Next Article in Journal
A Pattern of Care Report on the Management of Patients with Squamous Cell Carcinoma of the Anus—A Study by the Italian Association of Radiotherapy and Clinical Oncology (AIRO) Gastrointestinal Tumors Study Group
Previous Article in Journal
A Warning Call for Fertility Preservation Methods for Women Undergoing Gonadotoxic Cancer Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Nitric Oxide Synthase 2 Promoter Polymorphism Is a Risk Factor for Allergic Asthma in Children

by
Joanna Nowakowska
1,*,†,
Paulina Sobkowiak
2,†,
Anna Bręborowicz
2,
Magdalena Mrówczyńska
2,
Irena Wojsyk-Banaszak
2 and
Aleksandra Szczepankiewicz
1
1
Molecular and Cell Biology Unit, Poznan University of Medical Sciences, 60-572 Poznan, Poland
2
Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Medicina 2021, 57(12), 1341; https://doi.org/10.3390/medicina57121341
Submission received: 8 November 2021 / Revised: 3 December 2021 / Accepted: 5 December 2021 / Published: 8 December 2021
(This article belongs to the Section Pediatrics)

Abstract

:
Background and Objectives: In paediatric population, atopic asthma is associated with increased eosinophil counts in patients, that correlate with the airway inflammation measured by the concentration of nitric oxide in exhaled air (FeNO). As the FeNO level is a biomarker of atopic asthma, we assumed that polymorphisms in nitric synthases genes may represent a risk factor for asthma development. The purpose of this study was to analyse the association of NOS genetic variants with childhood asthma in the Polish population. Materials and methods: In study we included 443 children—220 patients diagnosed with atopic asthma and 223 healthy control subjects. We have genotyped 4 single nucleotide polymorphisms (SNP) from 3 genes involved in the nitric oxide synthesis (NOS1, NOS2 and NOS3). All analyses were performed using polymerase chain reaction with restriction fragments length polymorphism (PCR-RFLP). Results: We observed significant differences between cases and controls in SNP rs10459953 in NOS2 gene, considering both genotypes (p = 0.001) and alleles (p = 0.0006). The other analyzed polymorphisms did not show association with disease. Conclusions: According to our results, 5′UTR variant within NOS2 isoform may have an impact of asthma susceptibility in the population of Polish children. Further functional studies are required to understand the role of iNOS polymorphism in NOS2 translation and to consider it as a novel risk factor in childhood asthma. The next step would be to apply this knowledge to improve diagnosis and develop novel personalized asthma therapies.

1. Introduction

Asthma is a heterogeneous disease characterized by chronic airway inflammation, variable airflow limitation and airway hyperresponsiveness. Type of airway inflammation differs between asthmatic patients, but in childhood asthma up to 80% of patients demonstrates T helper cells subtype 2 (Th2)-mediated allergic inflammation with overexpression of Th2 cytokines (e.g., IL-4, IL-5 and IL-13), increased IgE production and enhanced recruitment of eosinophils to the site of inflammation in the airways [1]. In these cases, asthma is frequently associated with atopic dermatitis and rhinitis [2,3]. Increased eosinophil counts correlate with the fractional concentration of nitric oxide in exhaled air (fraction of exhaled NO; FeNO) and this parameter is an useful and non-invasive biomarker of airway inflammation severity [4]. Previous studies have shown increased levels of FeNO in asthmatic individuals that associated with eosinophilic inflammation in the airways [5,6]. Moreover, several previous papers reported increased FeNO concentration in paediatric and adult asthmatic patients [7,8,9], also in particularly in relation to atopic phenotype [10,11]. In a 3-year-follow-up study elevated FeNO was associated with an increased risk for new-onset asthma in schoolchildren, thus making this parameter a potential asthma predictor [12]. Despite many years of research, as well as awareness of the relationship of FeNO with asthma exacerbations, its clinical value in the population of children is still not fully understood [13]. Nitric oxide (NO) is formed by conversion of L-arginine to L-citrulline in the presence of one of three distinct isoforms of NO synthases (NOSs): a neuronal isoform (nNOS or NOS1), an endothelial isoform (eNOS or NOS3) and inducible NOS (iNOS or NOS2). The NOS1 and 3 each generate small amount of NO in the lungs whereas NOS2 generates the highest amounts of NO during inflammation [14]. These isoforms are encoded by three different genes (NOS1, NOS2 and NOS3, respectively). NOS1 is expressed in human airway nerves, NOS3 in endothelial cells of the pulmonary circulation and NOS2 in T cells, macrophages, epithelial cells, mast cells, eosinophils and neutrophils in response to inflammatory stimuli such as cytokines, oxidants, viral infections and allergen exposure [15,16]. Previous genetic studies showed that NOS polymorphisms were associated with asthma and FeNO levels in adult and paediatric populations worldwide, including large cohort study of over 2000 white children of Hispanic and non-Hispanic origin from California [17]. However, no previous study analysed the association between asthma and NOS genetic variants in paediatric asthmatics of Central European origin. Therefore, the aim of our study was to determine whether the previously reported associations of NOS variants with asthma are also observed in Polish asthmatic children. In this case-control study we investigated a possible association of the single nucleotide polymorphisms (SNPs) in all three NOS genes with IgE-mediated childhood asthma and FeNO levels.

2. Materials and Methods

In this case-control study, we included 443 Polish children: 220 patients diagnosed with atopic asthma and 223 healthy control subjects. Asthma diagnosis was made according to Global Initiative for Asthma (GINA) guidelines and included lung function test, fractionated exhaled nitric oxide (FeNO) measurement and atopic parameters (serum total IgE level, skin prick tests with common inhaled allergens). The detailed clinical description of the performed tests was previously described by Szczepankiewicz et al. [18].
Control group: Control group included 223 healthy subjects from the same geographic region. In the control group, clinical examination excluded current and past asthma or allergy symptoms, we also confirmed normal spirometry and normal FeNO concentration (<20 ppb) in these children.
Genotyping: We have genotyped 4 SNPs from 3 genes involved in the nitric oxide synthesis (NOS1, NOS2 and NOS3). The SNP selection was based on at least two of the following criteria: functionality demonstrated in the previous experimental studies, minor allele frequency above 0.05 or previous association with asthma or atopy. DNA was extracted from 10 mL of EDTA anticoagulated whole blood using the salting out method presented by Miller et al. [19] or from the saliva using Oragene DNA isolation system (DNA Genotek, Ottawa, ON, Canada, ). Genotyping of polymorphisms was performed using PCR-RFLP (polymerase chain reaction with restriction fragments length polymorphism) method. PCR reactions with specific primers were performed in MasterCycler (Eppendorf, Hamburg, Germany) thermal cycler. PCR products for each sample were then digested overnight (37 °C for Eco72I, FspBI and MboI, 65 °C for FspBI and TasI) with 0.5 U of appropriate restriction endonuclease. After RFLP, the digested products were resolved in agarose gel to identify the genotypes. Undigested PCR products for analyzed polymorphisms were re-digested to confirm the results. Primer sequences, PCR product size, restriction endonucleases used for genotyping and band size for particular alleles were shown in Table 1. For each reaction plate genomic control DNA samples and non-template controls were included. To check for RFLP genotyping accuracy, 25% of randomly chosen samples from both groups for each SNP were repeatedly analyzed. Genotyping was performed without knowing the clinical status of the subjects.
Statistical analysis: Pearson’s Chi-square (χ2) test and Fisher’s exact test were used to test differences in the genotypic and allelic (respectively) distribution in case control analysis. Stepwise logistic regression analysis was performed using disease status (case-control) as dichotomous dependent variable and the following predictors included in the model: analyzed polymorphisms, exhaled nitric oxide level, age and gender. Calculations were performed using Statistica version 10.0 software. Concordance with Hardy–Weinberg law was performed using a calculator developed by Court lab (Court M 2008). Higher-order gene-gene interactions were analyzed using genetic model-free multifactor dimensionality reduction (MDR) approach (v.2.0 beta 8.3) [20] using criteria described in the previous study [18].

3. Results

The group of patients and control subjects were similar in age and gender (p > 0.05). The asthmatic children showed significantly decreased lung function and increased FeNO levels (p < 0.001). The results were shown in Table 2.

3.1. Hardy–Weinberg Equilibrium Test

All studied polymorphisms were tested for concordance with the Hardy–Weinberg equilibrium law (HWE). No deviation was observed for asthma patients and control group (p > 0.05).

3.2. Single Marker Association Results

Genotype distribution and allele frequency were significantly different for rs10459953 polymorphism in NOS2 gene and TT genotype and T allele were significantly more frequent in the asthmatic patients (p = 0.001 and p = 0.0006, respectively). The other analyzed polymorphisms did not show association with the disease. The results of association analysis were presented in Table 3. The analysis stratified by gender confirmed strong association of rs10459953 polymorphism in NOS2 gene with asthma showing significantly higher frequency of TT genotype and T allele among asthmatic boys (p = 0.00002 for genotypes, p = 0.000001 for alleles), but not in girls (p > 0.05) (data not shown).

3.3. Gene × Gene Interaction Analysis

We found a significant epistatic interaction between two variants of NOS2 gene (rs2297518 and rs10459953, 2-locus model) and childhood asthma risk. The testing balanced accuracy was 63.3% and the cross-validation consistency was 80% (p = 0.025 after 1000 permutations). One of these polymorphisms (C/T in NOS2 gene) showed a significant association with the risk of asthma in a single marker analysis. We did not observe any epistatic interaction between NOS polymorphisms and exhaled NO level. Results of interaction analysis of analyzed SNPs for each comparison were summarized in Table 4.

4. Discussion

Our study shows an association between rs10459953 in NOS2 gene and childhood asthma, which was the first to explore it in Polish population. Only a few studies have analyzed this polymorphism in the context of various diseases, such as depression [21], benign prostatic hyperplasia [22], diabetes mellitus [23], male infertility [24] and stroke [25], but none of them investigated it in the context of asthma. Some studies considered NOS2 polymorphisms in various populations. Salam et al. analysed childhood asthma in US population, stratified by origin (Hispanic versus non—Hispanic), and identified complex sets of haplotypes in the NOS2 promoter coding regions and these haplotypes were associated with FeNO in children with and without asthma [17]. In turn, Bouzigon et al. found significant associations of FeNO levels with two SNPs in NOS3 (rs2853796 and rs1549758) and one SNP in NOS2 (rs6505510) in non-asthmatic adults. Considering asthmatic subjects, a single association was detected between FeNO levels and one SNP in NOS3 (rs743507) [26]. In a Japanese population, the number of CCTTT repeats in the NOS2 promoter region appeared to be significantly associated with FeNO levels in adults, but only before the treatment with inhaled corticosteroids. The same study researched also seven SNPs in this gene, none of them were related to FeNO levels [27]. Dahgam et al. analysed 49 SNPs in NOS2, NOS1 and NOS3 in Swedish adult general population, considered two in NOS2 (rs9901734 and rs3729508) and one in NOS3 (rs7830) as independently associated with FeNO levels. In rs7830, the relation depended on asthma status, suggesting that NOS3 genetic variation plays the most important role among all nitric oxide synthases in asthmatic adults [28]. In our study, we found that NOS2 variant located in the 5′UTR region was associated with childhood asthma in the Polish population. In addition, result of epistatic analysis showed only significant epistatic effect between two variants within NOS2, suggesting that this combination affects asthma susceptibility, but not FeNO levels. This is the first analysis showing that 5′UTR variant in NOS2 gene may be associated with the risk of childhood asthma. Although the functional effect of this polymorphism on NOS2 posttranscriptional regulation has not been determined yet, the recent study by Sample et al. [29] showed that 5′UTR variants may affect 5 cap structure and ribosome loading and thus affect translation.
The results of NOS1 and NOS3 association studies were inconsistent depending on the studied population. For example, NOS1 polymorphism (AAT repeats in intron 20) was associated with higher FeNO concentration in adult asthmatics of Caucasian origin [30], but later studies showed a relationship between NOS1 variants and FeNO also in healthy population and suggested they were associated with FeNO levels in women. Such association was not observed for NOS3 variants [31]. Some NOS1 and NOS3 polymorphisms studied in various populations showed a positive correlation with atopy [32], total IgE concentration [33,34] or sensitization to inhaled allergens (e.g., Dermatophagoides pteronyssinus) and a possible relationship with the risk of asthma [35]. The polymorphism in NOS1 gene correlated with the physician-diagnosed asthma and the risk of severe exacerbation in children, as demonstrated by Duckworth et al., but such relationship was not observed in regard to the polymorphism in NOS3 gene [36].
As genetic variants within NOS genes may influence NO levels in asthmatic airways, they may also influence allergic response and secretion of Th2 cytokines. The most important cytokines associated with allergic asthma are IL-4, IL-13 and IL-5, that are related to the activity of immune cells such as T cells, B cells producing specific IgE antibodies, eosinophils and mast cells [37]. The secretion of IL-5 results from an increased number of eosinophils recruited in the process of allergic inflammation, while enhanced release of IL-4 and IL-13 from Th2 cells, affect epithelial cells, thus contributing to increased fractional exhaled nitric oxide levels in the asthmatic airways [38]. Our previous study showed the association of IL-4, its receptor IL4Rα and IL-13 polymorphisms with asthma in the same pediatric cohort that was analyzed in this study [39]. Therefore, these studies confirm that Th2 cytokines are associated with allergic asthma in our pediatric population.

5. Conclusions

Taken together, our results suggest, that 5′UTR variant within inducible NOS isoform may affect susceptibility of childhood asthma in Polish population. However, further functional studies are required to elucidate if this variant affects NOS2 translation and thus may represent of novel risk factor in the allergic asthma. The next step would be to apply this knowledge to improve diagnosis and develop novel personalized asthma therapies.

Author Contributions

Conceptualization, P.S. and A.S.; data curation, J.N.; methodology, J.N. and A.S.; resources, A.B., M.M. and I.W.-B.; supervision, A.S.; writing—original draft, J.N.; writing—review and editing, P.S., A.B., I.W.-B. and A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Polish National Science Centre, Grant No. 2019/35/B/NZ5/02906 (A. Szczepankiewicz).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Local ethics committee of Poznan University of Medical Sciences (Agreement No. 248/08, 6 March 2008).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study, as well as their parents.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hering, T. [Update of the GINA-Recommendations]. MMW Fortschr. Med. 2017, 159, 63–64. [Google Scholar] [CrossRef] [PubMed]
  2. Nettis, E.; Ortoncelli, M.; Pellacani, G.; Foti, C.; di Leo, E.; Patruno, C.; Rongioletti, F.; Argenziano, G.; Ferrucci, S.M.; Macchia, L.; et al. A Multicenter Study on the Prevalence of Clinical Patterns and Clinical Phenotypes in Adult Atopic Dermatitis. J. Investig. Allergol. Clin. Immunol. 2020, 30, 448–450. [Google Scholar] [CrossRef] [PubMed]
  3. Dattola, A.; Bennardo, L.; Silvestri, M.; Nisticò, S.P. What’s new in the treatment of atopic dermatitis? Dermatol. Ther. 2019, 32, e12787. [Google Scholar] [CrossRef] [PubMed]
  4. Kharitonov, S.A.; Barnes, P.J. Exhaled Markers of Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2001, 163, 1693–1722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Alving, K.; Weitzberg, E.; Lundberg, J.M. Increased amount of nitric oxide in exhaled air of asthmatics. Eur. Respir. J. 1993, 6, 1368–1370. [Google Scholar] [PubMed]
  6. Deykin, A.; Massaro, A.F.; Drazen, J.M.; Israel, E. Exhaled nitric oxide as a diagnostic test for asthma: Online versus offline techniques and effect of flow rate. Am. J. Respir. Crit. Care Med. 2002, 165, 1597–1601. [Google Scholar] [CrossRef] [PubMed]
  7. van der Valk, R.J.P.; Baraldi, E.; Stern, G.; Frey, U.; de Jongste, J.C. Daily exhaled nitric oxide measurements and asthma exacerbations in children. Allergy 2012, 67, 265–271. [Google Scholar] [CrossRef] [PubMed]
  8. Avital, A.; Uwyyed, K.; Berkman, N.; Godfrey, S.; Bar-Yishay, E.; Springer, C. Exhaled nitric oxide and asthma in young children. Pediatr. Pulmonol. 2001, 32, 308–313. [Google Scholar] [CrossRef]
  9. Malmberg, L.P.; Pelkonen, A.S.; Haahtela, T.; Turpeinen, M. Exhaled nitric oxide rather than lung function distinguishes preschool children with probable asthma. Thorax 2003, 58, 494–499. [Google Scholar] [CrossRef] [Green Version]
  10. Stern, G.; de Jongste, J.; van der Valk, R.; Baraldi, E.; Carraro, S.; Thamrin, C.; Frey, U. Fluctuation phenotyping based on daily fraction of exhaled nitric oxide values in asthmatic children. J. Allergy Clin. Immunol. 2011, 128, 293–300. [Google Scholar] [CrossRef]
  11. Franklin, P.J.; Turner, S.W.; Le Souëf, P.N.; Stick, S.M. Exhaled nitric oxide and asthma: Complex interactions between atopy, airway responsiveness, and symptoms in a community population of children. Thorax 2003, 58, 1048–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bastain, T.M.; Islam, T.; Berhane, K.T.; McConnell, R.S.; Rappaport, E.B.; Salam, M.T.; Linn, W.S.; Avol, E.L.; Zhang, Y.; Gilliland, F.D. Exhaled nitric oxide, susceptibility and new-onset asthma in the Children’s Health Study. Eur. Respir J. 2011, 37, 523–531. [Google Scholar] [CrossRef] [Green Version]
  13. di Palmo, E.; Cantarelli, E.; Catelli, A.; Ricci, G.; Gallucci, M.; Miniaci, A.; Pession, A. The Predictive Role of Biomarkers and Genetics in Childhood Asthma Exacerbations. Int. J. Mol. Sci. 2021, 22, 4651. [Google Scholar] [CrossRef] [PubMed]
  14. Anderson, J.T.; Zeng, M.; Li, Q.; Stapley, R.; Moore, D.R., 2nd; Chenna, B.; Fineberg, N.; Zmijewski, J.; Eltoum, I.E.; Siegal, G.P.; et al. Elevated levels of NO are localized to distal airways in asthma. Free Radic. Biol. Med. 2011, 50, 1679–1688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Fischer, A.; Folkerts, G.; Geppetti, P.; Groneberg, D.A. Mediators of asthma: Nitric oxide. Pulm. Pharmacol. Ther. 2002, 15, 73–81. [Google Scholar] [CrossRef]
  16. Ricciardolo, F.L.M.; Sterk, P.J.; Gaston, B.; Folkerts, G. Nitric oxide in health and disease of the respiratory system. Physiol. Rev. 2004, 84, 731–765. [Google Scholar] [CrossRef] [PubMed]
  17. Salam, M.T.; Bastain, T.M.; Rappaport, E.B.; Islam, T.; Berhane, K.; Gauderman, W.J.; Gilliland, F.D. Genetic variations in nitric oxide synthase and arginase influence exhaled nitric oxide levels in children. Allergy 2010, 66, 412–419. [Google Scholar] [CrossRef] [Green Version]
  18. Szczepankiewicz, A.; Sobkowiak, P.; Rachel, M.; Breborowicz, A.; Schoneich, N.; Bruce, K.; Kycler, Z.; Wojsyk-Banaszak, I.; Dmitrzak-Weglarz, M. Multilocus Analysis of Candidate Genes Involved in Neurogenic Inflammation in Pediatric Asthma and Related Phenotypes: A Case–Control Study. J. Asthma 2012, 49, 329–335. [Google Scholar] [CrossRef]
  19. Miller, S.A.; Dykes, D.D.; Polesky, H.F. A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res. 1988, 16, 1215. [Google Scholar] [CrossRef] [Green Version]
  20. Ritchie, M.D.; Hahn, L.W.; Roodi, N.; Bailey, L.R.; Dupont, W.D.; Parl, F.F.; Moore, J.H. Multifactor-Dimensionality Reduction Reveals High-Order Interactions among Estrogen-Metabolism Genes in Sporadic Breast Cancer. Am. J. Hum. Genet. 2001, 69, 138–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Wigner, P.; Czarny, P.; Synowiec, E.; Bijak, M.; Białek, K.; Talarowska, M.; Galecki, P.; Szemraj, J.; Sliwinski, T. Variation of genes involved in oxidative and nitrosative stresses in depression. Eur. Psychiatry 2018, 48, 38–48. [Google Scholar] [CrossRef]
  22. Yoo, K.H.; Kim, S.K.; Chung, J.-H.; Chang, S.-G. Nitric oxide synthase 2 gene polymorphisms are associated with prostatic volume in Korean men with benign prostatic hyperplasia. Asian J. Androl. 2010, 12, 690–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Chen, F.; Li, Y.-M.; Yang, L.-Q.; Zhong, C.-G.; Zhuang, Z.-X. Association of NOS2 and NOS3 gene polymorphisms with susceptibility to type 2 diabetes mellitus and diabetic nephropathy in the Chinese Han population. IUBMB Life 2016, 68, 516–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Yan, L.; Guo, W.; Wu, S.; Liu, J.; Zhang, S.; Shi, L.; Ji, G.; Gu, A. Genetic variants in nitric oxide synthase genes and the risk of male infertility in a Chinese population: A case-control study. PLoS ONE 2014, 9, e115190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Synowiec, E.; Wigner, P.; Cichon, N.; Watala, C.; Czarny, P.; Saluk-Bijak, J.; Miller, E.; Sliwinski, T.; Zielinska-Nowak, E.; Bijak, M. Single-Nucleotide Polymorphisms in Oxidative Stress-Related Genes and the Risk of a Stroke in a Polish Population-A Preliminary Study. Brain Sci. 2021, 11, 391. [Google Scholar] [CrossRef] [PubMed]
  26. Bouzigon, E.; Monier, F.; Boussaha, M.; Le Moual, N.; Huyvaert, H.; Matran, R.; Letort, S.; Bousquet, J.; Pin, I.; Lathrop, M.; et al. Associations between Nitric Oxide Synthase Genes and Exhaled NO-Related Phenotypes according to Asthma Status. PLOS ONE 2012, 7, e36672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Sato, S.; Wang, X.; Saito, J.; Fukuhara, A.; Uematsu, M.; Suzuki, Y.; Sato, Y.; Misa, K.; Nikaido, T.; Fukuhara, N.; et al. Exhaled nitric oxide and inducible nitric oxide synthase gene polymorphism in Japanese asthmatics. Allergol. Int. 2016, 65, 300–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Dahgam, S.; Nyberg, F.; Modig, L.; Naluai, Å.T.; Olin, A.-C. Single nucleotide polymorphisms in theNOS2andNOS3genes are associated with exhaled nitric oxide. J. Med. Genet. 2012, 49, 200–205. [Google Scholar] [CrossRef]
  29. Sample, P.J.; Wang, B.; Reid, D.W.; Presnyak, V.; McFadyen, I.J.; Morris, D.R.; Seelig, G. Human 5’ UTR design and variant effect prediction from a massively parallel translation assay. Nat. Biotechnol. 2019, 37, 803–809. [Google Scholar] [CrossRef]
  30. Wechsler, M.E.; Grasemann, H.; Deykin, A.; Silverman, E.K.; Yandava, C.N.; Israel, E.; Wand, M.; Drazen, J.M. Exhaled nitric oxide in patients with asthma: Association with NOS1 genotype. Am. J. Respir. Crit. Care Med. 2000, 162, 2043–2047. [Google Scholar] [CrossRef] [PubMed]
  31. Grasemann, H.; Gravesande, K.S.V.; Büscher, R.; Drazen, J.M.; Ratjen, F. Effects of Sex and of Gene Variants in Constitutive Nitric Oxide Synthases on Exhaled Nitric Oxide. Am. J. Respir. Crit. Care Med. 2003, 167, 1113–1116. [Google Scholar] [CrossRef] [PubMed]
  32. Ali, M.; Khoo, S.-K.; Turner, S.; Stick, S.; Le Souëf, P.; Franklin, P. NOS1 polymorphism is associated with atopy but not exhaled nitric oxide levels in healthy children. Pediatr. Allergy Immunol. 2003, 14, 261–265. [Google Scholar] [CrossRef]
  33. Holla, L.I.; Schuller, M.; Buckova, D.; Vacha, J. Neuronal nitric oxide synthase gene polymorphism and IgE-mediated allergy in the Central European population. Allergy 2004, 59, 548–552. [Google Scholar] [CrossRef]
  34. Leung, T.F.; Liu, E.K.H.; Tang, N.L.S.; Ko, F.W.S.; Li, C.Y.; Lam, C.W.K.; Wong, G.W.K. Nitric oxide synthase polymorphisms and asthma phenotypes in Chinese children. Clin. Exp. Allergy 2005, 35, 1288–1294. [Google Scholar] [CrossRef]
  35. Martínez, B.; Barrios, K.; Vergara, C.; Mercado, D.; Jiménez, S.; Gusmão, L.; Caraballo, L. A NOS1 Gene Polymorphism Associated with Asthma and Specific Immunoglobulin E Response to Mite Allergens in a Colombian Population. Int. Arch. Allergy Immunol. 2007, 144, 105–113. [Google Scholar] [CrossRef] [PubMed]
  36. Duckworth, L.; Hsu, L.; Feng, H.; Wang, J.; Sylvester, J.E.; Kissoon, N.; Sandler, E.; Lima, J.J. Physician-diagnosed asthma and acute chest syndrome: Associations with NOS Polymorphisms. Pediatr. Pulmonol. 2007, 42, 332–338. [Google Scholar] [CrossRef]
  37. Diamant, Z.; Vijverberg, S.; Alving, K.; Bakirtas, A.; Bjermer, L.; Custovic, A.; Dahlen, S.; Gaga, M.; Van Wijk, R.G.; Del Giacco, S.; et al. Toward clinically applicable biomarkers for asthma: An EAACI position paper. Allergy 2019, 74, 1835–1851. [Google Scholar] [CrossRef] [Green Version]
  38. Mogensen, I.; James, A.; Malinovschi, A. Systemic and breath biomarkers for asthma: An update. Curr. Opin. Allergy Clin. Immunol. 2020, 20, 71–79. [Google Scholar] [CrossRef]
  39. arożna, B.; Hoffmann, A.; Sobkowiak, P.; Schoneich, N.; Bręborowicz, A.; Szczepankiewicz, A. Polymorphisms in the interleukin 4, interleukin 4 receptor and interleukin 13 genes and allergic phenotype: A case control study. Adv. Med Sci. 2016, 61, 40–45. [Google Scholar] [CrossRef]
Table 1. The sequences of PCR primers and restriction enzymes used in genotyping analysis.
Table 1. The sequences of PCR primers and restriction enzymes used in genotyping analysis.
GeneSNPPrimer SequencesProduct
Size (bp)
Restriction EnzymeAlleles (bp)
NOS1C5266T
rs2682826
F: 5-ACTCCTTGAGTTTTCCTGCTGCGATG-3
R: 5-CCATGTTCCAGTGGTTTCATGCACAC-3
128Eco72IT: 128
C: 100, 28
NOS2-1659C/T
rs10459953
F: 5′-TGCTGCAGGTATAGCCAGAAT-3′
R: 5′-ATGGAGGGATGGTATGGTGCTGAT-3′
159FspBIC: 160
T: 129, 31
608C/T
rs2297518
F: 5′-ATCCCCTGAACCCAGACTTT-3′
R: 5′-GGCCAGGTTTCCAGAAGAA-3′
198TasIC: 175, 113
T: 142, 113
NOS3G894T
rs1799983
F: 5′-AAGGCAGGAGACAGTGGATG-3′
R: 5′-CAGTCAATCCCTTTGGTGCT-3′
246MboIG: 246
T: 158, 88
SNP—single nucleotide polymorphism; Set of F—forward and R—reverse primers sequences, bp—base pairs.
Table 2. Clinical description of analyzed population.
Table 2. Clinical description of analyzed population.
ParameterPatients Group
(n = 220)
Control Group
(n = 223)
Gender, male (%)59%48%
Age, years (mean ± SD)11.45 ± 3.5612.14 ± 3.31
Asthma severity -
Mild, n (%)29 (13.2)-
Moderate, n (%)121 (55.0)-
Severe, n (%)70 (31.8)-
Positive SPT (%)68.2-
FEV1/FVC pred (mean ± SD)88.5 ± 12.5104.35 ± 8.18
FEV1% pred (mean ± SD)85.3 ± 15.1106.15 ± 12.1
FVC% pred (mean ± SD)89.9 ± 15.1100.5 ± 10.9
PEF% pred (mean ± SD)81.3 ± 15.890.3 ± 12.9
FeNO (ppb) (mean ± SD)28.6 ± 34.316.6 ± 14.4
IgE (IU/mL) (mean ± SD)257.5 ± 326.7-
FEV1—forced expiratory volume in the first second, FVC—forced vital capacity, PEF—peak expiratory flow, FeNO—fraction of exhaled NO.
Table 3. The association analysis for analyzed polymorphisms between asthmatic patients and the control group.
Table 3. The association analysis for analyzed polymorphisms between asthmatic patients and the control group.
GenePolymorphismAsthma (%)Control
(%)
p Value
NOS1C5266T
rs2682826
GenotypesCC
CT
TT
57.3
37.3
5.45
58.6
33.3
8.1
0.659
AllelesC
T
24.1
75.9
24.7
75.3
0.912
NOS2-1659C/T
rs10459953
GenotypesCC
CT
TT
67.2
31.3
1.5
86.8
12.4
0.8
0.001 *
AllelesC
T
82.8
17.2
93.0
7.0
0.0006 *
608C/T
rs2297518
GenotypesCC
CT
TT
70.8
26.9
2.3
68.5
29.7
1.8
0.866
AllelesC
T
84.2
15.8
84.1
15.9
0.532
NOS3G894T
rs1799983
GenotypesGG
GT
TT
54.5
40.0
5.5
51.2
42.2
6.6
0.855
AllelesG
T
74.5
25.5
72.3
27.7
0.590
* Indicates significance.
Table 4. The results of multifactor dimensionality reduction analysis for epistatic interaction between analyzed variants in NOS genes.
Table 4. The results of multifactor dimensionality reduction analysis for epistatic interaction between analyzed variants in NOS genes.
ModelLoci CombinationTesting Balanced
Accuracy (%)
Cross-Validation Consistencyp-Value
Asthma
2-locusNOS2 (608) and NOS2 (C/T)62.380.025 *
3-locusNOS1, NOS2 (608) and NOS2 (C/T)58.460.201
4-locusNOS1, NOS3, NOS2 (608) and NOS2 (C/T)53.7100.640
Increased FeNO
2-locusNOS2 608 and NOS3 44.960.961
3-locusNOS1, NOS2 608 and NOS351.180.763
4-locusNOS1, NOS3, NOS2 (608) and NOS2 (C/T)55.7100.476
* Indicates significance.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nowakowska, J.; Sobkowiak, P.; Bręborowicz, A.; Mrówczyńska, M.; Wojsyk-Banaszak, I.; Szczepankiewicz, A. Nitric Oxide Synthase 2 Promoter Polymorphism Is a Risk Factor for Allergic Asthma in Children. Medicina 2021, 57, 1341. https://doi.org/10.3390/medicina57121341

AMA Style

Nowakowska J, Sobkowiak P, Bręborowicz A, Mrówczyńska M, Wojsyk-Banaszak I, Szczepankiewicz A. Nitric Oxide Synthase 2 Promoter Polymorphism Is a Risk Factor for Allergic Asthma in Children. Medicina. 2021; 57(12):1341. https://doi.org/10.3390/medicina57121341

Chicago/Turabian Style

Nowakowska, Joanna, Paulina Sobkowiak, Anna Bręborowicz, Magdalena Mrówczyńska, Irena Wojsyk-Banaszak, and Aleksandra Szczepankiewicz. 2021. "Nitric Oxide Synthase 2 Promoter Polymorphism Is a Risk Factor for Allergic Asthma in Children" Medicina 57, no. 12: 1341. https://doi.org/10.3390/medicina57121341

Article Metrics

Back to TopTop