Next Article in Journal
An Overview of Fruit Allergens: Structural, Functional, Phylogenetical, and Clinical Aspects
Previous Article in Journal
Is Switching of Adrenaline Auto Injector Devices a Concern for Anaphylaxis Management? A CROSS-Sectional Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic and Epigenetic Factors in Risk and Susceptibility for Childhood Asthma

by
Dimitrina Miteva
1,
Snezhina Lazova
2,3 and
Tsvetelina Velikova
4,*
1
Department of Genetics, Faculty of Biology, Sofia University St. Kliment Ohridski, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
2
Pediatric Department, University Hospital “N. I. Pirogov”, 21 “General Eduard I. Totleben” Blvd, 1606 Sofia, Bulgaria
3
Health Care Department, Faculty of Public Health, Medical University Sofia, Bialo More 8 Str., 1527 Sofia, Bulgaria
4
Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
*
Author to whom correspondence should be addressed.
Allergies 2023, 3(2), 115-133; https://doi.org/10.3390/allergies3020009
Submission received: 4 October 2022 / Revised: 19 December 2022 / Accepted: 29 May 2023 / Published: 14 June 2023
(This article belongs to the Section Asthma/Respiratory)

Abstract

:
Asthma is a common respiratory disease that affects people of all ages, characterized by considerable heterogeneity in age, clinical presentation, genetics, epigenetics, environmental factors, treatment response, and prognostic outcomes. Asthma affects more than 330 million people worldwide, of which 33% are children under 14 years, and 27% are adults whose first symptoms occurred in childhood. However, the genetic and epigenetic mechanisms of childhood allergic diseases and asthma are still not fully understood. Here, we conducted a biomedical narrative review of genes associated with the risk, severity, and susceptibility of childhood asthma since it differs from asthma in adults regarding their pathophysiology, development, and outcomes. We also systematized the available information on epigenetic changes associated with childhood asthma.

1. Introduction

Asthma is a common respiratory disease that affects people of all ages. It is characterized by considerable heterogeneity in age, clinical presentation, genetics, epigenetics, environmental factors, treatment response, and outcomes [1]. Asthma affects more than 330 million people worldwide [1], of which 33% are children under 14 years of age, 27% are adults who had their first symptoms in childhood [2] and 40% are adults [3].
The clinical syndrome of childhood asthma is characterized by common symptoms, including wheezing, shortness of breath, chest tightness, and cough, combined with variable airflow limitation and bronchial hyperreactivity [1]. In contrast, the most common symptoms in adults with asthma are coughing (especially at night, during exercise, etc.), difficulty breathing, chest tightness, shortness of breath, and wheezing [3].
With the accumulation of epidemiological, pathogenetic, and clinical data, it became clear that asthma is not a single disease but a syndrome. In addition, the symptoms of asthma are associated with different (inflammatory) subtypes. Therefore, predicting childhood asthma’s clinical course and evolution has important practical implications for patients, their parents, clinicians, and researchers [1]. Besides, the best way to study childhood-onset and adult-onset asthma is to look at them separately, as their pathophysiology, development, and outcomes may differ.
Early asthma studies used positional cloning techniques to look for risk or susceptibility genes in families. With the development of genome-wide association studies (GWAS), the discovery of new genetic biomarkers has become possible [4]. Advances in genome sequencing technology have allowed scientists to understand asthma’s complexity more clearly, and GWAS has become one of the preferred research methods. Over the past ten years, numerous alleles and loci have been identified as associated with asthma susceptibility, childhood asthma development, and asthma treatment response. Recent efforts to elucidate the mechanisms of asthma have focused on revealing epigenetics and environmental factors that influence gene expression without altering the DNA sequence at the molecular level [4].
Furthermore, asthma is common in families, determined by 55–74% genetic risk in adults [5,6] and almost 90% in children [7]. Higher estimates are typically found among boys than girls, with a ratio of up to 2:1, and in early-onset cases [8].
The rise in asthma cases worldwide over the past few decades clearly indicates that environmental factors are crucial for asthma development. Environmental risk factors for asthma are respiratory viral infections, tobacco smoke, animals (farm and domestic), microbial exposure, medications, air pollution, and other allergens [9,10,11,12,13]. In addition, all environmental exposures, especially those starting early in life, play a critical role in individual development and genetic responses and, thus, could lead to asthma [14].
We conducted a modified form of a narrative review, adhering to recent recommendations for writing a biomedical narrative review [15]. We searched through scientific databases and evidence from studies on childhood asthma regarding genetic and epigenetic factors. Initially, a comprehensive literature search was carried out in the bibliographic databases Medline (PubMed) and Scopus. Both MeSH (Medical Subject Headings) and relevant free-text terms were used, as follows: (“childhood asthma”) AND (“genetic factors”) AND (“epigenetics”) AND (“GWAS”). Our search was confined to articles published up to September 2022. Relevant data were also derived from other papers identified using the search engine Google Scholar. Finally, references of retrieved publications were further hand-searched for supplements.

2. Genome-Wide Association Studies (GWAS) on Childhood Asthma

The Collaborative Study on the Genetics of Asthma (CSGA), one of the first studies to identify the critical loci corresponding to asthma, was published in 1997 [16]. This paper is the first attempt to consider whether different genes regulate the heterogeneity of asthma among distinct ethnic groups. The study analyzed asthma cases in sibling pairs with asthma from 3 different racial groups (African Americans, Hispanics, and Caucasians) and identified six loci: 5p15 and 17p11.1—qll.2 in African Americans, 2q33 and 21q21 in Hispanics, and 11p15 and 19q13 in Caucasians [16].
In 2007, a GWAS study found that several single nucleotide polymorphisms (SNPs) on chromosome 17q21 were associated with childhood-onset asthma and early-onset asthma [17]. This research group analyzed more than 317,000 SNPs in DNA from 994 children with asthma and 1243 controls. In addition, the study found that genetic variants that regulate ORMDL3 gene expression are significant risk factors for childhood asthma [17].
In 2010, Sleiman et al. identified SNPs associated with childhood asthma in the previously established locus on 17q21 [18]. In addition, they found eight new SNPs related to childhood asthma at a novel locus on 1q31. One of them, rs2786098, was strongly associated with childhood asthma. The DENND1B gene in the 1q31 locus, expressed by dendric cells and natural killer cells, was also associated with susceptibility to asthma [18]. Other asthma studies provided information that the dipeptidyl peptidase 10 (DPP10) gene had particular relevance to the risk of childhood asthma [19]. In 2010, Mathias et al. published a GWAS on two independent populations of African ancestry (asthmatic cases vs. controls and asthmatic subjects vs. family members) [20]. The study provided evidence for three SNPs in genes of potential biologic relevance to allergy and asthma, including rs1435879, mapping to the DPP10 gene on chromosome 2q12.3-q14.2.
Other studies linked the CHI3L1 gene to a predisposition to asthma [21,22]. For example, the genotype and allele frequencies of rs4950928 of the CHI3L1 gene in the Chinese Han children population showed a significant difference between asthmatic children and controls. However, for the rs12141494 SNP, no significant differences were found between the two groups [22].
The SNP allele frequencies in 24,000 asthma patients from the Transnational Asthma Genetic Consortium were analyzed to identify 22 genes associated with asthma [23]. The study found that the locus with the highest risk was 17q21 in the GSDMB and ORMDL3 genes. In addition, the relationship of two SNPs, rs9273349 and rs9272346, in the HLA-DQ region was confirmed to relate to asthma [23]. These SNPs were found to be more strongly associated with asthma in children and early-onset asthma [17,24]. Ferreira et al. conducted a meta-analysis of European children, including 360,838 people (180,129 patients and 180,709 controls [25]. They detected 99 loci associated with asthma, atopic dermatitis/eczema, and hay fever. Two SNPs near IL18R1 and in the GSDMB gene (17q21) were associated with childhood asthma.
Ferreira et al. and Pividori et al. compared 9,433 child cases of asthma, 21,564 adult cases of asthma, and 318,237 controls from the UK Biobank (UKBB) [25,26]. Pividori et al. identified 61 independent asthma loci, of which 23 were specific to childhood asthma, 37 were shared between children and adults, and only one was specific for adults [26]. In addition, Ferreira et al. reported 25 new loci linked with childhood asthma in/near genes IRF4, NOD2, IL4R and IL2RA [27].
Hayden et al. surveyed 10,199 adult smokers from the COPD Gene Study [28]. The GWAS showed that the availability of SNPs for childhood asthma increased the risk of developing chronic obstructive pulmonary disease (COPD) later in life. The study identified a significant association of genes KIAA1958 (rs59289606), GSDMB, IL1RL1, IL13, LINC01149, and C11orf30-LRRC32 with asthma in children [28].
Other GWAS also confirm loci associated with genetic susceptibility to childhood asthma in ORMDL3/GSDMB (17q21), HLA-DQ (6p21), IL1RL1 (2q11) and IL18R1 (2q12), but also variants in IL33 (9p24), RAD50 (5q31) and CDHR3 (7q22) [17,29,30,31,32,33,34]. In addition, the ORMDL3 gene was associated with childhood-onset asthma, while the HLA-DQ gene was linked to asthma developed later in life. All these findings indicate that 38% of all cases of childhood asthma are the result of the combination of the identified genes [35].
A review published in 2019 summarized the GWAS and admixture mapping studies of asthma from the past two years [33]. Hernandez-Pacheco et al. identified new genes associated with asthma and confirmed previously identified ones. The authors also discussed the data on genetic variations of asthma susceptibility, asthma treatment response, and asthma predictive biomarkers. Moreover, they concluded that genetic information could be used to clarify the mechanisms of asthma development and evolution, to make a more accurate diagnosis and adequate treatment [33].
A search in PubMed and NHGRI-EBI GWAS Catalog revealed 15 GWAS studies involving asthma onset and related asthma traits, analyzed by MacArthur et al. [36]. Four of them concerned children [37,38,39,40], five adults [41,42,43,44,45], and another five studies identified genetic markers shared between children and adults [23,25,46,47,48]. Four-hundred fifty-one genetic variants (SNPs and short deletions/insertions) were associated with asthma and related traits by the GWAS.
A study by Schieck et al. on children with asthma showed that the disease affects more boys than girls [37]. Polymorphisms in the DMRT1 gene were associated with the sex of asthmatic children, assessed in 703 children with asthma, where 65.3% of the participations were males and 658 non-asthmatic controls, where 49.7% were males [37]. DMRT1 may play a role in developing sex-specific patterns of childhood asthma, but the mechanisms for these sex-specific differences are not yet elucidated.
More studies on childhood asthma included multi-ethnic asthma populations, but the presentation of non-European populations is not yet sufficient [49,50]. Most GWAS of childhood asthma have identified several risk alleles and loci, but most have been conducted on European descent. One recent study aimed to identify asthma susceptibility loci in Korean children [51]. They included 741 children with persistent asthma, 589 healthy children, and 551 healthy adults as controls. The results were validated using UK Biobank data. This GWAS identified an association between the TNFSF15 gene and childhood asthma in Korean children. The four identified variants of this gene were also associated with childhood asthma in the UK Biobank replication sample with the same effect [51].
All discussed genes identified by GWAS and important for childhood asthma are summarized in Table 1 [17,18,19,21,22,23,25,28,29,30,31,35,37,51]. Genes for childhood asthma severity or/and drug response [42,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82] are presented in Table 2.

3. Epigenetic and Childhood Asthma Development

In recent years, efforts to understand the etiology of childhood asthma have focused not only on genetics but also on epigenetics. Epigenetics is the inherited features that affect gene expression without changing the DNA sequence [83]. Epigenetic markers can be inherited by the next generation [84]. Epigenetic modifications may occur during prenatal development, childhood development, and adolescence. During these periods of life, people are most susceptible to various asthma-triggering factors [85]. Prenatal external environmental factors such as cigarette smoke, drugs, and alcohol and other postnatal factors such as air pollution, radiation, insufficient sleep, smoking, and improper diet may be the triggering factors for epigenetic changes [86,87]. We summarized some of these factors in Figure 1.
Obesity and childhood asthma are connected. Gene-environment interactions also influence both conditions. Because childhood obesity and asthma are significant pediatric health problems worldwide, they are included in the WHO Global NCD Action Plan 2023–2030 and the United Nations 2030 Agenda for Sustainable Development [86]. Furthermore, in recent years, obesity has been recognized as an independent risk factor for asthma development [88,89,90]. Obesity is associated with increased risk and morbidity in children with asthma compared to children with normal weight [91,92] (Table 3).
Moreover, in addition to the environmental factors, different patterns of epigenetic modifications have been identified associated with asthma, allergies, atopic dermatitis, and airway inflammation. Epigenetic regulation occurs through three main mechanisms: DNA methylation, histone modifications, and non-coding RNAs (microRNAs).

3.1. DNA Methylation and Studies of DNA Methylation and Childhood Asthma

The best-described mechanism in the literature is CpG-DNA methylation [85]. DNA methyltransferases added a methyl group to cytosine in a CpG dinucleotide, converting cytosine to 5’-methylcytosine. Over 80% of CpG dinucleotide sites are located outside of CpG islands [93]. Methylation of CpG islands near the promoters leads to transcription repression, while hypomethylation leads to genomic instability, upregulation of gene expression, and elevated mutation rates [94].
The associations between DNA methylation and diseases can be identified by an epigenome-wide association study (EWAS). In EWAS, where thousands of CpGs are being evaluated, multiple testing issues should be addressed to reduce false-positive findings [95].
EWAS are primarily cross-sectional studies and cannot differentiate whether epigenetic changes precede the disease or are a consequence of it. According to available data on childhood asthma, blood-based EWAS provides information mainly at the eosinophilic level. However, nasal epithelium EWAS provides information on methylation changes and regulatory perturbations of epithelial cells in the respiratory tract [96]. In addition, nasal epithelium EWAS was more reproducible in other cohorts than EWAS conducted in blood samples. All these data suggest that the methylation is tissue- and cell-specific [97]. Therefore, several EWAS studies and projects have attempted to describe the DNA methylation of different cell types and tissues of asthma patients [85,94,96,98].
A significant overlap of methylation changes was found in nasal and bronchial epithelial cells of children with asthma [96]. When conducting blood-based EWAS and nasal epithelium EWAS, genes such as ACOT7, EPX, GJA4, and METTL were found to be common [99]. This indicates that, except for the cell specificity of CpG methylation sites, there is also an epigenetic effect across tissues [100].
Recent studies by Xu [101] and Qi [102] focused on epigenetics and childhood asthma development in detail. The studies described gene/genetic variants associated with childhood asthma and epigenetic modifications connected with asthma development. All data discussed above [100,101,103,104,105,106,107,108,109,110,111] are systematized in Table 3 (modified from Qi et al., 2019).
Table 3. Summary of the major childhood asthma DNA methylation studies.
Table 3. Summary of the major childhood asthma DNA methylation studies.
Gene/Candidate-Gene *AncestryTissueRef.
LMAN2; STX3; LPIN1; DICER1; SLC25A25;EuropeanWhole bloodXu et al., 2018 [101]
ACOT7, EPX, GJA4 and METTL1;American AfricanNasal brush epithelial cellsYang et al., 2018 [100]
ZFPM1; AP2A2; IL5RA;EuropeanPeripheral bloodArathimos et al., 2017 [103]
CDHR3; CDH26; FBXL7;Hispanic/LatinoNasal brushes cellsForno et al., 2018 [104]
EVL; NTRK1; SLC9A3; ACOT7;Mixed ancestryNasal swab cellsCardenas et al., 2019 [105]
CLNS1A, Mir_548; SUB1, LOC100129858; RUNX1; GPATCH2; WDR20; IL5RA; ACOT7; KCNH2;-13 European cohorts;
-3 mixed ancestry cohorts
-1 African/American cohort
Whole bloodReese et al., 2019 [106]
ORMDL3; CCL26African American and European AmericanBronchial airway epithelial cellsNicodemus-Johnson et al., 2016 [107]
SMAD3; DDO/METTL24EuropeanWhole bloodLund et al., 2018 [108]
CCL5, IL2RA, TBX21, FCER2, TGFB1, LIF, ADAM17, NHEJ1, AHR, PGDR, PI3K, GNA12 etc.American cohortPeripheral bloodRastogi et al., 2013 [109]
ARG1, ARG2, IL6, iNOSnon-Hispanic white and Hispanic white ethnicityBuccal cells/Nasal epitheliumBreton et al., 2011 [110]
KRT5, CRIP1, STAT5AEuropeanBronchial epitheliumStefanowicz et al., 2012 [111]
* Some genes were identified in more than one study.

3.2. Histone Modification and Studies of Histone Modification and Asthma

DNA condensation occurs with the help of core histones (H2A, H2B, H3, and H4) to form a chromatin structure [112]. Histone modifications are methylation, acetylation, phosphorylation, ubiquitination, sumoylation, and ADP-ribosylation. Disturbance in histone modifications has many clinical implications, leading to inflammatory responses by the immune system and the development of various diseases, including allergic asthma, rhinitis, atopic dermatitis, and allergies (Figure 2).
Histone modifications alter chromatin accessibility. For example, histone acetylation is carried out by the enzyme histone acetyltransferase (HAT), leading to a loose chromatin structure. This makes it available to transcription factors and activates gene expression. On the other hand, histone deacetylation is carried out by the enzyme histone deacetylase (HDAC), resulting in gene silencing and a lack of gene expression. These histone modifications are associated with the risk of asthma development or disease severity (in children and adults). They affect the gene expression of genes responsible for the differentiation and maturation of various immune cells, some of which are involved in asthma development [113].
HAT activity has been increased in adults [114] and children with asthma [115], and the ratio of HAT/HDAC changes also depending on the presence or/and severity of asthma.
To date, no publications discuss in detail the role of histone modifications in the development of asthma in children. Some studies have been conducted in animal models [116,117,118,119]. The results of other studies established the role of HDACs in T-cell development and asthma susceptibility. Their suppressed expression can lead to asthma and/or allergic airway diseases with varying severity, as well as resistance to corticosteroid therapy [114,120]. In HDAC1-deficient T-cell mice, there is an increased number of eosinophils and the production of Th2 cytokines, which is a prerequisite for asthma development [121]. These findings indicate that HDACs are essential in repairing airway epithelium, especially in airway remodeling observed in severe asthma [122].
A genome-wide histone modification study from 12 asthma patients and 12 controls identified 200 enhancer regions in three immune cell subpopulations (naive T cells, Th1 and Th2 cells). In addition, a selective enrichment in demethylation of the Lys at the fourth position of the N-terminal tail of histone 3 (H3K4me2) was shown. This marker indicates gene activation in promoters and enhancers of genes [123]. One hundred sixty-three regions were specific for the Th2 cells, 29 were specific for naïve T cells, and only 11 for Th1 cells.
A study of children with allergic asthma (n = 14) and controls (n = 18) identified a high level of histone acetylation (histone 3 at the IL13 and FOXP3 loci). The study also indicates a potential regulatory role of IL13 acetylation on the amount of protein product [124].
Similarly, acetylation of histone H3 lysine 18 (H3K18) is related to increased expression of STAT6, EGFR, and ΔNp63 [125]. The expression of epidermal growth factor receptor (EGFR) is vital for the proliferation, differentiation, and repair processes. It was increased in both healthy and damaged areas of the epithelium of pediatric and adult patients with asthma [125,126,127,128]. In addition, the signal transducer and activator of transcription 6 (STAT6) are also overexpressed in the epithelium of patients with severe asthma [129,130].
In a recent study, loss of HAT was found to be associated with a downregulation of the ORMDL3 gene, which in turn led to the development of childhood asthma [131].
Some other studies have also identified a link between childhood asthma and histone modifications. For example, Wawrzyniak et al. found higher expression of histone deacetylases 1 and 9 in epithelial cells from 18 asthmatic patients and 9 controls [132]. In addition, increased expression of HDACs led to increased expression of IL-13 and IL-4, which are critical genes for childhood asthma severity and treatment response.
It is well-known that prenatal and/or childhood exposure to environmental factors can influence the immune system and responses [133,134]. In addition, environmental factors can strongly induce epigenetic changes in immune cells, leading to the development of allergies/asthma [135,136].
In 2011, Brand et al. investigated epigenetic alteration in pregnant mice and offspring [137]. They demonstrated that changing H4 acetylation led to a childhood asthma phenotype.
Another research group evaluated the acetylation of histones H3 and H4 in the promoter regions of six immunoregulatory genes associated with childhood allergy in 173 placentas [138]. Three genes were found to be associated with high histone acetylation levels and risk of allergic sensitization—H3 acetylation in placentas at the IFNG and SH2B3 genes and H4 acetylation at HDAC4. These data suggest that placental histone acetylation levels are associated with a reduced risk of allergen sensitization and have the potential to predict the development of childhood asthma/allergies.
Inhibition of HDAC activity has a preventive effect on epithelial barrier integrity in pediatric and adult patients with asthma [132].
To sum up, there is no doubt that histone modifications play a crucial role in the development and pathogenesis of asthma. However, more studies with larger cohorts are needed to obtain more accurate and transparent information about their contribution to immune-mediated and allergic diseases, such as asthma. Moreover, maternal asthma remains among the highest risk factors for childhood-onset asthma. Data show that epigenetic alterations may occur in the embryo of pregnant asthmatic women that may lead to severe asthma development in the offspring.
A summary of the studies on histone modification and childhood asthma [124,125,131,132,138] are presented in Table 4.

3.3. Non-Coding RNAs and Studies of Non-Coding RNA and Asthma

Non-coding RNAs, such as microRNAs (miRNAs, miRs), are single-strand RNAs of 21–25 nucleotides that regulate gene expression at the post-transcriptional level [139]. They act as dynamic regulators of gene networks by exercising control over the translation of many mRNAs by mRNA destabilization [140]. miRNAs play a significant role in biochemical pathways, cell signaling, cell and tissue development, and regulating pathological processes in various diseases. They have been identified as related to childhood/adult asthma, expressed in different tissues, including blood cells, airway epithelial cells, and smooth muscle. They can also serve as potential biomarkers for diagnosis and therapy targets for asthma [141]. Differences in miRNA expression between asthmatic and healthy subjects were found [98]. Some asthma-related miRNAs may act on genes associated with respiratory tract function (i.e., miR-10, miR-140-3p, miR-708), and others may target genes related to immune system function (i.e., miR-21, miR-19a, miR-155, miR-210, miR-125b, miR-223) [142].
MiRNAs, such as miRNA-17-18-19-20, miRNA-26a/b, miRNA-27a/b, miRNA-125b, miRNA-155, and others, polarize macrophages as a reaction to specific environmental stimuli and signals toward an M1-phenotype (classically activated). Others as miRNA-21, miRNA-124, miRNA-223-3p, and miRNA-511-3p, e.g., polarize macrophages towards an M2-phenotype (alternatively activated) [143].
In a recent study, two miRNAs (miR-21 and miR-126) were found to be upstream-regulated in patients with asthma compared to controls [144]. Another study showed that miR-21 contributes to the production of eosinophils and proliferation [145]. MiRNA-21 is one of the most well-studied miRNAs in relation to asthma. In asthmatic patients, its high amount suppresses IL-12p35 and causes resistance to steroids [146]. IL-12p35 is predominantly produced by dendritic cells, monocytes, and macrophages and inhibits cytokine-induced STAT pathways and cell-cycle regulators. Serum miRNA-21 is related to asthma onset or allergies and could be used as a biomarker [146,147]. Both miRNA-21 and miRNA-146a were associated with increased levels of eosinophils and could be biomarkers for the eosinophilic endotype of asthma [148].
Another study identified increased levels of miRNA-155 and decreased levels of Let-7a in the blood of children with asthma (n = 100) compared to controls (n = 100) [149]. To distinguish the severity of the disease, different expression profiles of miRNA-155 and Let-7a were found [149]. MiRNA-155 plays a critical role in IL-33 regulation of airway inflammation in mouse models of asthma [150]. In addition, the Let-7 miRNA has been shown to target IL13 and exert anti-inflammatory effects in mice [151]. These data suggest that miRNA-155 could be one of the biomarkers suitable for childhood asthma diagnosis.
Another miRNA, miRNA-146, has also been studied in relation to asthma [152]. HLA-G variation in children also increased the risk of developing asthma, along with miRNA-152 [108]. In an examination of serum samples from 160 children in the Childhood Asthma Management Program (CAMP), 155 circulating miRNAs were detected [153]. Of these, eight were significantly related to PC20 (asthma test to determine the concentration of inhaled methacholine), but the most substantial relationship was with miRNA-296-5p. Two others, miRNA-30d-5p, and miRNA-16-5p, were found to be critical in the development of asthma via regulating airway smooth muscles [153].
MiRNAs may also be used as biomarkers to assess the risk of childhood asthma development or to predict the outcomes from treatment, as several studies showed [142,154,155,156]. All these findings demonstrated the importance of miRNAs in controlling inflammation, diagnosing disease severity, and treatment response in childhood asthma, as well as their potential use as disease biomarkers.
The miRNA studies associated with asthma [157,158,159,160,161,162,163,164] are summarized in Table 5.
Recently, our team also discussed the miRNA involvement in allergic rhinitis and mild asthma in adults as a part of united airway disease [165].
Our review paper has some limitations. First, we focused mainly on the GWAS outcomes related to the development and severe course of childhood asthma. We acknowledge that there are also numerous genes associated with asthma, including genes related to external risk factors and medications, responses to therapy, etc. Secondly, most of the cited studies have other limitations, such as a limited number of patients, some limitations in the design and methodology, or their failure to adjust the impact of different factors (i.e., environmental, etc.). Furthermore, new data is expected to be published since childhood asthma and overall asthma is a highly researched topic. We believe that this limitation is not fatal but a prospect for other reviews.

4. Conclusions

Recent efforts to elucidate the mechanisms of asthma have focused on elucidating epigenetics and environmental factors that influence gene expression without altering the DNA sequence at the molecular level. This biomedical narrative review summarized the available information on genes associated with risk and susceptibility to childhood asthma development. We also covered some of the epigenetic modifications associated with the development of children’s asthma.
The accumulated data from GWAS on the numerous alleles and loci associated with allergies and asthma in children and adults made it possible to discover new genetic biomarkers for asthma that can be implemented in clinical practice.
Although histone modifications contribute to the development of childhood asthma, there is still a small number of studies in this area. Improving our knowledge in this area may lead to the discovery of effective diagnostic tools, therapeutic targets, and drugs for different types of asthma.

Author Contributions

Conceptualization, D.M. and T.V.; methodology, D.M.; software, D.M.; validation, D.M., S.L. and T.V.; formal analysis, S.L.; investigation, D.M.; resources, D.M.; data curation, T.V.; writing—original draft preparation, D.M.; writing—review and editing, T.V.; visualization, D.M.; supervision, T.V.; project administration, T.V.; funding acquisition, T.V. All authors have read and agreed to the published version of the manuscript.

Funding

This study is financed by the European Union-NextGenerationEU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, project № BG-RRP-2.004-0008-C01.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Global Asthma Report 2018, Global Asthma Network. Available online: http://www.globalasthmanetwork.org (accessed on 20 July 2022).
  2. Rhodes, L.; Moorman, J.E.; Redd, S.C. Sex Differences in Asthma Prevalence and Other Disease Characteristics in Eight States. J. Asthma 2005, 42, 777–782. [Google Scholar] [CrossRef] [PubMed]
  3. Morales, E.; Duffy, D. Genetics and Gene-Environment Interactions in Childhood and Adult Onset Asthma. Front. Pediatr. 2019, 7, 499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Han, Y.; Jia, Q.; Jahani, P.S.; Hurrell, B.P.; Pan, C.; Huang, P.; Gukasyan, J.; Woodward, N.C.; Eskin, E.; Gilliland, F.D.; et al. Genome-wide analysis highlights contribution of immune system pathways to the genetic architecture of asthma. Nat. Commun. 2020, 11, 1776. [Google Scholar] [CrossRef] [Green Version]
  5. Thomsen, S.F.; Duffy, D.L.; Kyvik, K.O.; Backer, V. Genetic influence on the age at onset of asthma: A twin study. J. Allergy Clin. Immunol. 2010, 126, 626–630. [Google Scholar] [CrossRef] [PubMed]
  6. Polderman, T.J.C.; Benyamin, B.; de Leeuw, C.A.; Sullivan, P.F.; Van Bochoven, A.; Visscher, P.M.; Posthuma, D. Meta-analysis of the heritability of human traits based on fifty years of twin studies. Nat. Genet. 2015, 47, 702–709. [Google Scholar] [CrossRef] [Green Version]
  7. Ullemar, V.; Magnusson, P.K.E.; Lundholm, C.; Zettergren, A.; Melén, E.; Lichtenstein, P.; Almqvist, C. Heritability and confirmation of genetic association studies for childhood asthma in twins. Allergy 2016, 71, 230–238. [Google Scholar] [CrossRef] [Green Version]
  8. Almqvist, C.; Worm, M.; Leynaert, B. Working group of GA2LEN WP 2.5 Gender. Impact of gender on asthma in childhood and adolescence: A GA2LEN review. Allergy 2008, 63, 47–57. [Google Scholar] [CrossRef]
  9. Beasley, R.; Semprini, A.; Mitchell, E.A. Risk factors for asthma: Is prevention possible? Lancet 2015, 386, 1075–1085. [Google Scholar] [CrossRef]
  10. West, C.E.; Renz, H.; Jenmalm, M.C.; Kozyrskyj, A.L.; Allen, K.J.; Vuillermin, P.; Prescott, S.L.; in-FLAME Microbiome Interest Group. The gut microbiota and inflammatory noncommunicable diseases: Associations and potentials for gut microbiota therapies. J. Allergy Clin. Immunol. 2015, 135, 3–13. [Google Scholar] [CrossRef] [Green Version]
  11. Bisgaard, H.; Li, N.; Bonnelykke, K.; Chawes, B.L.K.; Skov, T.; Paludan-Müller, G.; Stokholm, J.; Smith, B.; Krogfelt, K.A. Reduced diversity of the intestinal microbiota during infancy is associated with increased risk of allergic disease at school age. J. Allergy Clin. Immunol. 2011, 128, 646–652.e5. [Google Scholar] [CrossRef]
  12. Hylkema, M.N.; Blacquière, M.J. Intrauterine Effects of Maternal Smoking on Sensitization, Asthma, and Chronic Obstructive Pulmonary Disease. Proc. Am. Thorac. Soc. 2009, 6, 660–662. [Google Scholar] [CrossRef]
  13. Gehring, U.; Gruzieva, O.; Agius, R.M.; Beelen, R.; Custovic, A.; Cyrys, J.; Eeftens, M.; Flexeder, C.; Fuertes, E.; Heinrich, J.; et al. Air Pollution Exposure and Lung Function in Children: The ESCAPE Project. Environ. Health Perspect. 2013, 121, 1357–1364. [Google Scholar] [CrossRef] [Green Version]
  14. Bønnelykke, K.; Ober, C. Leveraging gene-environment interactions and endotypes for asthma gene discovery. J. Allergy Clin. Immunol. 2016, 137, 667–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Gasparyan, A.Y.; Ayvazyan, L.; Blackmore, H.; Kitas, G.D. Writing a narrative biomedical review: Considerations for authors, peer reviewers, and editors. Rheumatol. Int. 2011, 31, 1409–1417. [Google Scholar] [CrossRef] [PubMed]
  16. Banks-Schlegle, S. A genome-wide search for asthma susceptibility loci in ethnically diverse populations. Nat. Genet. 1997, 15, 389–392. [Google Scholar] [CrossRef]
  17. Moffatt, M.; Kabesch, M.; Liang, L.; Dixon, A.L.; Strachan, D.; Heath, S.; Depner, M.; von Berg, A.; Bufe, A.; Rietschel, E.; et al. Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature 2007, 448, 470–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Sleiman, P.M.; Flory, J.; Imielinski, M.; Bradfield, J.P.; Annaiah, K.; Willis-Owen, S.A.; Wang, K.; Rafaels, N.M.; Michel, S.; Bonnelykke, K.; et al. Variants of DENND1B associated with asthma in children. N. Engl. J. Med. 2010, 362, 36–44. [Google Scholar] [CrossRef]
  19. Allen, M.; Heinzmann, A.; Noguchi, E.; Abecasis, G.; Broxholme, J.; Ponting, C.; Bhattacharyya, S.; Tinsley, J.; Zhang, Y.; Holt, R.; et al. Positional cloning of a novel gene influencing asthma from Chromosome 2q14. Nat. Genet. 2003, 35, 258–263. [Google Scholar] [CrossRef]
  20. Mathias, R.A.; Grant, A.V.; Rafaels, N.; Hand, T.; Gao, L.; Vergara, C.; Tsai, Y.J.; Yang, M.; Campbell, M.; Foster, C.; et al. A genome-wide association study on African-ancestry populations for asthma. J. Allergy Clin. Immunol. 2010, 125, 336–346.e4. [Google Scholar] [CrossRef] [Green Version]
  21. Ober, C.; Tan, Z.; Sun, Y.; Possick, J.D.; Pan, L.; Nicolae, R.; Radford, S.; Parry, R.R.; Heinzmann, A.; Deichmann, K.A.; et al. Effect of variation in CHI3L1 on serum YKL-40 level, risk of asthma, and lung function. N. Engl. J. Med. 2008, 358, 1682–1691. [Google Scholar] [CrossRef] [Green Version]
  22. Chen, F.; An, Y.; Wang, J. CHI3L1 is correlated with childhood asthma. Int. J. Clin. Exp. Pathol. 2017, 10, 10559–10564. [Google Scholar]
  23. Demenais, F.; Margaritte-Jeannin, P.; Barnes, K.C.; Cookson, W.O.C.; Altmüller, J.; Ang, W.; Barr, R.G.; Beaty, T.H.; Becker, A.B.; Beilby, J.; et al. Multiancestry association study identifies new asthma risk loci that colocalize with immune-cell enhancer marks. Nat. Genet. 2017, 50, 42–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Lasky-Su, J.; Himes, B.E.; Raby, B.A.; Klanderman, B.J.; Sylvia, J.S.; Lange, C.; Melen, E.; Martinez, F.D.; Israel, E.; Gauderman, J.; et al. HLA-DQ strikes again: Genome-wide association study further confirmsHLA-DQin the diagnosis of asthma among adults. Clin. Exp. Allergy 2012, 42, 1724–1733. [Google Scholar] [CrossRef] [PubMed]
  25. Ferreira, M.A.; Vonk, J.M.; Baurecht, H.; Marenholz, I.; Tian, C.; Hoffman, J.D.; Helmer, Q.; Tillander, A.; Ullemar, V.; van Dongen, J.; et al. Shared genetic origin of asthma, hay fever and eczema elucidates allergic disease biology. Nat. Genet. 2017, 49, 1752–1757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Pividori, M.; Schoettler, N.; Nicolae, D.L.; Ober, C.; Im, H.K. Shared and distinct genetic risk factors for childhood-onset and adult-onset asthma: Genome-wide and transcriptome-wide studies. Lancet Respir. Med. 2019, 7, 509–522. [Google Scholar] [CrossRef]
  27. Ferreira, M.A.; Mathur, R.; Vonk, J.M.; Szwajda, A.; Brumpton, B.; Granell, R.; Brew, B.K.; Ullemar, V.; Lu, Y.; Jiang, Y.; et al. Genetic Architectures of Childhood- and Adult-Onset Asthma Are Partly Distinct. Am. J. Hum. Genet. 2019, 104, 665–684. [Google Scholar] [CrossRef] [Green Version]
  28. Hayden, L.P. on behalf of the COPDGene Investigators; Cho, M.H.; Raby, B.A.; Beaty, T.H.; Silverman, E.K.; Hersh, C.P. Childhood asthma is associated with COPD and known asthma variants in COPDGene: A genome-wide association study. Respir. Res. 2018, 19, 209. [Google Scholar] [CrossRef] [Green Version]
  29. Moffatt, M.F.; Gut, I.G.; Demenais, F.; Strachan, D.P.; Bouzigon, E.; Heath, S.; von Mutius, E.; Farrall, M.; Lathrop, M.; Cookson, W.O.C.M.; et al. A large-scale, consortium-based genome-wide association study of asthma. N. Engl. J. Med. 2010, 363, 1211–1221. [Google Scholar] [CrossRef] [Green Version]
  30. Li, X.; Howard, T.D.; Zheng, S.L.; Haselkorn, T.; Peters, S.P.; Meyers, D.A.; Bleecker, E.R. Genome-wide association study of asthma identifies RAD50-IL13 and HLA-DR/DQ regions. J. Allergy Clin. Immunol. 2010, 125, 328–335.e11. [Google Scholar] [CrossRef] [Green Version]
  31. Bønnelykke, K.; Sleiman, P.; Nielsen, K.; Kreiner-Møller, E.; Mercader, J.M.; Belgrave, D.; Dekker, H.T.D.; Husby, A.; Sevelsted, A.; Faura-Tellez, G.; et al. A genome-wide association study identifies CDHR3 as a susceptibility locus for early childhood asthma with severe exacerbations. Nat. Genet. 2013, 46, 51–55. [Google Scholar] [CrossRef]
  32. Vicente, C.T.; Revez, J.A.; Ferreira, M.A.R. Lessons from ten years of genome-wide association studies of asthma. Clin. Transl. Immunol. 2017, 6, e165. [Google Scholar] [CrossRef] [Green Version]
  33. Hernandez-Pacheco, N.; Pino-Yanes, M.; Flores, C. Genomic Predictors of Asthma Phenotypes and Treatment Response. Front. Pediatr. 2019, 7, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Eliasen, A.U.; Pedersen, C.E.T.; Rasmussen, M.A.; Wang, N.; Soverini, M.; Fritz, A.; Stokholm, J.; Chawes, B.L.; Morin, A.; Bork-Jensen, J.; et al. Genome-wide study of early and severe childhood asthma identifies interaction between CDHR3 and GSDMB. J. Allergy Clin. Immunol. 2022, 150, 622–630. [Google Scholar] [CrossRef]
  35. Thomsen, S.F. Genetics of asthma: An introduction for the clinician. Eur. Clin. Respir. J. 2015, 2, 24643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. MacArthur, J.; Bowler, E.; Cerezo, M.; Gil, L.; Hall, P.; Hastings, E.; Junkins, H.; McMahon, A.; Milano, A.; Morales, J.; et al. The new NHGRI-EBI Catalog of published genome-wide association studies (GWAS Catalog). Nucleic Acids Res. 2016, 45, D896–D901. [Google Scholar] [CrossRef]
  37. Schieck, M.; Schouten, J.P.; Michel, S.; Suttner, K.; Toncheva, A.A.; Gaertner, V.D.; Illig, T.; Lipinski, S.; Franke, A.; Klintschar, M.; et al. Doublesex and mab-3 related transcription factor 1 (DMRT1) is a sex-specific genetic determinant of childhood-onset asthma and is expressed in testis and macrophages. J. Allergy Clin. Immunol. 2016, 138, 421–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Gref, A.; Merid, S.K.; Gruzieva, O.; Ballereau, S.; Becker, A.; Bellander, T.; Bergström, A.; Bossé, Y.; Bottai, M.; Chan-Yeung, M.; et al. Genome-Wide Interaction Analysis of Air Pollution Exposure and Childhood Asthma with Functional Follow-up. Am. J. Respir. Crit. Care Med. 2017, 195, 1373–1383. [Google Scholar] [CrossRef]
  39. Marques, C.R.; Costa, G.N.; da Silva, T.M.; Oliveira, P.; Cruz, A.A.; Alcantara-Neves, N.M.; Fiaccone, R.L.; Horta, B.L.; Hartwig, F.P.; Burchard, E.G.; et al. Suggestive association between variants in IL1RAPL and asthma symptoms in Latin American children. Eur. J. Hum. Genet. 2017, 25, 439–445. [Google Scholar] [CrossRef] [Green Version]
  40. Spear, M.L.; Hu, D.; Pino-Yanes, M.; Huntsman, S.; Eng, C.; Levin, A.M.; Ortega, V.E.; White, M.J.; McGarry, M.E.; Thakur, N.; et al. A genome-wide association and admixture mapping study of bronchodilator drug response in African Americans with asthma. Pharm. J. 2018, 19, 249–259. [Google Scholar] [CrossRef]
  41. Yatagai, Y.; Hirota, T.; Sakamoto, T.; Yamada, H.; Masuko, H.; Kaneko, Y.; Iijima, H.; Naito, T.; Noguchi, E.; Tamari, M.; et al. Variants near the HLA complex group 22 gene (HCG22) confer increased susceptibility to late-onset asthma in Japanese populations. J. Allergy Clin. Immunol. 2016, 138, 281–283.e13. [Google Scholar] [CrossRef] [Green Version]
  42. Almoguera, B.; Vazquez, L.; Mentch, F.; Connolly, J.; Pacheco, J.; Sundaresan, A.S.; Peissig, P.L.; Linneman, J.G.; McCarty, C.; Crosslin, D.; et al. Identification of Four Novel Loci in Asthma in European American and African American Populations. Am. J. Respir. Crit. Care Med. 2017, 195, 456–463. [Google Scholar] [CrossRef]
  43. Condreay, L.; Chiano, M.; Ortega, H.; Buchan, N.; Harris, E.; Bleecker, E.R.; Thompson, P.J.; Humbert, M.; Gibson, P.; Yancey, S.; et al. No genetic association detected with mepolizumab efficacy in severe asthma. Respir. Med. 2017, 132, 178–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Mosteller, M.; Hosking, L.; Murphy, K.; Shen, J.; Song, K.; Nelson, M.; Ghosh, S. No evidence of large genetic effects on steroid response in asthma patients. J. Allergy Clin. Immunol. 2017, 139, 797–803.e7. [Google Scholar] [CrossRef] [PubMed]
  45. Vonk, J.M.; Scholtens, S.; Postma, D.S.; Moffatt, M.F.; Jarvis, D.; Ramasamy, A.; Wjst, M.; Omenaas, E.R.; Bouzigon, E.; Demenais, F.; et al. Adult onset asthma and interaction between genes and active tobacco smoking: The GABRIEL consortium. PLoS ONE 2017, 12, e0172716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Murk, W.; DeWan, A.T. Genome-wide search identifies a gene-gene interaction between 20p13 and 2q14 in asthma. BMC Genet. 2016, 17, 102. [Google Scholar] [CrossRef] [Green Version]
  47. Nieuwenhuis, M.A.; Siedlinski, M.; van den Berge, M.; Granell, R.; Li, X.; Niens, M.; van der Vlies, P.; Altmüller, J.; Nürnberg, P.; Kerkhof, M.; et al. Combining genome-wide association study and lung eQTL analysis provides evidence for novel genes associated with asthma. Allergy 2016, 71, 1712–1720. [Google Scholar] [CrossRef] [Green Version]
  48. Yan, Q.; Brehm, J.; Pino-Yanes, M.; Forno, E.; Lin, J.; Oh, S.S.; Acosta-Perez, E.; Laurie, C.C.; Cloutier, M.M.; Raby, B.A.; et al. A meta-analysis of genome-wide association studies of asthma in Puerto Ricans. Eur. Respir. J. 2017, 49, 1601505. [Google Scholar] [CrossRef] [Green Version]
  49. Burchard, E.G.; Oh, S.S.; Foreman, M.G.; Celedon, J.C. Moving toward true inclusion of racial/ethnic minorities in federally funded studies. A key step for achieving respiratory health equality in the United States. Am. J. Respir. Crit. Care Med. 2015, 191, 514–521. [Google Scholar] [CrossRef] [Green Version]
  50. Popejoy, A.B.; Fullerton, S.M. Genomics is failing on diversity. Nature 2016, 538, 161–164. [Google Scholar] [CrossRef] [Green Version]
  51. Kim, K.W.; Kim, D.Y.; Yoon, D.; Kim, K.; Jang, H.; Schoettler, N.; Kim, E.G.; Na Kim, M.; Hong, J.Y.; Lee, J.; et al. Genome-wide association study identifies TNFSF15 associated with childhood asthma. Allergy 2021, 77, 218–229. [Google Scholar] [CrossRef]
  52. Israel, E.; Drazen, J.M.; Liggett, S.B.; Boushey, H.A.; Cherniack, R.M.; Chinchilli, V.M.; Cooper, D.M.; Fahy, J.V.; Fish, J.E.; Ford, J.G.; et al. The effect of polymorphisms of the beta(2)-adrenergic receptor on the response to regular use of albuterol in asthma. Am. J. Respir. Crit. Care Med. 2000, 162, 75–80. [Google Scholar] [CrossRef] [Green Version]
  53. Litonjua, A.; Gong, L.; Duan, Q.L.; Shin, J.; Moore, M.J.; Weiss, S.T.; Johnson, J.A.; Klein, T.E.; Altman, R.B. Very important pharmacogene summary ADRB2. Pharm. Genom. 2010, 20, 64–69. [Google Scholar] [CrossRef] [Green Version]
  54. Sood, N.; Connolly, J.J.; Mentch, F.D.; Vazquez, L.; Sleiman, P.M.; Hysinger, E.B.; Hakonarson, H. Leveraging electronic health records to assess the role of ADRB2 single nucleotide polymorphisms in predicting exacerbation frequency in asthma patients. Pharm. Genom. 2018, 28, 256–259. [Google Scholar] [CrossRef] [PubMed]
  55. Scaparrotta, A.; Franzago, M.; Marcovecchio, M.L.; Di Pillo, S.; Chiarelli, F.; Mohn, A.; Stuppia, L. Role of THRB, ARG1, and ADRB2 Genetic Variants on Bronchodilators Response in Asthmatic Children. J. Aerosol Med. Pulm. Drug Deliv. 2019, 32, 164–173. [Google Scholar] [CrossRef]
  56. Burchard, E.G.; Silverman, E.K.; Rosenwasser, L.J.; Borish, L.; Yandava, C.; Pillari, A.; Weiss, S.T.; Hasday, J.; Lilly, C.M.; Ford, J.G.; et al. Association Between a Sequence Variant in the IL-4 Gene Promoter and FEV1 in Asthma. Am. J. Respir. Crit. Care Med. 1999, 160, 919–922. [Google Scholar] [CrossRef] [PubMed]
  57. Drazen, J.M.; Yandava, C.N.; Dubé, L.; Szczerback, N.; Hippensteel, R.; Pillari, A.; Israel, E.; Schork, N.; Silverman, E.S.; Katz, D.A.; et al. Pharmacogenetic association between ALOX5 promoter genotype and the response to anti-asthma treatment. Nat. Genet. 1999, 22, 168–170. [Google Scholar] [CrossRef]
  58. Mougey, E.; Lang, J.E.; Allayee, H.; Teague, W.G.; Dozor, A.J.; Wise, R.A.; Lima, J.J. ALOX5Polymorphism associates with increased leukotriene production and reduced lung function and asthma control in children with poorly controlled asthma. Clin. Exp. Allergy 2012, 43, 512–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Tantisira, K.G.; Lake, S.; Silverman, E.S.; Palmer, L.; Lazarus, R.; Silverman, E.K.; Liggett, S.B.; Gelfand, E.W.; Rosenwasser, L.J.; Richter, B.; et al. Corticosteroid pharmacogenetics: Association of sequence variants in CRHR1 with improved lung function in asthmatics treated with inhaled corticosteroids. Hum. Mol. Genet. 2004, 13, 1353–1359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Choudhry, S.; Avila, P.C.; Nazario, S.; Ung, N.; Kho, J.; Rodriguez-Santana, J.R.; Casal, J.; Tsai, H.-J.; Torres, A.; Ziv, E.; et al. CD14 Tobacco Gene–Environment Interaction Modifies Asthma Severity and Immunoglobulin E Levels in Latinos with Asthma. Am. J. Respir. Crit. Care Med. 2005, 172, 173–182. [Google Scholar] [CrossRef]
  61. Martin, A.C.; Laing, I.A.; Khoo, S.K.; Zhang, G.; Rueter, K.; Teoh, L.; Taheri, S.; Hayden, C.M.; Geelhoed, G.C.; Goldblatt, J.; et al. Faculty Opinions recommendation of Acute asthma in children: Relationships among CD14 and CC16 genotypes, plasma levels, and severity. Am. J. Respir. Crit. Care Med. 2006, 173, 617–622. [Google Scholar] [CrossRef]
  62. Foley, S.C.; Mogas, A.K.; Olivenstein, R.; Fiset, P.O.; Chakir, J.; Bourbeau, J.; Ernst, P.; Lemière, C.; Martin, J.G.; Hamid, Q. Increased expression of ADAM33 and ADAM8 with disease progression in asthma. J. Allergy Clin. Immunol. 2007, 119, 863–871. [Google Scholar] [CrossRef] [PubMed]
  63. Tavendale, R.; Macgregor, D.F.; Mukhopadhyay, S.; Palmer, C.N. A polymorphism controlling ORMDL3 expression is associated with asthma that is poorly controlled by current medications. J. Allergy Clin. Immunol. 2008, 121, 860–863. [Google Scholar] [CrossRef] [PubMed]
  64. Bisgaard, H.; Bønnelykke, K.; Sleiman, P.M.A.; Brasholt, M.; Chawes, B.; Kreiner-Møller, E.; Stage, M.; Kim, C.; Tavendale, R.; Baty, F.; et al. Chromosome 17q21 Gene Variants Are Associated with Asthma and Exacerbations but Not Atopy in Early Childhood. Am. J. Respir. Crit. Care Med. 2009, 179, 179–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Berce, V.; Kozmus, C.; Potočnik, U. Association among ORMDL3 gene expression, 17q21 polymorphism and response to treatment with inhaled corticosteroids in children with asthma. Pharm. J. 2012, 13, 523–529. [Google Scholar] [CrossRef] [PubMed]
  66. Farzan, N.; Vijverberg, S.J.; Hernandez-Pacheco, N.; Bel, E.H.D.; Berce, V.; Bønnelykke, K.; Bisgaard, H.; Burchard, E.G.; Canino, G.; Celedón, J.C.; et al. 17q21 variant increases the risk of exacerbations in asthmatic children despite inhaled corticosteroids use. Allergy 2018, 73, 2083–2088. [Google Scholar] [CrossRef] [Green Version]
  67. Tantisira, K.G.; Lasky-Su, J.; Harada, M.; Murphy, A.; Litonjua, A.A.; Himes, B.E.; Lange, C.; Lazarus, R.; Sylvia, J.; Klanderman, B.; et al. Genome-wide association between GLCCI1 and response to glucocorticoid therapy in asthma. N. Engl. J. Med. 2011, 365, 1173–1183. [Google Scholar] [CrossRef] [Green Version]
  68. Himes, B.E.; Jiang, X.; Hu, R.; Wu, A.C.; Lasky-Su, J.A.; Klanderman, B.J.; Ziniti, J.; Senter-Sylvia, J.; Lima, J.J.; Irvin, C.G.; et al. Genome-Wide Association Analysis in Asthma Subjects Identifies SPATS2L as a Novel Bronchodilator Response Gene. PLOS Genet. 2012, 8, e1002824. [Google Scholar] [CrossRef] [Green Version]
  69. Israel, E.; Lasky-Su, J.; Markezich, A.; Damask, A.; Szefler, S.J.; Schuemann, B.; Klanderman, B.; Sylvia, J.; Kazani, S.; Wu, R.; et al. Genome-Wide Association Study of Short-Acting β2-Agonists. A Novel Genome-Wide Significant Locus on Chromosome 2 near ASB3. Am. J. Respir. Crit. Care Med. 2015, 191, 530–537. [Google Scholar] [CrossRef] [Green Version]
  70. Dijk, F.N.; Vijverberg, S.J.; Hernandez-Pacheco, N.; Repnik, K.; Karimi, L.; Mitratza, M.; Farzan, N.; Nawijn, M.C.; Burchard, E.G.; Engelkes, M.; et al. IL1RL1 gene variations are associated with asthma exacerbations in children and adolescents using inhaled corticosteroids. Allergy 2019, 75, 984–989. [Google Scholar] [CrossRef] [Green Version]
  71. Wan, Z.; Tang, Y.; Song, Q.; Zhang, J.; Xie, W.; He, Y.; Huang, R.; Zheng, X.; Liu, C.; Liu, J. Gene polymorphisms in VEGFA and COL2A1 are associated with response to inhaled corticosteroids in children with asthma. Pharmacogenomics 2019, 20, 947–955. [Google Scholar] [CrossRef]
  72. Dragicevic, S.; Kosnik, M.; Rankov, A.D.; Rijavec, M.; Milosevic, K.; Korosec, P.; Kavalar, M.S.; Nikolic, A. The Variants in the 3′ Untranslated Region of the Matrix Metalloproteinase 9 Gene as Modulators of Treatment Outcome in Children with Asthma. Lung 2018, 196, 297–303. [Google Scholar] [CrossRef] [PubMed]
  73. Levin, A.M.; Gui, H.; Hernandez-Pacheco, N.; Yang, M.; Xiao, S.; Yang, J.J.; Hochstadt, S.; Barczak, A.J.; Eckalbar, W.L.; Rynkowski, D.; et al. Integrative approach identifies corticosteroid response variant in diverse populations with asthma. J. Allergy Clin. Immunol. 2018, 143, 1791–1802. [Google Scholar] [CrossRef] [PubMed]
  74. Himes, B.E.; Jiang, X.; Wagner, P.; Hu, R.; Wang, Q.; Klanderman, B.; Whitaker, R.; Duan, Q.; Lasky-Su, J.; Nikolos, C.; et al. RNA-Seq Transcriptome Profiling Identifies CRISPLD2 as a Glucocorticoid Responsive Gene that Modulates Cytokine Function in Airway Smooth Muscle Cells. PLoS ONE 2014, 9, e99625. [Google Scholar] [CrossRef]
  75. Ramasamy, A.; Curjuric, I.; Coin, L.J.; Kumar, A.; McArdle, W.L.; Imboden, M.; Leynaert, B.; Kogevinas, M.; Schmid-Grendelmeier, P.; Pekkanen, J.; et al. A genome-wide meta-analysis of genetic variants associated with allergic rhinitis and grass sensitization and their interaction with birth order. J. Allergy Clin. Immunol. 2011, 128, 996–1005. [Google Scholar] [CrossRef] [PubMed]
  76. Lee, J.H.; McDonald, M.-L.N.; Cho, M.H.; Wan, E.S.; Castaldi, P.J.; Hunninghake, G.M.; Marchetti, N.; Lynch, D.A.; Crapo, J.D.; Lomas, D.A.; et al. DNAH5 is associated with total lung capacity in chronic obstructive pulmonary disease. Respir. Res. 2014, 15, 1–10. [Google Scholar] [CrossRef] [Green Version]
  77. Mak, A.C.Y.; White, M.J.; Eckalbar, W.L.; Szpiech, Z.A.; Oh, S.S.; Pino-Yanes, M.; Hu, D.; Goddard, P.; Huntsman, S.; Galanter, J.; et al. Whole-Genome Sequencing of Pharmacogenetic Drug Response in Racially Diverse Children with Asthma. Am. J. Respir. Crit. Care Med. 2018, 197, 1552–1564. [Google Scholar] [CrossRef]
  78. Perez-Garcia, J.; Espuela-Ortiz, A.; Lorenzo-Diaz, F.; Pino-Yanes, M. Pharmacogenetics of Pediatric Asthma: Current Perspectives. Pharm. Pers. Med. 2020, 13, 89–103. [Google Scholar] [CrossRef] [Green Version]
  79. Lutz, S.M.; Cho, M.H.; Young, K.; Hersh, C.P.; Castaldi, P.J.; McDonald, M.-L.; Regan, E.; Mattheisen, M.; DeMeo, D.L.; Parker, M.; et al. A genome-wide association study identifies risk loci for spirometric measures among smokers of European and African ancestry. BMC Genet. 2015, 16, 138. [Google Scholar] [CrossRef] [Green Version]
  80. Wain, L.V.; Shrine, N.; Artigas, M.S.; Erzurumluoglu, A.M.; Noyvert, B.; Bossini-Castillo, L.; Obeidat, M.; Henry, A.P.; Portelli, M.A.; Hall, R.J.; et al. Genome-wide association analyses for lung function and chronic obstructive pulmonary disease identify new loci and potential druggable targets. Nat. Genet. 2017, 49, 416–425. [Google Scholar] [CrossRef] [Green Version]
  81. Karimi, L.; Vijverberg, S.J.; Farzan, N.; Ghanbari, M.; Verhamme, K.M.; der Zee, A.H.M. FCER2 T2206C variant associated with FENO levels in asthmatic children using inhaled corticosteroids: The PACMAN study. Clin. Exp. Allergy 2019, 49, 1429–1436. [Google Scholar] [CrossRef] [Green Version]
  82. Balantic, M.; Rijavec, M.; Kavalar, M.S.; Suskovic, S.; Silar, M.; Kosnik, M.; Korosec, P. Asthma Treatment Outcome in Children Is Associated with Vascular Endothelial Growth Factor A (VEGFA) Polymorphisms. Mol. Diagn. Ther. 2012, 16, 173–180. [Google Scholar] [CrossRef] [PubMed]
  83. DeVries, A.; Vercelli, D. Epigenetic Mechanisms in Asthma. Ann. Am. Thorac. Soc. 2016, 13, S48–S50. [Google Scholar] [CrossRef] [PubMed]
  84. Kelsey, G.; Stegle, O.; Reik, W. Single-cell epigenomics: Recording the past and predicting the future. Science 2017, 358, 69–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Bae, D.-J.; Jun, J.A.; Chang, H.S.; Park, J.S.; Park, C.-S. Epigenetic Changes in Asthma: Role of DNA CpG Methylation. Tuberc. Respir. Dis. 2020, 83, 1–13. [Google Scholar] [CrossRef] [PubMed]
  86. WHO Global NCD Action Plan 2023-2030 and the United Nations 2030 Agenda for Sustainable Development. Available online: https://www.who.int/teams/noncommunicable-diseases/governance/roadmap (accessed on 4 June 2023).
  87. Bibi, H.; Shoseyov, D.; Feigenbaum, D.; Genis, M.; Friger, M.; Peled, R.; Sharff, S. The relationship between asthma and obesity in children: Is it real or a case of over diagnosis? J. Asthma 2004, 41, 403–410. [Google Scholar] [CrossRef]
  88. Lang, J.E.; Feng, H.; Lima, J.J. Body Mass Index-Percentile and Diagnostic Accuracy of Childhood Asthma. J. Asthma 2009, 46, 291–299. [Google Scholar] [CrossRef]
  89. Di Genova, L.; Penta, L.; Biscarini, A.; Di Cara, G.; Esposito, S. Children with Obesity and Asthma: Which Are the Best Options for Their Management? Nutrients 2018, 10, 1634. [Google Scholar] [CrossRef] [Green Version]
  90. Belamarich, P.F.; Luder, E.; Kattan, M.; Mitchell, H.; Islam, S.; Lynn, H.; Crain, E.F. Do obese inner-city children with asthma have more symptoms than nonobese children with asthma? Pediatrics 2000, 106, 1436–1441. [Google Scholar] [CrossRef]
  91. Sithole, F.; Douwes, J.; Burstyn, I.; Veugelers, P. Body Mass Index and Childhood Asthma: A Linear Association? J. Asthma 2008, 45, 473–477. [Google Scholar] [CrossRef]
  92. Ginde, A.A.; Santillan, A.A.; Clark, S.; Camargo, C.A., Jr. Body mass index and acute asthma severity among children presenting to the emergency department. Pediatr. Allergy Immunol. 2009, 21, 480–488. [Google Scholar] [CrossRef]
  93. Lovinsky-Desir, S.; Miller, R.L. Epigenetics, Asthma, and Allergic Diseases: A Review of the Latest Advancements. Curr. Allergy Asthma Rep. 2012, 12, 211–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Jones, P.A. Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 2012, 13, 484–492. [Google Scholar] [CrossRef] [PubMed]
  95. Thibeault, A.-A.H.; Laprise, C. Cell-Specific DNA Methylation Signatures in Asthma. Genes 2019, 10, 932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Gruzieva, O.; Merid, S.K.; Melén, E. An update on epigenetics and childhood respiratory diseases. Paediatr. Respir. Rev. 2014, 15, 348–354. [Google Scholar] [CrossRef]
  97. Kabesch, M.; Tost, J. Recent findings in the genetics and epigenetics of asthma and allergy. Semin. Immunopathol. 2020, 42, 43–60. [Google Scholar] [CrossRef] [Green Version]
  98. Brook, P.O.; Perry, M.M.; Adcock, I.M.; Durham, A.L. Epigenome-modifying tools in asthma. Epigenomics 2015, 7, 1017–1032. [Google Scholar] [CrossRef] [Green Version]
  99. Ntontsi, P.; Photiades, A.; Zervas, E.; Xanthou, G.; Samitas, K. Genetics and Epigenetics in Asthma. Int. J. Mol. Sci. 2021, 22, 2412. [Google Scholar] [CrossRef]
  100. Yang, I.V.; Pedersen, B.S.; Liu, A.H.; O’Connor, G.T.; Pillai, D.; Kattan, M.; Misiak, R.T.; Gruchalla, R.; Szefler, S.J.; Khurana Hershey, G.; et al. The nasal methylome and childhood atopic asthma. J. Allergy Clin. Immunol. 2017, 139, 1478–1488. [Google Scholar] [CrossRef] [Green Version]
  101. Xu, C.-J.; Söderhäll, C.; Bustamante, M.; Baïz, N.; Gruzieva, O.; Gehring, U.; Mason, D.; Chatzi, L.; Basterrechea, M.; Llop, S.; et al. DNA methylation in childhood asthma: An epigenome-wide meta-analysis. Lancet Respir. Med. 2018, 6, 379–388. [Google Scholar] [CrossRef] [Green Version]
  102. Qi, C.; Xu, C.-J.; Koppelman, G.H. The role of epigenetics in the development of childhood asthma. Expert Rev. Clin. Immunol. 2019, 15, 1287–1302. [Google Scholar] [CrossRef] [Green Version]
  103. Arathimos, R.; Suderman, M.; Sharp, G.C.; Burrows, K.; Granell, R.; Tilling, K.; Gaunt, T.R.; Henderson, J.; Ring, S.; Richmond, R.C.; et al. Epigenome-wide association study of asthma and wheeze in childhood and adolescence. Clin. Epigenetics 2017, 9, 112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Forno, E.; Wang, T.; Qi, C.; Yan, Q.; Xu, C.-J.; Boutaoui, N.; Han, Y.-Y.; Weeks, D.E.; Jiang, Y.; Rosser, F.; et al. DNA methylation in nasal epithelium, atopy, and atopic asthma in children: A genome-wide study. Lancet Respir. Med. 2018, 7, 336–346. [Google Scholar] [CrossRef]
  105. Cardenas, A.; Sordillo, J.E.; Rifas-Shiman, S.L.; Chung, W.; Liang, L.; Coull, B.A.; Hivert, M.-F.; Lai, P.S.; Forno, E.; Celedón, J.C.; et al. The nasal methylome as a biomarker of asthma and airway inflammation in children. Nat. Commun. 2019, 10, 3095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Reese, S.E.; Xu, C.-J.; Dekker, H.T.D.; Lee, M.K.; Sikdar, S.; Ruiz-Arenas, C.; Merid, S.K.; Rezwan, F.I.; Page, C.M.; Ullemar, V.; et al. Epigenome-wide meta-analysis of DNA methylation and childhood asthma. J. Allergy Clin. Immunol. 2018, 143, 2062–2074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Nicodemus-Johnson, J.; Myers, R.A.; Sakabe, N.J.; Sobreira, D.R.; Hogarth, D.K.; Naureckas, E.T.; Sperling, A.; Solway, J.; White, S.R.; Nobrega, M.A.; et al. DNA methylation in lung cells is associated with asthma endotypes and genetic risk. J. Clin. Investig. 2016, 1, e90151. [Google Scholar] [CrossRef]
  108. Lund, R.J.; Osmala, M.; Malonzo, M.; Lukkarinen, M.; Leino, A.; Salmi, J.; Vuorikoski, S.; Turunen, R.; Vuorinen, T.; Akdis, C.; et al. Atopic asthma after rhinovirus-induced wheezing is associated with DNA methylation change in the SMAD3 gene promoter. Allergy 2018, 73, 1735–1740. [Google Scholar] [CrossRef] [Green Version]
  109. Rastogi, D.; Suzuki, M.; Greally, J.M. Differential epigenome-wide DNA methylation patterns in childhood obesity-associated asthma. Sci. Rep. 2013, 3, srep02164. [Google Scholar] [CrossRef] [Green Version]
  110. Breton, C.V.; Byun, H.-M.; Wang, X.; Salam, M.T.; Siegmund, K.; Gilliland, F.D. DNA Methylation in the Arginase–Nitric Oxide Synthase Pathway Is Associated with Exhaled Nitric Oxide in Children with Asthma. Am. J. Respir. Crit. Care Med. 2011, 184, 191–197. [Google Scholar] [CrossRef] [Green Version]
  111. Stefanowicz, D.; Hackett, T.-L.; Garmaroudi, F.S.; Günther, O.; Neumann, S.; Sutanto, E.N.; Ling, K.-M.; Kobor, M.; Kicic, A.; Stick, S.; et al. DNA Methylation Profiles of Airway Epithelial Cells and PBMCs from Healthy, Atopic and Asthmatic Children. PLoS ONE 2012, 7, e44213. [Google Scholar] [CrossRef] [Green Version]
  112. Salam, M.T. Asthma Epigenetics. Heterog. Asthma 2013, 795, 183–199. [Google Scholar] [CrossRef]
  113. Kidd, C.D.A.; Thompson, P.J.; Barrett, L.; Baltic, S. Histone Modifications and Asthma. The Interface of the Epigenetic and Genetic Landscapes. Am. J. Respir. Cell Mol. Biol. 2016, 54, 3–12. [Google Scholar] [CrossRef] [PubMed]
  114. Ito, K.; Caramori, G.; Lim, S.; Oates, T.; Chung, K.F.; Barnes, P.J.; Adcock, I.M. Expression and Activity of Histone Deacetylases in Human Asthmatic Airways. Am. J. Respir. Crit. Care Med. 2002, 166, 392–396. [Google Scholar] [CrossRef] [PubMed]
  115. Su, R.-C.; Becker, A.B.; Kozyrskyj, A.L.; HayGlass, K.T. Epigenetic regulation of established human type 1 versus type 2 cytokine responses. J. Allergy Clin. Immunol. 2008, 121, 57–63.e3. [Google Scholar] [CrossRef] [PubMed]
  116. Choi, J.-H.; Oh, S.-W.; Kang, M.-S.; Kwon, H.; Oh, G.-T.; Kim, D.-Y. Trichostatin A attenuates airway inflammation in mouse asthma model. Clin. Exp. Allergy 2004, 35, 89–96. [Google Scholar] [CrossRef]
  117. Yang, S.R.; Wright, J.; Bauter, M.; Seweryniak, K.; Kode, A.; Rahman, I. Sirtuin regulates cigarette smoke-induced proinflammatory mediator release via RelA/p65 NF-κB in macrophages in vitro and in rat lungs in vivo: Implications for chronic inflammation and aging. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007, 292, L567–L576. [Google Scholar] [CrossRef] [Green Version]
  118. Abbring, S.; Wolf, J.; Ayechu-Muruzabal, V.; Diks, M.A.; Alhamdan, F.; Harb, H.; Renz, H.; Garn, H.; Potaczek, D.P.; Van Esch, B.C.; et al. Raw Cow’s Milk Reduces Allergic Symptoms in a Murine Model for Food Allergy—A Potential Role for Epigenetic Modifications. Nutrients 2019, 11, 1721. [Google Scholar] [CrossRef] [Green Version]
  119. Ren, Y.; Li, M.; Bai, S.; Kong, L.; Su, X. Identification of histone acetylation in a murine model of allergic asthma by proteomic analysis. Exp. Biol. Med. 2020, 246, 929–939. [Google Scholar] [CrossRef]
  120. Chen, L.-F.; Fischle, W.; Verdin, E.; Greene, W.C. Duration of Nuclear NF-κB Action Regulated by Reversible Acetylation. Science 2001, 293, 1653–1657. [Google Scholar] [CrossRef] [Green Version]
  121. Grausenburger, R.; Bilic, I.; Boucheron, N.; Zupkovitz, G.; El-Housseiny, L.; Tschismarov, R.; Zhang, Y.; Rembold, M.; Gaisberger, M.; Hartl, A.; et al. Conditional Deletion of Histone Deacetylase 1 in T Cells Leads to Enhanced Airway Inflammation and Increased Th2 Cytokine Production. J. Immunol. 2010, 185, 3489–3497. [Google Scholar] [CrossRef] [Green Version]
  122. Wang, Y.; Tian, Y.; Morley, M.P.; Lu, M.M.; DeMayo, F.J.; Olson, E.N.; Morrisey, E.E. Development and Regeneration of Sox2+ Endoderm Progenitors Are Regulated by a HDAC1/2-Bmp4/Rb1 Regulatory Pathway. Dev. Cell 2013, 24, 345–358. [Google Scholar] [CrossRef] [Green Version]
  123. Seumois, G.; Chavez, L.; Gerasimova, A.; Lienhard, M.; Omran, N.; Kalinke, L.; Vedanayagam, M.; Ganesan, A.P.V.; Chawla, A.; Djukanović, R.; et al. Epigenomic analysis of primary human T cells reveals enhancers associated with TH2 memory cell differentiation and asthma susceptibility. Nat. Immunol. 2014, 15, 777–788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Harb, H.; Raedler, D.; Ballenberger, N.; Böck, A.; Kesper, D.A.; Renz, H.; Schaub, B. Childhood allergic asthma is associated with increased IL-13 and FOXP3 histone acetylation. J. Allergy Clin. Immunol. 2015, 136, 200–202. [Google Scholar] [CrossRef] [PubMed]
  125. Stefanowicz, D.; Lee, J.Y.; Lee, K.; Shaheen, F.; Koo, H.-K.; Booth, S.; Knight, D.A.; Hackett, T.-L. Elevated H3K18 acetylation in airway epithelial cells of asthmatic subjects. Respir. Res. 2015, 16, 95. [Google Scholar] [CrossRef] [Green Version]
  126. Puddicombe, S.M.; Polosa, R.; Richter, A.; Krishna, M.T.; Howarth, P.H.; Holgate, S.T.; Davies, D.E. Involvement of the epidermal growth factor receptor in epithelial repair in asthma. FASEB J. 2000, 14, 1362–1374. [Google Scholar] [CrossRef] [Green Version]
  127. Boxall, C.; Holgate, S.T.; Davies, D.E. The contribution of transforming growth factor-beta and epidermal growth factor signalling to airway remodelling in chronic asthma. Eur. Respir. J. 2006, 27, 208–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Amishima, M.; Munakata, M.; Nasuhara, Y.; Sato, A.; Takahashi, T.; Homma, Y.; Kawakami, Y. Expression of Epidermal Growth Factor and Epidermal Growth Factor Receptor Immunoreactivity in the Asthmatic Human Airway. Am. J. Respir. Crit. Care Med. 1998, 157, 1907–1912. [Google Scholar] [CrossRef] [Green Version]
  129. Mullings, R.E.; Wilson, S.J.; Puddicombe, S.M.; Lordan, J.L.; Bucchieri, F.; Djukanović, R.; Howarth, P.H.; Harper, S.; Holgate, S.T.; Davies, D.E. Signal transducer and activator of transcription 6 (STAT-6) expression and function in asthmatic bronchial epithelium. J. Allergy Clin. Immunol. 2001, 108, 832–838. [Google Scholar] [CrossRef]
  130. Tomita, K.; Caramori, G.; Ito, K.; Sano, H.; Lim, S.; Oates, T.; Cosio, B.; Chung, K.F.; Tohda, Y.; Barnes, P.J.; et al. STAT6 expression in T cells, alveolar macrophages and bronchial biopsies of normal and asthmatic subjects. J. Inflamm. 2012, 9, 5. [Google Scholar] [CrossRef] [Green Version]
  131. Cheng, Q.; Shang, Y.; Huang, W.; Zhang, Q.; Li, X.; Zhou, Q. p300 mediates the histone acetylation of ORMDL3 to affect airway inflammation and remodeling in asthma. Int. Immunopharmacol. 2019, 76, 105885. [Google Scholar] [CrossRef]
  132. Wawrzyniak, P.; Wawrzyniak, M.; Wanke, K.; Sokolowska, M.; Bendelja, K.; Rückert, B.; Globinska, A.; Jakiela, B.; Kast, J.I.; Idzko, M.; et al. Regulation of bronchial epithelial barrier integrity by type 2 cytokines and histone deacetylases in asthmatic patients. J. Allergy Clin. Immunol. 2016, 139, 93–103. [Google Scholar] [CrossRef] [Green Version]
  133. Harb, H.; Alhamwe, B.A.; Garn, H.; Renz, H.; Potaczek, D.P. Recent developments in epigenetics of pediatric asthma. Curr. Opin. Pediatr. 2016, 28, 754–763. [Google Scholar] [CrossRef] [PubMed]
  134. García-Serna, A.M.; Martín-Orozco, E.; Hernández-Caselles, T.; Morales, E. Prenatal and Perinatal Environmental Influences Shaping the Neonatal Immune System: A Focus on Asthma and Allergy Origins. Int. J. Environ. Res. Public Health 2021, 18, 3962. [Google Scholar] [CrossRef] [PubMed]
  135. Kabesch, M. Early origins of asthma (and allergy). Mol. Cell. Pediatr. 2016, 3, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Potaczek, D.P.; Harb, H.; Michel, S.; Alhamwe, B.A.; Renz, H.; Tost, J. Epigenetics and allergy: From basic mechanisms to clinical applications. Epigenomics 2017, 9, 539–571. [Google Scholar] [CrossRef]
  137. Brand, S.; Teich, R.; Dicke, T.; Harb, H.; Yildirim, A.O.; Tost, J.; Schneider-Stock, R.; Waterland, R.A.; Bauer, U.M.; von Mutius, E. Epigenetic regulation in mouse offspring as a novel mechanism for transmaternal protection against microbial-induced asthma. J. Allergy Clin. Immunol. 2011, 128, 618–625.e7. [Google Scholar] [CrossRef]
  138. Harb, H.; Alhamwe, B.A.; Acevedo, N.; Frumento, P.; Johansson, C.; Eick, L.; Papadogiannakis, N.; Alm, J.; Renz, H.; Potaczek, D.P.; et al. Epigenetic Modifications in Placenta are Associated with the Child’s Sensitization to Allergens. BioMed Res. Int. 2019, 2019, 1–11. [Google Scholar] [CrossRef]
  139. Ambros, V. The functions of animal microRNAs. Nature 2004, 431, 350–355. [Google Scholar] [CrossRef]
  140. Ariel, D.; Upadhyay, D. The role and regulation of microRNAs in asthma. Curr. Opin. Allergy Clin. Immunol. 2012, 12, 49–52. [Google Scholar] [CrossRef]
  141. Specjalski, K.; Jassem, E. MicroRNAs: Potential biomarkers and targets of therapy in allergic diseases? Arch. Immunol. Ther. Exp. 2019, 67, 213–223. [Google Scholar] [CrossRef] [Green Version]
  142. Narożna, B.; Langwiński, W.; Szczepankiewicz, A. Non-coding RNAs in pediatric airway diseases. Genes 2017, 8, 348. [Google Scholar] [CrossRef] [Green Version]
  143. Feketea, G.; Bocsan, C.I.; Popescu, C.; Gaman, M.; Stanciu, L.A.; Zdrenghea, M.T. A Review of Macrophage MicroRNAs’ Role in Human Asthma. Cells 2019, 8, 420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Wu, X.B.; Wang, M.Y.; Zhu, H.Y.; Tang, S.Q.; You, Y.D.; Xie, Y.Q. Overexpression of microRNA-21 and microRNA-126 in the patients of bronchial asthma. Int. J. Clin. Exp. Med. 2014, 7, 1307–1312. [Google Scholar] [PubMed]
  145. Pua, H.H.; Ansel, K.M. MicroRNA regulation of allergic inflammation and asthma. Curr. Opin. Immunol. 2015, 36, 101–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Elbehidy, R.M.; Youssef, D.; El-Shal, A.S.; Shalaby, S.M.; Sherbiny, H.S.; Sherief, L.M.; E Akeel, N. MicroRNA–21 as a novel biomarker in diagnosis and response to therapy in asthmatic children. Mol. Immunol. 2016, 71, 107–114. [Google Scholar] [CrossRef] [PubMed]
  147. Sawant, D.V.; Yao, W.; Wright, Z.; Sawyers, C.; Tepper, R.S.; Gupta, S.K.; Kaplan, M.H.; Dent, A.L. Serum MicroRNA-21 as a Biomarker for Allergic Inflammatory Disease in Children. MicroRNA 2015, 4, 36–40. [Google Scholar] [CrossRef] [Green Version]
  148. Hammad Mahmoud Hammad, R.; Hamed, D.H.E.D.; Eldosoky, M.A.E.R.; Ahmad, A.A.E.S.; Osman, H.M.; Abd Elgalil, H.M.; Mahmoud Hassan, M.M. Plasma microRNA-21, microRNA-146a and IL-13 expression in asthmatic children. Innate Immun. 2018, 24, 171–179. [Google Scholar] [CrossRef] [Green Version]
  149. Karam, R.A.; Elrahman, D.M.A. Differential expression of miR-155 and Let-7a in the plasma of childhood asthma: Potential biomarkers for diagnosis and severity. Clin. Biochem. 2019, 68, 30–36. [Google Scholar] [CrossRef]
  150. Johansson, K.; Malmhäll, C.; Ramos-Ramírez, P.; Rådinger, M. MicroRNA-155 is a critical regulator of type 2 innate lymphoid cells and IL-33 signaling in experimental models of allergic airway inflammation. J. Allergy Clin. Immunol. 2017, 139, 1007–1016.e9. [Google Scholar] [CrossRef] [Green Version]
  151. Kumar, M.; Ahmad, T.; Sharma, A.; Mabalirajan, U.; Kulshreshtha, A.; Agrawal, A.; Ghosh, B. Let-7 microRNA-mediated regulation of IL-13 and allergic airway inflammation. J Allergy Clin. Immunol. 2011, 128, 1077–1085.e10. [Google Scholar] [CrossRef]
  152. Su, X.W.; Yang, Y.; Lv, M.L.; Li, L.J.; Dong, W.; Liao, M.; Gao, L.B.; Luo, H.B.; Liu, Y.; Cong, R.J.; et al. Association between single-nucleotide polymorphisms in pre-miRNAs and the risk of asthma in a Chinese population. DNA Cell Biol. 2011, 30, 919–923. [Google Scholar] [CrossRef]
  153. Davis, J.S.; Sun, M.; Kho, A.T.; Moore, K.G.; Sylvia, J.M.; Weiss, S.T.; Lu, Q.; Tantisira, K.G. Circulating microRNAs and association with methacholine PC20 in the Childhood Asthma Management Program (CAMP) cohort. PLoS ONE 2017, 12, e0180329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Panganiban, R.P.; Wang, Y.; Howrylak, J.; Chinchilli, V.M.; Craig, T.J.; August, A.; Ishmael, F.T. Circulating microRNAs as biomarkers in patients with allergic rhinitis and asthma. J. Allergy Clin. Immunol. 2016, 137, 1423–1432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. McGeachie, M.J.; Davis, J.S.; Kho, A.T.; Dahlin, A.; Sordillo, J.E.; Sun, M.; Lu, Q.; Weiss, S.T.; Tantisira, K.G. Asthma remission: Predicting future airways responsiveness using an miRNA network. J. Allergy Clin. Immunol. 2017, 140, 598–600.e8. [Google Scholar] [CrossRef] [Green Version]
  156. Milger, K.; Götschke, J.; Krause, L.; Nathan, P.; Alessandrini, F.; Tufman, A.; Fischer, R.; Bartel, S.; Theis, F.J.; Behr, J.; et al. Identification of a plasma miRNA biomarker signature for allergic asthma: A translational approach. Allergy 2017, 72, 1962–1971. [Google Scholar] [CrossRef]
  157. Lu, T.X.; Hartner, J.; Lim, E.J.; Fabry, V.; Mingler, M.K.; Cole, E.T.; Orkin, S.H.; Aronow, B.J.; Rothenberg, M.E. Microrna-21 limits in vivo immune response-mediated activation of the il-12/ifn-gamma pathway, th1 polarization, and the severity of delayed-type hypersensitivity. J. Immunol. 2011, 187, 3362–3373. [Google Scholar] [CrossRef] [Green Version]
  158. Comer, B.; Camoretti-Mercado, B.; Kogut, P.C.; Halayko, A.J.; Solway, J.; Gerthoffer, W.T. MicroRNA-146a and microRNA-146b expression and anti-inflammatory function in human airway smooth muscle. Am. J. Physiol. Cell. Mol. Physiol. 2014, 307, L727–L734. [Google Scholar] [CrossRef] [PubMed]
  159. Zhou, H.; Li, J.; Gao, P.; Wang, Q.; Zhang, J. miR-155: A Novel Target in Allergic Asthma. Int. J. Mol. Sci. 2016, 17, 1773. [Google Scholar] [CrossRef] [Green Version]
  160. Simpson, L.J.; Patel, S.; Bhakta, N.R.; Choy, D.; Brightbill, H.D.; Ren, X.; Wang, Y.; Pua, H.H.; Baumjohann, D.; Montoya, M.M.; et al. A microRNA upregulated in asthma airway T cells promotes TH2 cytokine production. Nat. Immunol. 2014, 15, 1162–1170. [Google Scholar] [CrossRef]
  161. Haj-Salem, I.; Fakhfakh, R.; Bérubé, J.-C.; Jacques, E.; Plante, S.; Simard, M.J.; Bossé, Y.; Chakir, J. MicroRNA-19a enhances proliferation of bronchial epithelial cells by targetingTGFβR2gene in severe asthma. Allergy 2014, 70, 212–219. [Google Scholar] [CrossRef]
  162. Mattes, J.; Collison, A.; Plank, M.; Phipps, S.; Foster, P.S. Antagonism of microRNA-126 suppresses the effector function of T H 2 cells and the development of allergic airways disease. Proc. Natl. Acad. Sci. USA 2009, 106, 18704–18709. [Google Scholar] [CrossRef] [Green Version]
  163. Tian, M.; Ji, Y.; Wang, T.; Zhang, W.; Zhou, Y.; Cui, Y. Changes in circulating microRNA-126 levels are associated with immune imbalance in children with acute asthma. Int. J. Immunopathol. Pharmacol. 2018, 32, 2058738418779243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Kho, A.T.; Sharma, S.; Davis, J.S.; Spina, J.; Howard, D.; McEnroy, K.; Moore, K.; Sylvia, J.; Qiu, W.; Weiss, S.T.; et al. Circulating MicroRNAs: Association with Lung Function in Asthma. PLoS ONE 2016, 11, e0157998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Naydenova, K.; Dimitrov, V.; Velikova, T. Immunological and microRNA Features of Allergic Rhinitis in the Context of United Airway Disease. Sinusitis 2021, 5, 45–52. [Google Scholar] [CrossRef]
Figure 1. Some of the common risk factors associated with childhood asthma.
Figure 1. Some of the common risk factors associated with childhood asthma.
Allergies 03 00009 g001
Figure 2. Clinical implications of epigenetic regulation (histone modifications, DNA methylation and non-coding RNAs).
Figure 2. Clinical implications of epigenetic regulation (histone modifications, DNA methylation and non-coding RNAs).
Allergies 03 00009 g002
Table 1. Summary of genes with relevance significant to asthma in children identified by the GWAS.
Table 1. Summary of genes with relevance significant to asthma in children identified by the GWAS.
Genome-Wide Association Studies
GeneLocus/Chr. Region *Function/BiomarkerPopulationRef.
DENND1B1q31Involved in T cell receptor signaling pathway and positive regulation of T-helper 2 cell cytokine productionNorth American children of European ancestrySleiman et al., 2010 [18]
DPP102q14Involved in positive regulation of protein localization to plasma membrane and proteolysis; IL-1 gene clusterEnglish and German;
American Blacks,
African Caribbean
Allen et al., 2003 [19]
ORMDL3
GSDMB
17q21Acts upstream or within negative regulation of B cell apoptotic process; Europeans, Asians,
Hispanics,
Moffatt et al., 2007 [17]
Moffatt et al., 2010 [29]
Thomsen et al., 2015 [35]
Demenais et al., 2019 [23]
CHI3LI1q32.1YKL-40 plays a role in the pathogenesis of autoimmune diseases; Europeans,
Hutterites, American,
German
Ober et al., 2008 [21]
Chen & Wang, 2017 [22]
HLA-DQ6p21MHC class II loci; involved in immunoglobulin production and immunoglobulin-mediated immune responseAmerican Whites,
Europeans
Li et al., 2010 [30]
Thomsen et al., 2015 [35] Demenais et al., 2019 [23]
RAD505q31Involved in DNA double-strand break processing; involved in mitotic G2/M transition checkpoint; Northeastern Han Chinese, Li et al., 2010 [30]
Thomsen, 2015 [35]
IL135q31Inflammation; involved in the positive regulation of macrophage activationNortheastern Han Chinese,
Europeans
Li et al., 2010 [30]
Thomsen, 2015 [35]
CDHR37q22Pronounced severe and early onset of disease; related to increased IL-17A response to viral infections; EuropeansBonnelykke et al., 2014 [31]
Eliasen et al., 2022 [34]
IL1RL1
IL18R1
2q11
2q12
Encodes a protein in the Toll-like receptor superfamily/encodes the IL-18 receptor;Icelanders, Europeans,
East Asians
Moffatt et al., 2010 [29]
Hayden et al., 2018 [28]
Ferreira et al., 2017 [25]
IL339p24Encodes IL-33, belongs to the IL-1 superfamily of proteins; EuropeansMoffat et al., 2007 [17]
IRF4
NOD2
IL4R
IL2RA
6p25
16q12
16p12
10p15
Involved in the regulation of T-helper cell differentiation and innate immune response; cytokine signaling in the immune system;EuropeansFerreira et al., 2017 [25]
KIAA1958
IL1RL1
IL13 LINC01149
9q32
2q12
5q31
6p21
Encodes a protein in the Toll-like receptor superfamily/encodes the IL-18 receptor; involved in positive regulation of macrophage activation; EuropeansLi et al., 2010 [30]
Hayden et al., 2018 [25]
DMRT19p24Influence sex-specific patterns of childhood asthma; potential involvement in hormone or immune cell regulation;EuropeansSchieck et al., 2016 [37]
TNFSF159q32Not expressed in B and T cells; the expression of the gene product is inducible by TNF and IL-1 alpha;Koreans, AsiaKim et al., 2022 [51]
* https://omim.org/; https://www.ncbi.nlm.nih.gov/gene. Last accessed on 7 June 2023.
Table 2. Summary of some genes critical for childhood asthma severity or/and drug/treatment response.
Table 2. Summary of some genes critical for childhood asthma severity or/and drug/treatment response.
GeneLocus/Chr. Region *Association Effect (Pathogenesis, Function, Severity, Treatment)Ref.
ADBR25q32Arg16-variant is associated with improved response to Beta-2 agonism; asthma severity;Israel et al., 2000 [52]
Litonjua et al., 2010 [53]
Sood et al., 2018 [54]
Scaparrotta et al., 2019 [55]
IL45q31Encodes interleukin-4, associated with reduced forced expiratory volume in 1 s; associated with asthma severity;Burchard et al., 1999 [56]
ALOX510q115-lipoxygenase, associated with improved response to ALOX5
inhibitor; worse lung function and asthma control;
Drazen et al., 1999 [57]
Mougey et al., 2012 [58]
CRHR117q21Intronic region of the corticotrophin-releasing hormone receptor 1 gene, response to inhaled corticosteroids;Tantisira et al., 2004 [59]
CD145q31Associated with asthma severity with modification from secondhand smoke; associated with asthma severity; Choudhry et al., 2005 [60]
Martin et al., 2006 [61]
ADAM3320p13Increased in severe asthma vs.
mild asthma or no asthma; moderate and severe asthma;
Foley et al., 2007 [62]
ORMDL3 cluster17q21Association with exacerbations and poor asthma control on controller medication; response to treatment with inhaled corticosteroids; proinflammatory airway epithelial response;Tavendale et al., 2008 [63]
Bisgaard et al., 2009 [64]
Berce et al., 2013 [65]
Farzan et al., 2018 [66]
GLCCI17p21Response to inhaled corticosteroids; Tantisira et al., 2011 [67]
SPATS2L2q33Related to bronchodilator responsiveness; related to different ranges of asthma severity, age, and baseline;Himes et al., 2012 [68]
ASB32p16Modulates pulmonary function; bronchodilator response to inhaledβ2-agonists; Israel et al., 2015 [69]
IL1RL12q12Response to inhaled corticosteroids; related to asthma susceptibility and severity;Dijk et al., 2019 [70]
COL2A112q13Response to inhaled corticosteroids; related to mild-to-moderate asthma;Wan et al., 2019 [71]
MMP920q13MMP-9 is released by inflammatory cells in response to allergen provocation in asthma, declining lung function; Dragicevic et al., 2018 [72]
EDDM3B14q11Inhaled corticosteroids in pediatric asthma;Levin et al., 2019 [73]
CRISPLD216q24Associated with an adaptive response to asthma; a glucocorticoid responsive gene in airway smooth muscle cells; Himes et al., 2014 [74]
Almoguera et al., 2017 [42]
DNAH55p15Associated with lung function or
the presence/absence of COPD; moderate-to-severe persistent asthma;
Ramasamy et al., 2011 [75]
Lee et al., 2014 [76]
Mak et al., 2018 [77]
Perez-Garcia et al., 2020 [78]
FCER219p13Associated with response to inhaled corticosteroids; associated with asthma severity;Lutz et al., 2015 [79]
Wain et al., 2017 [80]
Karimi et al., 2019 [81]
VEGFA6p21Associated with response to inhaled corticosteroids therapy severity of bronchial asthma;Balantic et al., 2012 [82]
Wan et al., 2019 [71]
* https://omim.org/; https://www.ncbi.nlm.nih.gov/gene. Last accessed on 7 June 2023.
Table 4. Summary of the studies about the role of histone modification and childhood asthma.
Table 4. Summary of the studies about the role of histone modification and childhood asthma.
Genes/Candidate-Genes *Histone ModificationsTissueRef.
IL-13, FOXP3histone H3 acetylationPeripheral bloodHarb et al., 2015 [124]
IL-4, IL-13higher expression of HDACs 1 and 9Bronchial epitheliumWawrzyniak et al., 2016 [132]
STAT6, EGFR and ΔNp63histone H3 acetylationBronchial airway epithelial cellsStefanowics et al., 2015 [125]
IFNG, SH2B3
HDAC4
histone H3 acetylation
histone H4 acetylation
Blood samples
Placenta samples
Harb et al., 2019 [138]
ORMDL3histone H3 acetylationBronchial epitheliumCheng et al., 2019 [131]
* Some genes were identified in more than one study.
Table 5. Summary of some significant miRNAs’ studies associated with asthma.
Table 5. Summary of some significant miRNAs’ studies associated with asthma.
miRNAsLevelFunction/Role in AsthmaTissueRef.
miR-21increased- eosinophilic accumulation
- correlation with IL-13
- Th2 response
- suppression of IL-12p35
bronchial epithelial cellsLu et al., 2011 [157]
Wu et al., 2014 [144]
Pua et al., 2015 [145]
Elbehidy R., 2016 [146]
miR-146increased- increase the eosinophilic level
- anti-inflammatory
- negative regulation of IL1β and COX2
blood samples,
human ASM cells
Su et al., 2011 [152]
Comer et al., 2014 [158]
Hammad et al., 2018 [148]
miR-155increased- decreased level of Let-7a
- regulation of IL-13
- in Th2 immune responses
blood samplesZhou et al., 2016 [159]
Karam et al., 2019 [149]
miR-19increased-mediate the production of IL-13
-targets mRNA of the TGFβR2 gene
-regulation in responses of the Th2 cell
bronchial epithelial cells,
airway T cells
Simpson et al., 2014 [160]
Haj-Salem et al., 20015 [161]
miR-126Increased- increase AHR and immune migration
- Th2 response
- increase the eosinophilic level
bronchial epithelial cells,
peripheral blood
Mattes et al., 2009 [162]
Wu et al., 2014 [144]
Tian et al., 2018 [163]
miR-15b, 126,
-139,
-142,
-186 etc.
- lung function parameters in childrenBlood samplesKho et al., 2016 [164]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Miteva, D.; Lazova, S.; Velikova, T. Genetic and Epigenetic Factors in Risk and Susceptibility for Childhood Asthma. Allergies 2023, 3, 115-133. https://doi.org/10.3390/allergies3020009

AMA Style

Miteva D, Lazova S, Velikova T. Genetic and Epigenetic Factors in Risk and Susceptibility for Childhood Asthma. Allergies. 2023; 3(2):115-133. https://doi.org/10.3390/allergies3020009

Chicago/Turabian Style

Miteva, Dimitrina, Snezhina Lazova, and Tsvetelina Velikova. 2023. "Genetic and Epigenetic Factors in Risk and Susceptibility for Childhood Asthma" Allergies 3, no. 2: 115-133. https://doi.org/10.3390/allergies3020009

Article Metrics

Back to TopTop