1017–24. 4. Asher MI, Montefort S, Björkstén B, Lai CK, Strachan DP, Weiland SK, Williams H; ISAAC Phase Three Study Group. Worldwide time trends in the prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and eczema in child- hood: ISAAC Phases One and Three repeat multicountry cross-sectional surveys. Lancet. 2006;368(9537):733–43. 5. Illi S, von Mutius E, Lau S, Niggemann B, Grüber C, Wahn U; Multicentre Allergy Study (MAS) group. Perennial aller- gen sensitisation early in life and chronic asthma in chil- dren: a birth cohort study. Lancet. 2006;368(9537):763–70. 6. Ducharme FM. Inhaled corticosteroids versus leukotriene antagonists as first-line therapy for asthma: a systematic re- view of current evidence. Treat Respir Med. 2004;3(6):399– 405. 7. Colucci R, Fornai M, Tuccori M, Antonioli L, Pasqualetti G, Blandizzi C, Del Tacca M. Tolerability profiles of leu- kotriene receptor antagonists and long-acting beta2-adre- noceptor agonists in combination with inhaled corticoste- roids for treatment of asthma: a review. J Asthma. 2007;44(6):411–22. 413 Nitrosative events in atopic asthma pathogenesis 8. Shorter JH, Nelson DD, McManus JB, Zahniser MS, Sama S, Milton DK. Clinical study of multiple breath biomarkers of asthma and COPD (NO, CO2, CO and N2O) by infrared laser spectroscopy. J Breath Res. 2011;5(3):037108. 9. Moncada S, Higgs A. The L-arginine-nitric oxide pathway. N Engl J Med. 1993;329(27):2002–12. 10. Ghosh S, Erzurum SC. Nitric oxide metabolism in asthma patho- physiology. Biochim Biophys Acta. 2011;1810(11):1008–16. 11. Ricciardolo FL, Sterk PJ, Gaston B, Folkerts G. Nitric ox- ide in health and disease of the respiratory system. Physiol Rev. 2004;84(3):731–65. 12. Dweik RA, Boggs PB, Erzurum SC, Irvin CG, Leigh MW, Lundberg JO, Olin AC, Plummer AL, Taylor DR; American Thoracic Society Committee on Interpretation of Exhaled Nitric Oxide Levels (FENO) for Clinical Applications. An official ATS clinical practice guideline: interpretation of ex- haled nitric oxide levels (FENO) for clinical applications. Am J Respir Crit Care Med. 2011;184(5):602–15. 13. American Thoracic Society; European Respiratory Society. ATS/ERS recommendations for standardized procedures for the online and offline measurement of exhaled lower respi- ratory nitric oxide and nasal nitric oxide, 2005. Am J Respir Crit Care Med. 2005;171(8):912–30. 14. Saleh D, Ernst P, Lim S, Barnes PJ, Giaid A. Increased for- mation of the potent oxidant peroxynitrite in the airways of asthmatic patients is associated with induction of nitric ox- ide synthase: effect of inhaled glucocorticoid. FASEB J. 1998;12(11):929–37. 15. Stuehr DJ, Cho HJ, Kwon NS, Weise MF, Nathan CF. Purifi- cation and characterization of the cytokine-induced macro- phage nitric oxide synthase: an FAD- and FMN-containing flavoprotein. Proc Natl Acad Sci U S A. 1991;88(17):7773–7. 16. Lyons CR, Orloff GJ, Cunningham JM. Molecular cloning and functional expression of an inducible nitric oxide syn- thase from a murine macrophage cell line. J Biol Chem. 1992;267(9):6370–4. 17. Xie QW, Cho HJ, Calaycay J, Mumford RA, Swiderek KM, Lee TD, Ding A, Troso T, Nathan C. Cloning and character- ization of inducible nitric oxide synthase from mouse mac- rophages. Science. 1992;256(5054):225–8. 18. Nunokawa Y, Ishida N, Tanaka S. Cloning of inducible nitric oxide synthase in rat vascular smooth muscle cells. Bio- chem Biophys Res Commun. 1993;191(1):89–94. 19. Warner RL, Paine R 3rd, Christensen PJ, Marletta MA, Rich- ards MK, Wilcoxen SE, Ward PA. Lung sources and cytokine requirements for in vivo expression of inducible nitric oxide synthase. Am J Respir Cell Mol Biol. 1995;12(6):649–61. 20. Yates DH. Role of exhaled nitric oxide in asthma. Immunol Cell Biol. 2001;79(2):178–90. 21. Bogdan C. Nitric oxide and the immune response. Nat Im- munol. 2001;2(10):907–16. 22. Clancy RM, Abramson SB. Nitric oxide: a novel mediator of inflammation. Proc Soc Exp Biol Med. 1995;210(2):93–101. 23. Uetani K, Thomassen MJ, Erzurum SC. Nitric oxide syn- thase 2 through an autocrine loop via respiratory epithelial cell-derived mediator. Am J Physiol Lung Cell Mol Physiol. 2001;280(6):L1179–88. 24. Fujii Y, Magder S, Cernacek P, Goldberg P, Guo Y, Hussain SN. Endothelin receptor blockade attenuates lipopolysac- charide-induced pulmonary nitric oxide production. Am J Respir Crit Care Med. 2000;161(3 Pt 1):982–9. 25. Wang HH, Hsieh HL, Yang CM. Nitric oxide production by endothelin-1 enhances astrocytic migration via the tyrosine nitration of matrix metalloproteinase-9. J Cell Physiol. 2011;226(9):2244–56. 26. Alderton WK, Cooper CE, Knowles RG. Nitric oxide syn- thases: structure, function and inhibition. Biochem J. 2001;357(Pt 3):593–615. 27. Barnes PJ, Dweik RA, Gelb AF, Gibson PG, George SC, Grasemann H, Pavord ID, Ratjen F, Silkoff PE, Taylor DR, Zamel N. Exhaled nitric oxide in pulmonary diseases: a comprehensive review. Chest. 2010;138(3):682–92. 28. Gaston B, Drazen JM, Loscalzo J, Stamler JS. The biology of nitrogen oxides in the airways. Am J Respir Crit Care Med. 1994;149(2 Pt 1):538–51. 29. Pechkovsky DV, Zissel G, Goldmann T, Einhaus M, Taube C, Magnussen H, Schlaak M, Müller-Quernheim J. Pattern of NOS2 and NOS3 mRNA expression in human A549 cells and primary cultured AEC II. Am J Physiol Lung Cell Mol Physiol. 2002;282(4):L684–92. 30. Hollá LI, Bucková D, Kuhrová V, Stejskalová A, Francová H, Znojil V, Vácha J. Prevalence of endothelial nitric oxide synthase gene polymorphisms in patients with atopic asth- ma. Clin Exp Allergy. 2002;32(8):1193–8. 31. Maarsingh H, Leusink J, Bos IS, Zaagsma J, Meurs H. Ar- ginase strongly impairs neuronal nitric oxide-mediated air- way smooth muscle relaxation in allergic asthma. Respir Res. 2006;7:6. 32. Zuo L, Koozechian MS, Chen LL. Characterization of reac- tive nitrogen species in allergic asthma. Ann Allergy Asthma Immunol. 2014;112(1):18–22. 33. Hecker M, Cattaruzza M, Wagner AH. Regulation of induc- ible nitric oxide synthase gene expression in vascular smooth muscle cells. Gen Pharmacol. 1999;32(1):9–16. 34. Romanska HM, Polak JM, Coleman RA, James RS, Harmer DW, Allen JC, Bishop AE. iNOS gene upregulation is associ- ated with the early proliferative response of human lung fibro- blasts to cytokine stimulation. J Pathol. 2002;197(3):372–9. 35. Ermert M, Ruppert C, Günther A, Duncker HR, Seeger W, Ermert L. Cell-specific nitric oxide synthase-isoenzyme ex- pression and regulation in response to endotoxin in intact rat lungs. Lab Invest. 2002;82(4):425–41. 36. Panaro MA, Brandonisio O, Acquafredda A, Sisto M, Mito- lo V. Evidences for iNOS expression and nitric oxide pro- duction in the human macrophages. Curr Drug Targets Im- mune Endocr Metabol Disord. 2003;3(3):210–21. 414 O. O. Parilova, T. T. Volodina, S. G. Shandrenko 37. Prado CM, Leick-Maldonado EA, Yano L, Leme AS, Capelozzi VL, Martins MA, Tibério IF. Effects of nitric ox- ide synthases in chronic allergic airway inflammation and remodeling. Am J Respir Cell Mol Biol. 2006;35(4):457–65. 38. del Pozo V, de Arruda-Chaves E, de Andrés B, Cárdaba B, López-Farré A, Gallardo S, Cortegano I, Vidarte L, Jurado A, Sastre J, Palomino P, Lahoz C. Eosinophils transcribe and translate messenger RNA for inducible nitric oxide syn- thase. J Immunol. 1997;158(2):859–64. 39. Evans TJ, Buttery LD, Carpenter A, Springall DR, Polak JM, Cohen J. Cytokine-treated human neutrophils contain induc- ible nitric oxide synthase that produces nitration of ingested bacteria. Proc Natl Acad Sci U S A. 1996;93(18):9553–8. 40. Webb JL, Polak JM, Evans TJ. Effect of adhesion on induc- ible nitric oxide synthase (iNOS) production in purified hu- man neutrophils. Clin Exp Immunol. 2001;123(1):42–8. 41. Lu L, Bonham CA, Chambers FG, Watkins SC, Hoffman RA, Simmons RL, Thomson AW. Induction of nitric oxide syn- thase in mouse dendritic cells by IFN-gamma, endotoxin, and interaction with allogeneic T cells: nitric oxide produc- tion is associated with dendritic cell apoptosis. J Immunol. 1996;157(8):3577–86. 42. McCauley SD, Gilchrist M, Befus AD. Nitric oxide: a major determinant of mast cell phenotype and function. Mem Inst Oswaldo Cruz. 2005;100 Suppl 1:11–4. 43. Gilchrist M, McCauley SD, Befus AD. Expression, localiza- tion, and regulation of NOS in human mast cell lines: ef- fects on leukotriene production. Blood. 2004;104(2):462–9. 44. Bidri M, Ktorza S, Vouldoukis I, Le Goff L, Debré P, Guil- losson JJ, Arock M. Nitric oxide pathway is induced by Fc epsilon RI and up-regulated by stem cell factor in mouse mast cells. Eur J Immunol. 1997;27(11):2907–13. 45. Inoue T, Suzuki Y, Yoshimaru T, Ra C. Nitric oxide protects mast cells from activation-induced cell death: the role of the phosphatidylinositol-3 kinase-Akt-endothelial nitric oxide synthase pathway. J Leukoc Biol. 2008;83(5):1218–29. 46. Gilchrist M, Savoie M, Nohara O, Wills FL, Wallace JL, Befus AD. Nitric oxide synthase and nitric oxide production in in vivo-derived mast cells. J Leukoc Biol. 2002;71(4):618–24. 47. Lin TJ, Hirji N, Nohara O, Stenton GR, Gilchrist M, Befus AD. Mast cells express novel CD8 molecules that selectively mod- ulate mediator secretion. J Immunol. 1998;161(11):6265–72. 48. Suzuki Y, Inoue T, Ra C. Endothelial nitric oxide synthase is essential for nitric oxide generation, L-type Ca2+ channel activation and survival in RBL-2H3 mast cells. Biochim Biophys Acta. 2010;1803(3):372–85. 49. Furuke K, Burd PR, Horvath-Arcidiacono JA, Hori K, Mo- stowski H, Bloom ET. Human NK cells express endothelial nitric oxide synthase, and nitric oxide protects them from activation-induced cell death by regulating expression of TNF-alpha. J Immunol. 1999;163(3):1473–80. 50. Lancaster JR Jr. Nitroxidative, nitrosative, and nitrative stress: kinetic predictions of reactive nitrogen species chem- istry under biological conditions. Chem Res Toxicol. 2006;19(9):1160–74. 51. Hess DT, Matsumoto A, Kim SO, Marshall HE, Stamler JS. Protein S-nitrosylation: purview and parameters. Nat Rev Mol Cell Biol. 2005;6(2):150–66. 52. Martínez MC, Andriantsitohaina R. Reactive nitrogen species: molecular mechanisms and potential significance in health and disease. Antioxid Redox Signal. 2009;11(3):669–702. 53. Alving K, Weitzberg E, Lundberg JM. Increased amount of nitric oxide in exhaled air of asthmatics. Eur Respir J. 1993;6(9):1368–70. 54. Kharitonov SA, Yates D, Robbins RA, Logan-Sinclair R, Shinebourne EA, Barnes PJ. Increased nitric oxide in exhaled air of asthmatic patients. Lancet. 1994;343(8890):133–5. 55. Massaro AF, Gaston B, Kita D, Fanta C, Stamler JS, Drazen JM. Expired nitric oxide levels during treatment of acute asthma. Am J Respir Crit Care Med. 1995;152(2):800–3. 56. Yates DH, Kharitonov SA, Robbins RA, Thomas PS, Barnes PJ. Effect of a nitric oxide synthase inhibitor and a gluco- corticosteroid on exhaled nitric oxide. Am J Respir Crit Care Med. 1995;152(3):892–6. 57. Hamid Q, Springall DR, Riveros-Moreno V, Chanez P, How- arth P, Redington A, Bousquet J, Godard P, Holgate S, Po- lak JM. Induction of nitric oxide synthase in asthma. Lan- cet. 1993;342(8886–8887):1510–3. 58. Guo FH, Comhair SA, Zheng S, Dweik RA, Eissa NT, Thom- assen MJ, Calhoun W, Erzurum SC. Molecular mechanisms of increased nitric oxide (NO) in asthma: evidence for tran- scriptional and post-translational regulation of NO synthe- sis. J Immunol. 2000;164(11):5970–80. 59. De Sanctis GT, MacLean JA, Hamada K, Mehta S, Scott JA, Jiao A, Yandava CN, Kobzik L, Wolyniec WW, Fabian AJ, Venugopal CS, Grasemann H, Huang PL, Drazen JM. Con- tribution of nitric oxide synthases 1, 2, and 3 to airway hy- perresponsiveness and inflammation in a murine model of asthma. J Exp Med. 1999;189(10):1621–30. 60. Feder LS, Stelts D, Chapman RW, Manfra D, Crawley Y, Jones H, Minnicozzi M, Fernandez X, Paster T, Egan RW, Kreutner W, Kung TT. Role of nitric oxide on eosinophilic lung inflammation in allergic mice. Am J Respir Cell Mol Biol. 1997;17(4):436–42. 61. Xiong Y, Karupiah G, Hogan SP, Foster PS, Ramsay AJ. Inhibition of allergic airway inflammation in mice lacking nitric oxide synthase 2. J Immunol. 1999;162(1):445–52. 62. Popov TA. Human exhaled breath analysis. Ann Allergy Asthma Immunol. 2011;106(6):451–6. 63. Jatakanon A, Lim S, Kharitonov SA, Chung KF, Barnes PJ. Correlation between exhaled nitric oxide, sputum eosino- phils, and methacholine responsiveness in patients with mild asthma. Thorax. 1998;53(2):91–5. 64. Mattes J, Storm van’s Gravesande K, Reining U, Alving K, Ihorst G, Henschen M, Kuehr J. NO in exhaled air is corre- lated with markers of eosinophilic airway inflammation in 415 Nitrosative events in atopic asthma pathogenesis corticosteroid-dependent childhood asthma. Eur Respir J. 1999;13(6):1391–5. 65. Piacentini GL, Bodini A, Costella S, Vicentini L, Mazzi P, Sperandio S, Boner AL. Exhaled nitric oxide and sputum eosinophil markers of inflammation in asthmatic children. Eur Respir J. 1999;13(6):1386–90. 66. Donohue JF, Jain N. Exhaled nitric oxide to predict cortico- steroid responsiveness and reduce asthma exacerbation rates. Respir Med. 2013;107(7):943–52. 67. Haldar P, Brightling CE, Hargadon B, Gupta S, Monteiro W, Sousa A, Marshall RP, Bradding P, Green RH, Wardlaw AJ, Pavord ID. Mepolizumab and exacerbations of refrac- tory eosinophilic asthma. N Engl J Med. 2009;360(10):973– 84. 68. Corren J, Lemanske RF, Hanania NA, Korenblat PE, Parsey MV, Arron JR, Harris JM, Scheerens H, Wu LC, Su Z, Mo- sesova S, Eisner MD, Bohen SP, Matthews JG. Lebrikizum- ab treatment in adults with asthma. N Engl J Med. 2011;365(12):1088–98. 69. Warke TJ, Fitch PS, Brown V, Taylor R, Lyons JD, Ennis M, Shields MD. Exhaled nitric oxide correlates with airway eo- sinophils in childhood asthma. Thorax. 2002;57(5):383–7. 70. Bousquet J, Chanez P, Lacoste JY, Barnéon G, Ghavanian N, Enander I, Venge P, Ahlstedt S, Simony-Lafontaine J, Go- dard P, et al. Eosinophilic inflammation in asthma. N Engl J Med. 1990;323(15):1033–9. 71. Meurs H, Maarsingh H, Zaagsma J. Arginase and asthma: novel insights into nitric oxide homeostasis and airway hyper- responsiveness. Trends Pharmacol Sci. 2003;24(9):450–5. 72. Yeadon M, Price R. Induction of calcium-independent nitric oxide synthase by allergen challenge in sensitized rat lung in vivo. Br J Pharmacol. 1995;116(6):2545–6. 73. Parilova OO, Shandrenko SG. Interconnection between ni- tric oxide formation and hypersensitivity parameters under guinea pig model of acute asthma with multiple challenges. Ukr Biochem J. 2015;87(5):113–23. 74. Trifilieff A, Fujitani Y, Mentz F, Dugas B, Fuentes M, Ber- trand C. Inducible nitric oxide synthase inhibitors suppress airway inflammation in mice through down-regulation of chemokine expression. J Immunol. 2000;165(3):1526–33. 75. Pautz A, Art J, Hahn S, Nowag S, Voss C, Kleinert H. Regu- lation of the expression of inducible nitric oxide synthase. Nitric Oxide. 2010;23(2):75–93. 76. Alving K, Malinovschi A. Basic aspects of exhaled nitric ox- ide. European Respiratory Monograph. Plymouth, UK, Lat- imer Trend & So Ltd, European Respiratory Society; 2010; 69:1–31. 77. Chibana K, Trudeau JB, Mustovich AT, Hu H, Zhao J, Bal- zar S, Chu HW, Wenzel SE. IL-13 induced increases in ni- trite levels are primarily driven by increases in inducible nitric oxide synthase as compared with effects on arginases in human primary bronchial epithelial cells. Clin Exp Al- lergy. 2008;38(6):936–46. 78. Bove PF, van der Vliet A. Nitric oxide and reactive nitrogen species in airway epithelial signaling and inflammation. Free Radic Biol Med. 2006;41(4):515–27. 79. Mgbemena V, Segovia JA, Chang TH, Tsai SY, Cole GT, Hung CY, Bose S. Transactivation of inducible nitric oxide synthase gene by Kruppel-like factor 6 regulates apoptosis during influenza A virus infection. J Immunol. 2012;189(2):606–15. 80. Ilmarinen-Salo P, Moilanen E, Kinnula VL, Kankaanranta H. Nitric oxide-induced eosinophil apoptosis is dependent on mitochondrial permeability transition (mPT), JNK and oxida- tive stress: apoptosis is preceded but not mediated by early mPT-dependent JNK activation. Respir Res. 2012;13:73. 81. Curran AD. The role of nitric oxide in the development of asthma. Int Arch Allergy Immunol. 1996;111(1):1–4. 82. Abbas AK, Murphy KM, Sher A. Functional diversity of helper T lymphocytes. Nature. 1996;383(6603):787–93. 83. Ouyang W, Ranganath SH, Weindel K, Bhattacharya D, Murphy TL, Sha WC, Murphy KM. Inhibition of Th1 devel- opment mediated by GATA-3 through an IL-4-independent mechanism. Immunity. 1998;9(5):745–55. 84. Ferber IA, Lee HJ, Zonin F, Heath V, Mui A, Arai N, O’Garra A. GATA-3 significantly downregulates IFN-gamma pro- duction from developing Th1 cells in addition to inducing IL-4 and IL-5 levels. Clin Immunol. 1999;91(2):134–44. 85. Chang RH, Feng MH, Liu WH, Lai MZ. Nitric oxide in- creased interleukin-4 expression in T lymphocytes. Immu- nology. 1997;90(3):364–9. 86. Niedbala W, Wei XQ, Campbell C, Thomson D, Komai-Ko- ma M, Liew FY. Nitric oxide preferentially induces type 1 T cell differentiation by selectively up-regulating IL-12 re- ceptor beta 2 expression via cGMP. Proc Natl Acad Sci U S A. 2002;99(25):16186–91. 87. Bauer H, Jung T, Tsikas D, Stichtenoth DO, Frölich JC, Neumann C. Nitric oxide inhibits the secretion of T-helper 1- and T-helper 2-associated cytokines in activated human T cells. Immunology. 1997;90(2):205–11. 88. Eriksson U, Egermann U, Bihl MP, Gambazzi F, Tamm M, Holt PG, Bingisser RM. Human bronchial epithelium con- trols TH2 responses by TH1-induced, nitric oxide-mediated STAT5 dephosphorylation: implications for the pathogene- sis of asthma. J Immunol. 2005;175(4):2715–20. 89. Adler KB, Fischer BM, Li H, Chloe NH, Wright DT. Hyper secretion of cumin is response to inflammatory mediators by guinea pig tracheal epithelial cells in vitro is blocked by inhibition of nitric oxide synthesis. Am J Respir Cell Mol Biol. 1995;13:526–530. 90. Angeli P, Prado CM, Xisto DG, Silva PL, Pássaro CP, Na- kazato HD, Leick-Maldonado EA, Martins MA, Rocco PR, Tibério IF. Effects of chronic L-NAME treatment lung tis- sue mechanics, eosinophilic and extracellular matrix re- sponses induced by chronic pulmonary inflammation. Am J Physiol Lung Cell Mol Physiol. 2008;294(6):L1197–205. 416 O. O. Parilova, T. T. Volodina, S. G. Shandrenko 91. Prado CM, Leick-Maldonado EA, Kasahara DI, Capelozzi VL, Martins MA, Tibério IF. Effects of acute and chronic nitric oxide inhibition in an experimental model of chronic pulmonary allergic inflammation in guinea pigs. Am J Physiol Lung Cell Mol Physiol. 2005;289(4):L677–83. 92. Iijima H, Duguet A, Eum SY, Hamid Q, Eidelman DH. Ni- tric oxide and protein nitration are eosinophil dependent in allergen-challenged mice. Am J Respir Crit Care Med. 2001;163(5):1233–40. 93. Starling CM, Prado CM, Leick-Maldonado EA, Lanças T, Reis FG, Aristóteles LR, Dolhnikoff M, Martins MA, Tibério IF. Inducible nitric oxide synthase inhibition attenuates lung tissue responsiveness and remodeling in a model of chronic pulmonary inflammation in guinea pigs. Respir Physiol Neurobiol. 2009;165(2–3):185–94. 94. Karpuzoglu E, Ahmed SA. Estrogen regulation of nitric ox- ide and inducible nitric oxide synthase (iNOS) in immune cells: implications for immunity, autoimmune diseases, and apoptosis. Nitric Oxide. 2006;15(3):177–86. 95. Thomassen MJ, Buhrow LT, Connors MJ, Kaneko FT, Er- zurum SC, Kavuru MS. Nitric oxide inhibits inflammatory cytokine production by human alveolar macrophages. Am J Respir Cell Mol Biol. 1997;17(3):279–83. 96. Raychaudhuri B, Dweik R, Connors MJ, Buhrow L, Malur A, Drazba J, Arroliga AC, Erzurum SC, Kavuru MS, Thom- assen MJ. Nitric oxide blocks nuclear factor-kappaB activa- tion in alveolar macrophages. Am J Respir Cell Mol Biol. 1999;21(3):311–6. 97. Sato E, Simpson KL, Grisham MB, Koyama S, Robbins RA. Reactive nitrogen and oxygen species attenuate interleukin- 8-induced neutrophil chemotactic activity in vitro. J Biol Chem. 2000;275(15):10826–30. 98. Kim YM, Talanian RV, Billiar TR. Nitric oxide inhibits apoptosis by preventing increases in caspase-3-like activ- ity via two distinct mechanisms. J Biol Chem. 1997;272(49):31138–48. Нітрозативні події в патогенезі атопічної астми О. О. Парилова, Т. Т. Володіна, С. Г. Шандренко Кореляцію між високим рівнем оксиду азоту, що видихається та еозинофіл-опосередкованим запаленням дихальних шляхів у пацієнтів з атопічною бронхіальною астмою добре доведено. Це створює передумови існування регуляторного механізму зворотнього зв’язку між ними. Тому стаття стисло наводить свідчення стосовно біосинтезу, структурних особливостей ен- зиму, регуляції експресії його ізоформ та дії оксиду азоту, що допомагає з’ясувати молекулярні механізми завдяки яким оксид азоту сприяє загостренню перебігу захворювання. В по- передній експериментальній роботі ми продемонстрували, що інфільтрат імунних клітин, які персистують в дихальних шля- хах, робить внесок у синтез оксиду азоту в респіраторному тракті під час пізньої алергічної реакції за моделі гострої брон- хіальної астми мурчаків з множинними провокаціями алерге- ну. На основі цих даних автори констатують, що оксид азоту є додатковим сигналом індукції відповіді Th2 ланки та залуче- ний в складну мережу імунної регуляції, характерної для фено- типу атопічної астми. Ключов і   слова: оксид азоту, атопічна астма, синтази оксиду азоту, алергічне запалення дихальних шляхів, еозино- філія. Нитрозативные события в патогенезе атопической  астмы Е. А. Парилова, Т. Т. Володина, С. Г. Шандренко Корреляция между высоким уровнем оксида азота в выдыхае- мом воздухе и эозинофил-опосредованным воспалением ды- хательных путей у пациентов с атопической бронхиальной астмой хорошо обоснована. Это порождает предпосылку, что существует механизм регулирования обратной связи между ними. Таким образом, статья кратко описывает данные касаю- щиеся биосинтеза, структурных особенностей энзима, регули- рования экспрессии его изоформ и воздействия оксида азота, которые помогают выяснить молекулярные механизмы, благо- даря чему оксид азота селективно содействует обострению астмы. В предыдущей экспериментальной работе мы показа- ли, что инфильтрат иммунных клеток в дыхательных путях дополняет синтез оксида азота в респираторном тракте во вре- мя аллергического воспаления на модели острой бронхиаль- ной астмы морских свинок с множественными провокациями аллергена. На основании этих данных авторы констатируют, что оксид азота представляет собой дополнительный сигнал индукции ответа Th2 звена и значительно вовлечен в сложную сеть иммунной регуляции, отличительной для фенотипа ато- пической астмы. Ключевые  слова: оксид азота, атопическая астма, синта- зы оксида азота, аллергическое воспаление дыхательных пу- тей, эозинофилия. Received 29.09.2015