Abstract
The clinical and socioeconomic burden of asthma exacerbations (AEs) represents a major public health problem. In the last four years, there has been an increase in ethnic diversity in candidate-gene and genome-wide association studies (GWAS) of AEs, which in the latter case has led to the identification of novel genes and underlying pathobiological processes. Pharmacogenomics, admixture mapping analyses, and the combination of multiple “omic” layers have contributed to prioritizing genomic regions of interest and/or understanding the functional consequences of genetic variation. Despite this, the field still lags behind the genomics of asthma, where a vast compendium of genetic approaches has been used (e.g., gene-environment interactions, next-generation sequencing, or polygenic risk scores). Furthermore, the roles of the DNA methylome and histone modifications in AEs have been scarcely investigated, and microRNA findings remain to be validated in independent studies. Likewise, the most recent transcriptomic studies highlight the importance of host-airway microbiome interaction in the modulation of AEs risk. Leveraging -omics and deep-phenotyping data from sub-types or homogenous subgroups of patients will be crucial to overcome the inherent heterogeneity of AEs, and boost the identification of potential therapeutic targets and the implementation of precision medicine in clinical practice for AEs.
Keywords: asthma exacerbations, genomics, epigenetics, transcriptomics
INTRODUCTION
Asthma exacerbations (AEs) are episodes of worsening symptoms requiring a change in treatment. AE events can be severe, and while multiple criteria have been used in the literature [1], a common definition comprises asthma-related hospitalizations, emergency department (ED) visits, and the use of systemic (oral, intramuscular, or intravenous) corticosteroids. However, this definition should be regarded cautiously because it often relies on a subject’s self-report without incorporating other clinical or physiological parameters capturing the underlying pathophysiology of the episode [2].
AEs are a major public health problem and a priority in asthma research. Each year, approximately 75,000 people are hospitalized, and 15,000 people die from asthma in the United Kingdom (UK) [3]. In the United States (US), there are ~170,000 asthma-related hospitalizations, 1.8 million ER visits, and ~4,000 asthma-related deaths per annum. In fact, yearly asthma healthcare expenditures amount to £1.1 billion in the UK [4] and $50.3 billion in the US [5]. Indirect asthma costs, such as work and school absences, further increase the economic impact of asthma [5]. Moreover, AEs affect the quality of life of individuals with asthma [6,7] and their caregivers [8]. Although several studies have found an association between AEs and lung function decline [9–13], infants with a reduced airway calibre may also be at higher risk of loss of lung function and AEs [14,15]. In fact, the baseline airway wall area percent, an indicator of airway remodelling, is associated with the annual rate of future AEs and long-term decline in lung function [16,17].
To date, the best predictor of AEs is having had one within the last year [18–20], highlighting the key roles of genetic factors and/or early-life determinants. In addition, identifying clinically relevant biomarkers or predictors of AEs is crucial to guide the reduction and prevention of AEs. AEs are likely due to the complex interplay of genetic, environmental, and behavioural factors [18,21–23]. In fact, risk factors for AEs comprise allergen, air pollution or tobacco exposure, viral infections, psychological stress, treatment adherence, obesity, or genetic factors, among others [18].
Ethnic differences in the patterns of AEs are evidenced worldwide. In the US, African Americans and Puerto Ricans exhibit higher rates of AEs [24–28], while in Europe, AE rates are higher in Southern European countries [29]. African Americans are also more likely to have longer lengths of stay in intensive care units compared with Europeans [30]. In fact, African ancestry has been associated with AEs among African Americans [31] and other individuals of African descent in the US [32]. However, this association has not been validated for the number of exacerbations [33] or in Puerto Ricans [34], a recently-admixed population with up to ~25% African ancestry [34,35]. More recent findings suggest that the association of African ancestry and asthma re-admissions in African Americans may be mediated by disease management and socioeconomic factors [36].
A detailed description of genetic association studies of AE published until 15th November 2018 was previously reported by Herrera-Luis et al. [16]. Gautam et al. [37] reviewed the transcriptomics of asthma susceptibility, disease severity, and AEs prior to 2022, but no extensive summary of epigenetic studies of AEs has been published. In this review, we discuss the latest findings from -omics studies of AEs, assess methodological challenges, and propose future directions in this research field. For that purpose, we provide an update on the state of the art of the genomics and transcriptomics of AEs from the aforementioned dates to 1st October 2022 and review the epigenetic articles of AEs ever published.
LITERATURE MINING
Literature mining of genomic, epigenetic, and transcriptomic studies of AEs was conducted in PubMed [38] applying the following search terms: (Transcriptomics [Title/Abstract] OR Candidate gene [Title/Abstract] OR polymorphism [Title/Abstract] OR SNP [Title/Abstract] OR Genetics [Title/Abstract] OR GWAS [Title/Abstract] OR EWAS [Title/Abstract] OR epigenetic [Title/Abstract] OR methylation [Title/Abstract] OR histone [Title/Abstract] OR micro RNA [Title/ Abstract] OR mRNA [Title/ Abstract] OR transcriptomics [Title/ Abstract]) AND (asthma with exacerbations [Title/Abstract]). We excluded manuscripts reporting 1) reviews, editorials, or opinion articles, 2) findings in animals or cell lines, or 3) no data on -omics or candidate-gene associations with AEs.
GENETIC ASSOCIATION STUDIES
The genetic determinants of AEs have been thoroughly investigated using hypothesis-driven approaches to select genomic regions of interest [1] (Figure 1A). However, candidate-gene association studies are hampered by non-reproducibility across studies and a low likelihood of identifying true biological risk variants because of the polygenic structure underlying complex human traits [39]. Conversely, genome-wide association studies (GWAS) allow for agnostic interrogation of genetic variation across the genome for association with a trait. These hypotheses-free strategies can uncover novel pathogenic mechanisms, potentially leading to new therapeutic targets [40] (Figure 1B). Most genetic association studies have investigated single nucleotide polymorphisms (SNPs), which are base substitutions at a single position in the genome sequence. Although rare genetic variation (<1% minor allele frequency) may be implicated in the pathophysiology of AEs, whole-genome or exome association studies are yet to be conducted for AEs (Figure 1C).
In populations resulting from the admixture of two or more ancestral populations, admixture mapping analysis can be an alternative strategy to avoid the high penalty of statistical significance in GWASs, particularly in genetically complex populations often underrepresented in biomedical research [41–43]. Briefly, differences in the number of copies of alleles inherited from distinct ancestral populations at a given locus, or “local ancestry”, can be leveraged to distinguish candidate regions where local ancestry is associated with a trait of interest (Figure 1D). Genetic variants within the biologically plausible candidate region are then interrogated for association with the trait to identify causal variants that usually evince distinct allele frequencies between ancestral populations [41,42,44].
Since the most frequently used approaches to identify susceptibility alleles for AEs are biased to uncover variants with modest effect sizes (i.e., candidates gene studies) or located in non-coding regions of the genome (i.e., GWAS), it is imperative to comprehensively assess the functional impact of genetic variation [1,45]. In this context, investigating the effect of variants on different -omic layers (Figure 1E) has become easier, given the availability of multiple free tools online (e.g., see [46–49]).
Candidate-gene association studies.
Most candidate-gene association studies of AEs focused on polymorphisms in genes previously implicated in asthma or in viral pathways [1], such as the Interleukin 33 (IL33) [50], vitamin D receptor (VDR) [54], or SERPINE1 gene encoding for the plasminogen activator inhibitor-1 (PAI-1) [52] (Table 1). For instance, adding several asthma-related variants at SPATS2L and IL33 that were associated with ED management failure to ED-related clinical scores improved the predictive capability for ED management failure compared with the clinical model alone (area under the curve [AUC]: 0.82 vs. 0.79, p = 0.0004) [50]. Moreover, an expression quantitative trait loci (eQTL) analysis of respiratory syncytial virus (RSV)-related genes narrowed down the modulatory effect of RSV infection on a CEACAM3 locus for AEs [53]. Likewise, a candidate-gene association study of six genomic regions harbouring genes whose combined sputum gene expression signature exhibited predictive capability for exacerbations uncovered a DNASE1L3 locus for AEs associated with DNASE1L3 transcript expression levels in asthma-related tissues [56].
Table 1.
rsID (Gene) | Subjects | Phenotype | EA/EG | Effect size (95%CI) or (SE) | P | PMID [reference] |
---|---|---|---|---|---|---|
| ||||||
rs295137 (SPATS2L) | 491 European children with moderate-to-severe asthma presenting to the ER | Hosp/active asthma management ≥8 h in ER after OCS/return visit within 72 h for one of events after presenting to the ER | T | OR: 1.77 (1.17, 2.68) | 0.006 | 30644648 [50] |
rs7037276 (IL33) | T | OR: 0.55 (0.33, 0.90) | 0.02 | |||
rs1342326 (IL33) | C | OR: 0.52 (0.32, 0.86) | 0.01 | |||
| ||||||
rs16986718 (NLRP4) | 1-year longitudinal study of 1,454 Korean subjects with asthma, including 955 never-smokers | Number of ER visits/Hosp/OCS/rescue bronchodilator/increase of asthma medication | G | Increased annual exacerbation episodes | 0.001 | 30526007 [51] |
≥2 exacerbations events vs one/none | OR: 2.56 (NA) | 6.7×10−5 | ||||
SNP × pack/years on exacerbations | NA | 0.014 | ||||
| ||||||
rs1799768 (SERPINE1) | 265 subjects (48% European, 43% African/African American) with poorly controlled asthma enrolled in a randomized clinical trial of soy isoflavones. | OCS | 4G4G/4G5G vs 5G/5G | IRR: 2.57 (1.09–6.07) | 0.031 | 30707970 [52] |
Soy isoflavones intake×SNP on OCS | NA | 0.005 | ||||
Number of OCS events/person-year | RR: 0.28 (0.12, 0.59) | <0.001 | ||||
| ||||||
rs7251960 (CEACAM3) | Discovery: 456 Taiwanese children with asthma Replication: 844 children with mild-to-moderate asthma recruited in the United States |
Discovery: Nocturnal wheezing/ cough or wheezing/dyspnea in the last 2 weeks. Replication: Asthma-related limitation of activity/wheezing-induced nocturnal awakenings at least once a month in the last 2 months |
CT/TT vs CC | ORDiscovery: 2.58 (1.65, 4.03) ORReplication: 1.53 (1.03,2.31) |
3.12×10−5; 0.035 | 32606071 [53] |
RSV × SNP on exacerbation. | NA | 0.03 | ||||
| ||||||
rs1544410 (VDR) | 657 Australian children (64.2% European) presenting to the ER with acute asthma/wheezing/ bronchiolitis | Exacerbation severity Z-scores | TT vs CC | Increased severity scores | 0.005 | 32380236 [54] |
rs2228570 (VDR) | Exacerbation severity Z-scores | AA vs GG | Increased severity scores | 0.001 | ||
Lifetime Hosp for acute respiratory illnesses | AG vs GG | Increased severity scores | 0.011 | |||
| ||||||
ADRB2 variants | 832 children with asthma | Hosp/ER visits/OCS in the last 6–12 months despite ICS plus LABA use | Arg16/Gln27 vs Gly16/Glu27 | 1.40 (1.05,1.87) | 0.022 | 34128573 [55] |
Arg16/Gln27 vs Gly16/Gln27 | 1.43 (1.05–1.94) | 0.023 | ||||
| ||||||
rs67622929 (DNASE1L3) | Discovery: 1,002 African American subjectss with asthma. Replication: 2,181 Hispanic/Latino children with asthma | Hosp/ER visits/OCS in the last 12 months | C | ORDiscovery: 1.48 (1.18,1.87) ORReplication: 1.18 (1.01,1.37) |
7.9×10−4; 0.03 | 33035569 [56] |
| ||||||
rs11681246 (LTBP1) | 2,681 European children | Hosp/ER visits/OCS in the last 6–12 months despite ICS use | G | OR: 0.72 (0.63, 0.83) | 3.28×10−6 | 32786158 [57] |
rs76390075 (LTBP1) | 1,347 Hispanic/Latino or African American children with asthma | C | OR: 0.40 (0.26, 0.63) | 6.76×10−5 | ||
| ||||||
rs2660845 (LTA4H) | Patients with asthma. Discovery: 523 Europeans with early-onset asthma R1: 2,514 Europeans with early-onset asthma R2: 486 Hispanic/Latino children R3: 71 African American children |
Hosp/ER visits/OCS in the last 6–12 months despite montelukast use | G | ORDiscovery: 2.92 (1.04,8.18) ORR1: 1.02 (0.87,1.19) ORR2: 1.04 (0.78,1.39) ORR3: 0.27 (0.09,0.80) |
0.041 R1: 0.833 R2: 0.788 R3: 0.019 |
34550981 [58] |
| ||||||
rs2517955 (PGAP3) | 3-year longitudinal study of 273 non-Hispanic white adolescents and adults with asthma | Number of asthma-related ER visits/Hosp in 3 years | C | β: 1.05 (NA) | 0.0034 | 32795586 [59] |
rs1031458 (GSDMB) | G | β: −0.77 (NA) | 0.028 | |||
rs3902920 (GSDMB) | T | β: −0.88 (NA) | 0.012 | |||
| ||||||
rs13431828 (IL1RL1) | 2,412 European, Hispanic/Latino or African American children | Hosp/ER visits in the last 6–12 months despite ICS use | C | OR: 1.32 (1.08, 1.62) | 0.02 | 31755552 [60] |
OR: 1.31 (1.07, 1.59) | 0.02 | |||||
| ||||||
rs242941 (CRHR1) | European adult patients with asthma (nDiscovery=597; nReplication: 9,842) | Hosp/ER visits/OCS despite ICS use | A | RRDiscovery: 6.11 (NA) RRReplication: 1.16 (NA) |
<0.005; 0.004 | 33428814 [61] |
rs1134481 (TBXT) | T | RRDiscovery: 0.36 (NA) RRReplication: 1.02 (NA) |
<0.005; 0.563 | |||
rs37973 (GLCCI1) | G | RRDiscovery: 1.88 (NA) RRReplication: 0.82 (NA) |
<0.005; <0.005 | |||
| ||||||
rs1384006 (OXSR1) | 1-year longitudinal study of 1,454 Korean subjects with asthma, including 955 never-smokers | Number of ER visits/Hosp/OCS/rescue bronchodilator/increase of asthma medication | C | Increased annual exacerbation episodes | 0.004 | 34983467 [62] |
≥2 exacerbations events vs one/none | OR: 0.36 (0.18, 0.72) | 0.004 | ||||
| ||||||
rs9665961 (BIRC3) * | 5,710 European adults, 166 European children, 854 Hispanic/Latino children and 493 African American children with asthma | Hosp/ER visits/OCS in the last 6–12 months despite ICS use/8% decrease in FEV1 in patients after 6 weeks of ICS therapy | A | OR: 0.81 (NA) | 3.77×10−4 | 34971648 [63] |
A total of 35 SNPs in linkage disequilibrium (r2≥0.8) were significantly associated with AEs on the sample size weighted meta-analysis based on p-values. For visual clarity, the most significant variant in the meta-analysis is shown here, accompanied by the odds ratio corresponding to the largest cohort contributing to the rs9665961 genotype data on the multi-ancestry meta-analysis. Abbreviation: β: Regression coefficient; CI: Confidence interval; EA/EG: Effect allele/genotype; ER: Emergency room; FEV1: Forced expiratory volume in the first second; Hosp: Hospitalizations; ICS: Inhaled corticosteroids; IRR: Incidence risk ratio; LABA: Long-acting beta2-agonists; NA: Not available; OCS: Oral corticosteroids use; OR: Odds ratio; RR: Relative risk; Rn: Replication study (number n); RSV: Respiratory syncytial virus; SE: Standard error of the beta coefficient; SNP: Single nucleotide polymorphism; P: P-value.
The first GWAS of asthma revealed variants at chromosome 17q12–21 with larger effects on asthma in children than adults [64], whose effects may be intensified by early-life and passive tobacco smoke exposure [65,66]. As expected for the most consistently replicated signal for asthma across populations, genetic variation and gene-by-environment (GXE) interactions for chromosome 17q12–21 have been investigated in relation to AEs [1,67]. More recently, the effect of GSDMB SNP rs7216389 on AEs was found not to be modulated by prenatal second-hand smoke exposure in Danish children [68]. Several 17q12–21 variants are associated with expression levels of nearby genes in bronchial epithelial cells and located within binding sites for Interferon regulatory factors, suggesting effects through antiviral pathways [59], consistent with previous GxE evidence on asthma susceptibility [67].
Two recent studies of Korean subjects with asthma revealed genetic associations for AEs in NLRP4 and OXSR1 that differed by smoking status [51,62]. NLRP4 is a regulator of the inflammasome acting as an inhibitor of type I interferon signalling, tumor necrosis factor (TNF)-α and IL-1β-mediated NF-κB activation [69]. Conversely, OXSR1 encodes an oxidative stress responsive kinase that participates in ion transport and cell volume homeostasis [62]. In fact, OXSR1 expression was increased by smoke exposure and glucocorticoid treatment in various airway cell types [62].
In the reviewed period, four candidate gene studies have explored susceptibility variants for response to inhaled corticosteroids (ICS) [60,61,63,70], long-acting beta2-agonists [55], or montelukast [58] using AEs as a clinical endpoint. Four of them assessed asthma-related genes: IL1RL1 [60] and CRHR1 [61] for ICS, ADRB2 [55] for LABA, and LTA4H [58] for montelukast response. The other two combined or integrated multiple omics to prioritize candidate genes [63,70]. Hernandez-Pacheco et al. [57] identified a member of the family of latent-transforming growth factor-beta binding proteins (LTPB1) as differentially expressed after glucocorticoid exposure in several transcriptomic datasets from airway smooth muscle cells and peripheral blood mononuclear cells. Within LTBP1, two polymorphisms associated with AEs exerted ethnic-specific effects [70]. Kan et al. [63] leveraged a previous GWAS of change in forced expiratory volume in one second (FEV1) after ICS treatment (p<1×10−4), chromatin immunoprecipitation sequencing (ChIP-Seq), and transcriptomics to develop a multi-omics integrative score that prioritized a locus harbouring a member of the family of inhibitor of apoptosis proteins (BIRC3) near glucocorticoid receptor-binding sites. The BIRC3 locus was significantly associated with AEs -despite ICS use- in Hispanics/Latinos, African Americans, and Europeans [63].
GWAS.
Six non-pharmacogenomic GWAS of AEs were published in the reviewed period (Table 2). An asthma-related HLA-DQB1 locus was associated with AEs in British adults and Hispanic/Latino children, possibly through regulatory effects over HLA genes [71]. In European children, a study comparing participants with AEs and individuals without asthma revealed a novel genome-wide signal at FUT2/MAMSTR, along with several previously asthma-related loci, as expected of such a comparison strategy [74]. Interestingly, the epistasis of a functional FUT2 SNP with an ABO SNP increased the risk of respiratory infection with Streptococcus pneumoniae [74].
Table 2.
Type of study | rsID (Gene) | Subjects | Phenotype | EA | Effect size (95%CI) or (SE) | P | PMID [reference] |
---|---|---|---|---|---|---|---|
| |||||||
GWAS | rs56151658 (HLA-DQB1) | Discovery: 34,167 white British adults with asthma Replication: 2,645 Hispanic/Latino children with asthma |
ER/Hosp/OCS | A | ORDiscovery:1.36 (1.22, 1.52) ORReplication: 1.19 (0.99, 1.42)*† |
3.11×10−8 5×10−3* |
32890573 [71] |
| |||||||
GWAS | rs2253681 (LINC03033) | 4,010 Hispanic/Latino youth with asthma | ER/Hosp/OCS | A | OR: 1.55 (1.34,1.79) | 6.3×10−9 | 33093117 [72] |
| |||||||
GWAS | rs4952375 (LINC01913) | Children with asthma. Discovery: 3,310 Hispanics/Latinos; replication: 1,043 African Americans. | ER/Hosp/OCS | A | ORDiscovery: 1.37 (1.20,1.55) ORReplication: 1.53 (1.12 ,2.08) |
1.24×10−6; 7.43×10−3 | 32841424 [73] |
| |||||||
GWAS | rs721992318 (GSDMB) | Discovery: 2,866 European children experiencing severe AE between ages 2 and 6 years, and 65,415 non-asthmatic controls. Replication: 1,118 children. |
Discovery: Asthma with Hosp; Replication: Asthma | T | OR: 1.65 (1.56,1.75) | 1.6×10−68 | 33328473 [74] |
rs696733010 (CDHR3) | A | OR: 1.41 (1.32,1.51) | 2.1×10−23 | ||||
rs107163018 (HLA-DQA1) | C | OR: 1.25 (1.18,1.32) | 8.0×10−14 | ||||
rs34093366 (IL33) | G | OR: 1.37 (1.26,1.49) | 1.6×10−13 | ||||
rs134232666 (IL33) | C | OR: 1.31 (1.22,1.40) | 1.7×10−13 | ||||
rs1018962918 (IL1RL1) | C | OR: 1.40 (1.27,1.54) | 7.7×10−12 | ||||
rs104382818 (WDR36) | C | OR: 1.20 (1.14,1.27) | 1.0×10−10 | ||||
rs2054118 (IL13) | A | OR: 1.21 (1.13,1.29) | 1.0×10−8 | ||||
rs281379 (FUT2/MAMSTR) | G | ORDiscovery 1.18 (1.11,1.25) ORReplication: 1.43 (1.16,1.79) |
2.6×10−9; 1.1×10−3 | ||||
| |||||||
GWAS | rs10519519 (MYT1L) | 1-year longitudinal study of 20 non-smoking and 188 smoking Korean patients with asthma | Annual rate of episodes of increased dyspnea, wheezing, or coughing with a >20%decrease in FEV1 |
A | β: 0.60 (0.11) | 8.32×10−7 | 35606283 [75] |
| |||||||
GWAS | rs12091010 (VCAM1/EXTL2) | Discovery (4,989 subjects with asthma): 53.1% Europeans, 23.2% Hispanics/Latinos, 13.3% Singaporean Chinese, and 10.3% African Americans. Replication: 36,477 European and 1078 non-European asthma patients |
ACC/ER/Hosp/OCS/SA | T | ORDiscovery: 0.82 (0.75–0.90) ORReplication: 0.89 (0.82–0.97) |
9.05×10−6; 5.35×10−3 | 35754128 [76] |
rs943126 (PANK1) | Children with asthma. Discovery: 854 Hispanic/Latino, 493 African Americans. Replication: 1,697 Europeans | ER/Hosp/OCS despite ICS use | C | ORDiscovery: 0.85 (0.78,0.92) ORReplication: 0.92 (0.86,0.98) |
3.10×10−5; 1.30×10−2 | ||
| |||||||
PGWAS (ICS) | rs5995653 (APOBEC3B/APOBEC3C) | Children with asthma. Discovery: 854 Hispanic/Latino and 493 African Americans. Replication: 1,697 Europeans | ER/Hosp/OCS despite ICS use | A | ORDiscovery 0.76 (0.62,0.93) ORReplication: 0.66 (0.56,0.79) |
4.80×10−6; 7.52×10−3 | 30697902 [77] |
rs62081416* (L3MBTL4/ARHGAP28) | 854 Hispanic/Latino and 493 African American children with asthma | A | OR: 2.44 (1.63,3.65) | 1.57×10−5 | |||
| |||||||
PGWAS (ICS) | rs3827907 (EDDM3B) | Patients with asthma. Discovery: 244 African Americans. Replication: African Americans (nR1=803 and nR2=563) and Latinos (nR3=1,461) |
Discovery: SNPxICS adherence on change in ACT score over 6 weeks of ICS treatment. Replication 1: SNPxICS adherence on time to ER/Hosp/OCS. Replication 2–3: SNPxICS use on ER/Hosp/OCS |
C | CoefDiscovery12.35 (NA) CoefR1: -0.07 (NA) CoefR2: 0.15 (NA) CoefR3: 0.96 (NA) |
7.79×10−8; 0.023; 0.029; 0.041 | 30367910 [78] |
| |||||||
PGWAS (LABA) | rs1947048 (EPHA7) | 1,425 children and young adults with asthma (23% Hispanic/Latino, 10.4% African American, 32.5 Singaporean Chinese) | ER/Hosp/OCS despite LABA use | G | OR: 2.50 (1.69, 3.69) | 4.36×10−6 | 33706416 [79] |
PGWAS (LABA) | rs6489992 (TBX3) | A | OR: 1.77 (1.40, 2.23) | 4.96×10−6 | |||
| |||||||
PGWAS (ICS) | rs67026078 (CACNA2D3/WNT5A) | Children with asthma. Discovery: 2,681 Europeans. Replication 1: 538 Europeans. Replication 2: 854 Hispanic/Latinos, 493 African Americans, 426 Singaporean Chinese | ER/Hosp/OCS/SA despite ICS use | C | ORDiscovery: 1.50 (0.93, 2.43) ORR1: 1.83 (1.16, 2.90) |
4.22×10−6; R1; 0.01; R2: NS | 33303529 [80] |
| |||||||
PGWAS (ICS) | rs1166980 (ROBO2) | Children with asthma. Discovery: 166 Europeans. Replication 1: 2,681 Europeans. Replication 2: 854 Hispanic/Latinos, 493 African Americans |
Discovery: ≥8% in FEV1 after 6 weeks of ICS treatment Replication: ER/Hosp/OCS/SA despite ICS use |
G | ORDiscovery: 7.01(3.29, 14.93) | 4.61×10−7, R1-R2: NS | 34442380 [81] |
PGWAS (ICS) | rs72891545‡ (ROBO2) | Discovery: 2,681 Europeans | ER/Hosp/OCS/SA despite ICS use | A | OR: 4.79 (2.36, 9.73) | 1.44×10−5 | |
| |||||||
PGWAS (ICS) | rs138717703 (RBMXP1/PTCHD4) | European adults with asthma (nDiscovery= 5,710; nReplication=1,141) | OCS despite ICS use | G | ORDiscovery: 1.73 (1.39, 2.16) ORReplication: 1.48 (0.75, 2.90)‡ |
7.91×10−7; 5.78×10−4 | 35501119 [82] |
PGWAS (ICS) | rs77506063 (RBMXP1/PTCHD4) |
C | ORDiscovery: 1.73 (1.39, 2.16) ORReplication: 1.48 (0.75, 2.90)‡ |
7.91×10−7; 5.78×10−4 | |||
PGWAS (ICS) | rs145325916 (RBMXP1/PTCHD4) | C | ORDiscovery: 1.74 (1.40, 2.16) ORReplication: 1.27 (0.66, 2.42)‡ |
6.18×10−7; 3.56×10−3 | |||
PGWAS (ICS) | rs116023293 (HNRNPA3/P4 PTCHD4) |
G | ORDiscovery: 1.74 (1.40, 2.16) ORReplication: 1.27 (0.66, 2.42)‡ |
5.28×10−7; 3.56×10−3 | |||
| |||||||
PGWIS (Age; ICS) | rs34631960 (THSD4) | 1,321 adult and child Europeans with asthma | SNPxAge use on ER/Hosp/OCS | C | ORDiscovery: 2.33 (1.61, 3.38) ORReplication: 1.82 (1.23, 2.7); |
7.08×10−6; 2.97×10−3 | 32119686 [83] |
PGWIS (Age; ICS) | rs2328386 (HIVEP2) | T | ORDiscovery: 0.33(0.2, 0.55) ORReplication: 0.51(0.34, 0.77) |
1.86×10−5; 1.49×10−3 |
rs56151658 was not available in Hispanics/Latinos. The results for the most significant proxy in Latinos (r2=0.71) are shown (rs9275356).
Effect size from the largest replication cohort is shown.
The genetic variant was identified using a candidate-gene approach using the GWAS summary statistics. Abbreviations: ACC: Acute asthma care; ACT: Asthma Control Test; Coef: Interaction coefficient estimate; CI: Confidence interval; EA: Effect allele; ER: Emergency room visits; FEV1: Forced expiratory volume in the first second; Hosp: Hospitalizations; GWAS: Genome-wide association study, ICS: Inhaled corticosteroids; LABA: Long-acting beta-agonists; OCS: Oral corticosteroids use; NA: Not available; NS: Non-significant; PGWAS: Pharmacogenomic GWAS (asthma treatment considered is shown within parenthesis); PGWIS: Pharmacogenomic genome-wide interaction study (tested environmental variable and asthma treatment considered are shown within parenthesis); Rn: Replication study (number n); SA: School absences; SE: Standard error of the beta coefficient; SNP: Single nucleotide polymorphism; P: P-value.
Most genetic association studies of AEs have been conducted in Europeans [16], but recent substantial efforts have increased ethnic diversity and representation [72,73,75–77]. As a result, the largest GWAS meta-analysis of severe AEs in Hispanics/Latinos children (n=4,010) uncovered a genome-wide significant association in LINC03033, a long non-coding RNA (lncRNA) that participates in myofibroblasts differentiation and airway remodelling [72]. The risk allele for AEs was associated with higher DNA methylation (DNAm) levels at LINC03033 in nasal epithelium, which in turn was associated with higher expression of KCNJ2-AS1 [72], also overexpressed in atopic asthma [84]. Another recent GWAS compared asthma cases with AEs to controls without asthma to overcome the reduced statistical power derived from the complex genetic structure in recently-admixed populations in order to identify genetic signals for asthma with AEs in Hispanic/Latino and African American children [73]: a genome-wide significant locus nearby lncRNA LINC01913 was associated with asthma with severe exacerbations, possibly through LINC01913 expression in lung and DNAm of PKDCC in blood. While LINC01913 function remains unknown, PKDCC is involved in lung development and mediates various homeostatic cellular processes [73]. Likewise, an intronic variant in the MYT1L gene, encoding for a regulator of proteins of the nervous system, was associated with the annual number of AEs in Koreans [75]. More recently, a multi-ancestry meta-analysis of GWAS of AEs identified two suggestive variants associated with blood DNAm or expression levels of genes participating in inflammation and host defence (VCAM1, EXTL2, and PANK1) [76].
Pharmacogenomic GWAS (PGWAS) of AEs have identified susceptibility variants for response to ICS [1,77,80,82,85] and LABA [79]. Genomic regions harbouring the loci suggestively associated with AEs in children receiving ICS are implicated in response to viral infections (APOBEC3B/APOBEC3C [77]), baseline lung function (CACNA2D3 [86]), bronchodilator responsiveness (CACNA2D3 [87]), or the Wingless/integrase 1 signalling (WNT5A) pathway [80]. Moreover, two studies considered AE despite ICS use as a secondary outcome to validate genetic associations for ICS response at EDDM3B [78] and ROBO2 [81]. While the EDDM3B and APOBEC3B/APOBEC3C variants exerted similar effects across several ethnic backgrounds [77,78]), CACNA2D3/WNT5A and ROBO2 loci exhibited effects specific in children of European descent [80,81].
In addition, a recent study in older adults of European ancestry with asthma treated with ICS uncovered 152 suggestive associations for AEs defined using diagnosis codes, and a genome-wide signal for oral corticosteroid (OCS) use nearby PTCHD4, which encodes a regulator of hedgehog signalling previously associated with airway disease [82]. Furthermore, a genome-wide interaction study (GWIS) of age on AEs despite ICS use found genome-wide significant signals in genes implicated in angiogenesis, lung function and chronic obstructive pulmonary disease (COPD) (THSD4), inflammatory and immune processes, and glucocorticoid response (HIVEP2) [83]. Moreover, the only multi-ancestry meta-analysis of GWAS of AEs despite LABA use discovered suggestive associations within genes previously implicated in lung function (TBX3 [88]) and response to short-acting beta2-agonists (EPHA7 [89]).
As previously indicated, some studies aimed to shed light into the role of genetic variation by assessing their functional and biological impact. For instance, CACNA2D3/WNT5A and ROBO2 variants were associated with the expression of proteins involved in asthma pathophysiology in plasma [80,81]. Gene-level analysis stratified by smoking status in Koreans revealed that significant genes in non-smokers were enriched for T-cell immune responses and DNA/RNA modifications, while tissue development and apoptosis were the most important processes in smokers [75].
Genetic variants associated with AEs despite ICS use in European adults are enriched in genes implicated in protein and fatty acid metabolism, toll-like receptor signalling, antigen cross-presentation, or vesicular transport [82]. Among European children treated with ICS, genetic variants associated with AEs were enriched in asthma-related genes that showed differential expression under trichostatin A exposure [80]. Trichostatin A is an antifungal antibiotic with histone deacetylase activity that has been shown to reduce airway inflammation and hyperresponsiveness [90]. Interestingly, histone deacetylase participates in the regulation of corticosteroid sensitivity [91]. Overall, these findings support further investigation of the therapeutic potential of trichostatin A in asthma.
Admixture mapping.
Admixture mapping studies have identified genetic variants associated with asthma, IgE levels, bronchodilator response, and lung function [42,94], but only two admixture studies of AEs have been published (Table 3). The only admixture mapping of AEs independent of treatment conducted in Hispanics/Latinos revealed significant associations for AEs with Indigenous American ancestry at chromosomal regions 5q32, 13q13-q13.2, and 3p13. The 5q32 SNP rs1144986 (C5orf46) was significantly and consistently associated with AEs in Mexican Americans and Puerto Ricans, but was not validated in non-Hispanics/Latinos. The risk allele of rs1144986 was associated with altered DPYSL3 DNAm levels and lower gene expression of SCGB3A2 in blood. While DPYSL3 may be involved in airway remodelling, SCGB3A2 is an upstream regulator of TGFβ-mediated antifibrotic processes in the lung [92].
Table 3.
rsID (Gene) Chromosomal band |
Subjects | Phenotype | EA | OR (95%CI) | P | PMID [reference] |
---|---|---|---|---|---|---|
| ||||||
rs1144986 (C5orf46) 5q32 |
Patients with asthma. Discovery: 625 Mexican Americans. R1: 1,124 Puerto Ricans R2: 1,001 African Americans, 1,250 Singaporeans, and 941 Europeans. |
ER/Hosp/OCS | G | Discovery: 0.43 (0.28, 0.66) R1: 0.79 (0.62, 1.00) |
9.45×10−5; 4.94×10−2; NS |
36180068 [92] |
| ||||||
rs5752429 (TPST2) 2q12.1 |
Discovery: 266 adolescents/adults of African-descent ancestry Replication: 222 African Americans |
Discovery: Better response to 5xICS vs 2.5xICS Replication: ER/Hosp/OCS despite high ICS use |
A | Discovery: 0.21 (0.09, 0.52)* Replication: 2.28 (1.33, 3.90) |
6×10−4; 0.003 |
34762840 [93] |
rs73399224 (RNFT2) 12q24.22 |
Discovery: 250 children of African-descent ancestry Replication: 379 African Americans |
Discovery: Better response to 5xICS vs 100 μg fluticasone plus salmeterol Replication: ER/Hosp/OCS despite ICS use |
G | Discovery: 0.17 (0.07, 0.42)* Replication: 1.97 (1.07, 3.64) |
8.0×10−5; 0.03 |
Another study investigated the association of local ancestry with response to different step-up regimens including ICS in 516 subjects with asthma of African descent [93]. The primary outcome was a composite score comprising AEs, a 31-day difference in annualized asthma-control days, and a 5% difference in percent predicted FEV1. African ancestry at 12q24.22-q24.23 was associated with better responsiveness in children that transitioned from low-dose ICS to the quintuple dose of ICS compared to those who received 100 μg fluticasone plus salmeterol. Moreover, African ancestry at chromosome 22q12.1 was associated with better responsiveness in adults that transitioned from low-dose ICS to the quintuple dose of ICS compared with those who received 2.5 times the ICS dose. Analysis of genetic variants within these regions revealed one SNP that was consistently replicated for association with AEs in African Americans treated with ICS [93].
EPIGENETICS
The three main epigenetic mechanisms that can act synergistically to regulate gene expression are DNAm, histone modifications, and non-coding RNAs such as micro-RNAs (miRNA). However, histone modifications have been investigated for asthma [95] but not for AEs.
DNAm.
DNAm consists of the addition of a methyl group to a cytosine, often within 5′-cytosine-phosphate-guanine-3′ dinucleotide sequences (or CpG sites). DNAm levels have been associated with disease risk and health outcomes, including asthma and allergy [96]. Most CpGs in the human DNA methylome are hypermethylated and located in regions of low CpG density but CpG-rich regions, known as CpG islands, are often hypomethylated. While promoter DNAm usually leads to reduced gene expression, gene body DNAm is associated with active transcription [97].
Although DNAm is the most extensively studied epigenetic mechanism, only one targeted DNAm study of AEs [98] and one epigenome-wide association study (EWAS) [99] of AEs as a proxy of treatment response have been published (Table 4). Curtin et al. [98] found that increased cord blood DNAm of the IL2 promoter was associated with AEs and hospitalizations for asthma or wheezing later in childhood. On the other hand, Wang et al. [99] conducted a multi-ancestry EWAS meta-analysis of peripheral blood CpG markers and AEs despite ICS treatment. Hypomethylation of cg00066816 upstream of IL12B, which encodes for a subunit of the heterodimeric IL-12, a pro-inflammatory cytokine involved in Th1 and Th17 signalling [100], was nominally associated with the absence of asthma-related ED visits or hospitalizations in the previous year in children on ICS. In a secondary analysis, 13 CpGs were differentially methylated in subjects who received OCS bursts in the past year despite ICS use. Although functional effects of DNAm over blood gene expression were explored, the CpG-gene pairs were not consistently replicated across studies [99].
Table 4.
Biological sample | Subjects | Phenotype | CpG | Gene/Nearest Gene | Regression coeficient (95%CI) | P | PMID [reference] |
---|---|---|---|---|---|---|---|
| |||||||
Cord blood | 303 children recruited by a birth cohort in Manchester (UK) | Asthma-related hospitalizations or ER after the first year of life | Promoter site 1 | IL2 | 1.07 (1.01,1.14) | 0.03 | 23414538 [98] |
Asthma/wheeze-related hospitalization after the first year of life | 1.12 (1.04,1.20) | 0.002 | |||||
| |||||||
Blood | 394 children treated with ICS (57.4% Europeans, 42.6% Hispanic/Latino) | Asthma-related hospitalizations or ER in the past year despite ICS use | cg00066816 | IL12B | −3.10 (NA) | 0.002 | 31187518 [99] |
OCS use in the past year despite ICS use. | cg00557354 | ARHGEF7 | −3.49 (NA) | 0.001 | |||
cg04256470 | CORT, CENPS | 3.62 (NA) | <0.001 | ||||
cg09495977 | HTRA3 | −2.42 (NA) | 0.017 | ||||
cg12333095 | ANKRD13A | −3.48 (NA) | 0.001 | ||||
cg13818573 | C1QL1 | −3.59 (NA) | <0.001 | ||||
cg21589280 | DDAH1 | −3.06 (NA) | 0.003 | ||||
cg03080985 | SH3BGRL2 | −3.07 (NA) | 0.003 | ||||
cg04330449 | NEUROG1 | −2.64 (NA) | 0.009 | ||||
cg05307923 | ADARB2 | −2.57 (NA) | 0.011 | ||||
cg08724517 | MAP9 | 2.95 (NA) | 0.004 | ||||
cg11665562 | PSMC1 | −3.25 (NA) | 0.001 | ||||
cg14269514 | OAZ3, MRPL9 | −3.11 (NA) | 0.002 | ||||
cg24322623 | MYOD1 | −2.96 (NA) | 0.004 |
Abbreviations: CI: Confidence interval; ER: Emergency room visits; ICS: Inhaled corticosteroids; OCS: Oral steroid bursts; NA: Not available; P: P-value; UK: United Kingdom.
miRNAs.
miRNA are post-transcriptional regulators that exert their effects by binding to the 3′ untranslated regions of mRNAs, leading to mRNA deadenylation and subsequent degradation. These small non-coding molecules are implicated in the regulation of multiple cellular processes and have recently gained attention in allergic and chronic lung diseases [101–103].
To our knowledge, seven studies have addressed the role of miRNAs in AEs, including three studies in blood, three in serum, and one in induced sputum (Table 5). All studies focusing on circulating miRNAs applied single marker approaches, while the study that performed miRNA profiling in induced sputum carried out a systems biology approach. Specifically, Gomez et al. [109] conducted a weighted gene co-expression network analysis (WGCNA) of miRNA and mRNA expression levels in induced sputum from 61 subjects with asthma. The analysis of 221 miRNAs revealed a 12-miRNA module directly correlated with asthma hospitalizations. In their cluster analysis, high expression levels of these 12 miRNAs were associated with neutrophilic inflammation, low T2 biomarkers, and airflow obstruction. Notably, the sputum 12-miRNA module correlated with mRNA modules implicated in the TLR9/Th17 signalling pathway and endoplasmic reticulum stress. One of the miRNA associated with high sputum neutrophil counts in response to ozone exposure, hsa-miR-223–3p, acted as a regulator of both of these two mRNA modules [109].
Table 5.
Biological sample | miRNA profiling | Subjects | Phenotype | Main findings | PMID [reference] |
---|---|---|---|---|---|
| |||||
Whole blood | Human MicroRNA v2.0 Assay Pool (Illumina). | Children with (n=100) and without asthma (n=100) recruited at a Turkish hospital | Asthma severity (GINA, 2008) and AEs severity | Increased expression of 10 miRNAs was associated with asthma severity and exacerbations severity: HS_108.1, HS_112, HS_182.1, HS_240, HS_261.1, HS_3, HS_55.1, HS_91.1, hsa-mir-604, and hsa-mir-638. | 26422695 [104] |
| |||||
Peripheral blood | q-PCR | Children with acute-stage asthma (n=100) and healthy children (n=100) recruited at a Chinese hospital | Acute asthma attacks (not defined) | miR-1 expression levels were reduced in acute-stage asthma compared with controls. miR-1 expression levels improved prediction of acute asthma attacks compared with IL-4, IL-5, IL-8, and TNF-α in the same population. |
30046607 [105] |
| |||||
Serum | q-PCR | Subjects with (n=59) and without asthma (n=11) recruited in the United States | Lifetime and past 12 months frequency of asthma-related hospitalizations | miR-1 levels were inversely correlated with sputum eosinophilia and asthma-related hospitalization frequency, and positively correlated with lung function and ACT scores. | 32035607 [106] |
| |||||
Serum | q-PCR | European children with asthma from CAMP: 38 with and 115 without exacerbations | OCS bursts in the past 12 months following randomization with ICS | Increased expression of 12 miRNAs was associated with OCS bursts: miR-206, miR-146b-5p, miR-222–3p, miR-409–3p, miR-223–5p, miR-126–5p, miR-339–3p, miR-30e-3p, miR-126–3p, miR-342–3p, miR-454–3p, and miR-720. A clinical and 3-miRNA model (miR-146b, miR-206, and miR-720) showed higher AUC for prediction of OCS use compared with the clinical model in the same population (AUC: 0.81 vs 0.67). |
29940952 [107] |
| |||||
Serum | q-PCR | 6-week longitudinal study of 21 adults with asthma recruited at a Polish hospital | Admission for an unplanned visit due to worsened symptoms of asthma accompanied by a decrease in ventilatory parameters | Reduced levels of miRNA-126a, miRNA-16, and miRNA-21 during the exacerbation compared with the follow-up visit. | 31743969 [108] |
| |||||
Induced sputum | Nanostring nCounter array v3.0a | Subjects with (n=62) and without asthma (n=9) recruited in the United States | Asthma-related hospitalizations in the past 12 months | A 12 miRNA.WGCNA module was directly correlated with asthma hospitalizations. Ten of these miRNA correlated significantly and consistently with sputum neutrophils, longer duration of asthma, decreased quality of life, impaired lung function, and/or increased BDR. The miRNA module correlated with a mRNA module enriched in genes participating in TLR/Th17 signalling and endoplasmic reticulum stress. | 32255668 [109] |
| |||||
Whole blood | Small-RNA sequencing | Costa Rican children with asthma (nFE=183; nIF=168) from GARCS | ≥3 events of asthma-related ER/AC visits and/or hospitalizations in the last 12 months (frequent exacerbations, FE) compared with no or infrequent exacerbation (IF) | 5 miRNA (miR-451b, hsa-miR-142–5p, hsa-miR-6739–3p, hsa-miR-7–5p, and hsa-miR-4433b-5p) were downregulated in FE compared with IF. 15 mIRNA (hsa-miR-93–3p, hsa-miR-766–3p, hsa-miR-331–3p, hsa-miR-532–3p, hsa-miR-664b-3p, hsa-miR-296–5p, hsa-miR-6515–3p, hsa-miR-4286, hsa-miR-1296–5p, hsa-miR-29b-2–5p, hsa-miR-500b-5p, hsa-miR-500a-5p, hsa-miR-642a-5p, hsa-miR-103a-2–5p, and hsa-miR-550a-3p) were upregulated in FE compared with IF. miR-532–3p, miR-296–5p, miR-766–3p, miR-7–5p, and miR-451b also showed significant association with COPD exacerbations. |
35447890 [110] |
Abbreviations: AC: Acute care; AUC: Area under the curve; CAMP: Childhood Asthma Management Program; COPD: Chronic obstructive pulmonary disease; ER: Emergency room; FE: Frequent exacerbations; GARCS: Genetics of Asthma in Costa Rica Study; GINA: Global Initiative for Asthma; ICS: Inhaled corticosteroids; IF: No or infrequent exacerbations; mRNA: Messenger RNA; OCS: Oral corticosteroids; q-PCR: Real-time quantitative polymerase chain reaction; RNA: Ribonucleic acid; Th17: T helper 17 cells; TLR: Toll-like receptor; WGCNA: Weighted gene co-expression network analysis.
Midyat et al. [104] reported that 10 of 739 tested miRNAs were differentially expressed by asthma and AE severity in children. Another study found that miR-1 is downregulated in acute-stage asthma and predicted asthma attacks with an AUC of 0.90, significantly higher than the AUC from asthma-related cytokines (e.g., IL-4 or IL-5) (p<0.05) [105]. Analysing animal models and primary human endothelial cells, miR-1 has been implicated in the regulation of airway eosinophilia through the inhibition of eosinophil binding to the endothelium by promoting RNA-induced gene silencing of eosinophil trafficking genes [106].
In a six-week longitudinal study, 3 of 7 circulating miRNAs tested were significantly lower during an AE episode than a follow-up visit: miRNA-126a, miRNA-16, and miRNA-21 [108]. Furthermore, miRNA-21 and miRNA-126a expression levels were positively correlated with FEV1%, whereas miRNA-21 levels were higher in participants with atopy or FeNO levels >25 parts per billion. MiRNA-126a and miRNA-21 are both considered promoters of Th2-mediated allergic inflammation [101,111], and miRNA-21 is a systemic oxidative stress marker dysregulated in the airways and/or blood in atopic dermatitis and allergic asthma [101].
In an analysis of subjects with frequent exacerbations and infrequent/no exacerbations, 20 of 649 tested blood miRNAs were differentially expressed by asthma [110]. In the COPDGene study, 5 of these 20 miRNAs were associated with COPD exacerbations, supporting some overlap in the pathogenesis of COPD and asthma. The gene targets of these 4 miRNAs participate in PI3K-Akt and MAPK signalling pathways [110], which are relevant in Th-2 inflammation and asthma pathogenesis [102].
Only one study has investigated the role of miRNAs in response to treatment using AEs as a clinical endpoint. In particular, miRNA profiling of serum samples was conducted in 153 children with asthma after randomization to ICS [107]. In univariate logistic regression models, 12 of the 125 tested serum miRNAs were significantly associated with OCS bursts in the previous year. Moreover, the combination of a clinical score for exacerbations along with 3 of these 12 miRNAs (miR-146b, miR-206, and miR-720) suggested a higher predictive capability for AEs compared with clinical score alone (AUC: 0.81 vs. 0.67) [107]. Of note, miR-146b-5b and miR-206 serum levels have been associated with asthma and COPD [103], as well as with baseline FEV1/FVC in individuals with asthma [112].
TRANSCRIPTOMICS
Studies of transcriptomics and AEs prior to 2022 were recently reviewed by Gautam et al. [37]. Such studies have identified distinct AE-related gene expression signatures implicated in innate and adaptative immunity, viral and non-viral exacerbations, and revealed genes implicated in frequent exacerbations (TNFR2) and in AEs triggered by colds (genes implicated in SMAD3 signalling pathways). Only one single-cell RNA-sequencing study in the context of AEs has been conducted, highlighting the implication of several cytokines and intracellular transduction regulators in multiple cell types in this trait [113].
Two transcriptomic studies of AEs have been published in the reviewed period. One focused on the interaction of transcriptional and bacterial networks in nasal epithelium on the risk of AEs in children [114]. Specifically, the risk of AEs increased along with the expression of genes implicated in SMAD3-related cell differentiation in a context of high abundance of a bacterial network dominated by Veillonella, Streptococcus, Neisseria, and Haemophilus and/or reduced abundance of a bacterial network dominated Staphylococcus [114]. Another study aimed to understand the pathophysiological factors underlying frequent exacerbations using transcriptomic data from bronchial biopsies. CEACAM5, encoding for a cell surface glycoprotein upregulated by interferon-gamma [115], was the only transcript differentially expressed in subjects with frequent exacerbations compared with those with infrequent exacerbations. However, no differential expression was found when subjects with persistent frequent exacerbations were compared with those with persistent infrequent exacerbations. An analysis of several gene signatures for viral infections and type 1 and type 2 inflammatory pathways revealed that subjects with frequent exacerbations had higher expression of those signatures than those with persistent frequent exacerbations [116].
CONCLUSION AND FUTURE DIRECTIONS
AEs constitute a major burden on individuals with asthma and their caregivers, healthcare systems, and society as a whole. Although preventing AEs is key in clinical practice, risk stratification of patients with AEs is challenging due to the inherent heterogeneity of the biological mechanisms underlying these events. Despite this, -omic studies have identified genes and biological processes associated with AEs and proposed potential therapeutic targets. These results need to be validated in independent cohorts and experimental studies, and much work remains to be done compared to -omics findings in other respiratory traits, such as asthma [37], or COPD [117–119].
Perhaps because of the heterogeneity of AEs, specific phenotyping approaches have been successful in identifying novel susceptibility variants [58,74,120]. Despite the increased statistical power derived from a large sample size, future studies should also consider analyzing subtypes or homogenous groups of individuals exposed to similar exacerbation triggers, which would allow the characterization of GxE interactions, almost unexplored in AEs [1]. An alternative approach to boost statistical power in recently admixed populations is to leverage local ancestry into GWAS models to increase the resolution of causal variant(s) identification [121]. Pointedly, differences in the definition of AEs, trigger agents, or clinical characteristics of individuals with asthma may have reduced statistical power in several GWASs [76,77,79,80,122]. This could also account, at least partially, for the lack of replication of SNPs associated with AEs across independent populations [76].
A combination of genetic variants into a single score of risk burden or polygenic risk score (PRS) for AEs is not feasible without additional risk stratification that also considers clinical and environmental parameters. Recently, multi-ancestral PRS for asthma developed using lasso sum [123] or Bayesian regression [124] have captured the risk of asthma, although other studies have failed to achieve this [125,126]. PRS incorporating DNAm or gene expression data may better capture environmental influences in order to improve risk stratification [127]. The extent to which methylation risk scores (MRS) or transcriptome risk scores (PTRS) may contribute to risk prediction is still to be determined, though promising findings have been published for other respiratory traits [128,129]. Within this context, it will be crucial to evaluate the predictive power of biomarkers in populations not included in the discovery phase or training datasets [130].
Similarly, the severity and number of AEs have a prognostic capability in risk-stratification [131], but only one GWAS of the annual number of exacerbations has been conducted [75], and no study has assessed temporal distance among events and/or the time-to-first exacerbation. Moreover, although bioinformatic tools have been used to evaluate the functional impact of potential susceptibility variants, many of these resources do not include data from tissues/cells obtained from asthma patients, across several asthma-relevant tissues, or diverse ethnic backgrounds. In this sense, experimental studies are required to understand the biological role of identified genes and establish their prognostic value to adequately implement precision medicine in patient risk stratification and prioritize potential therapeutic targets.
The role of rare variants in AEs has been poorly investigated, despite the fact that they may underlie ethnic/racial differences in the burden of AEs or interact with environmental exposures to modulate AEs. Furthermore, although ethnic diversity has increased in genetic studies of asthma, particularly for Hispanic/Latino populations, large-scale genome-wide studies of Asian and African-descent populations have not been implemented.
The contribution of the DNA methylome to AEs remains largely unexplored [99]. Thus, a priority in asthma epigenetics is to investigate the role of DNAm as a mediator of environmental effects or as a consequence of AEs, not only at the CpG level, but at differentially methylated regions (DMRs). Future research should also focus on the role of genetically-regulated DNAm and epigenetically-regulated gene expression. Furthermore, it will be key to consider that hypomethylation states in previous EWAS of asthma in blood were largely driven by a lower eosinophil count in blood [132], which is why specific cell types [132,133] or cell-type deconvolution algorithms to discern cell-type specific DNAm signals using whole-blood data [134] are gaining interest. Moreover, histone modifications have been implicated in asthma susceptibility and severity, ICS response, and immune responses to viral infections [95,135], yet little is known about histone modifications and AEs.
Most epigenetic asthma studies have focused on miRNAs in blood and serum, and validation of their findings is needed to exclude spurious results due to differences in sample processing [136,137]. Despite this concern, many miRNAs have been consistently implicated in chronic respiratory or allergic diseases, highlighting their potential as possible therapeutic targets (e.g., miR-206 and miRNA-21) [101–103]. Other plausible candidates to participate in AEs are miRNAs involved in airway inflammation or respiratory infections [101,102,138]. Undoubtedly, further exploration of the role of the miRNAome and its interaction with other -omics layers in the upper and lower airways is required to determine the role of altered miRNA expression in AEs.
Transcriptomic studies conducted in the reviewed period highlight important host-microbiome interactions in the upper and lower airways and open new directions for future research. Although McCauley et al. [114] found that the interaction of host gene expression levels and microbial networks in the upper airways promote AEs, the causative direction of those relationships is unclear. Still, they proposed several plausible candidate genes that could be evaluated in other cohorts. Interestingly, among individuals with frequent AEs, Hoda et al. [116] found increased CEACAM5 expression, which is also promoted by the interferon gamma [115].
Although there has been recent progress in genomic studies of AEs, the role and the interaction of different -omic layers in the modulation of the risk of AEs remain largely unexplored. In many cases, novel findings remain to be validated in independent populations, and their prognostic potential is unclear. Moving forward, multi-ethnic cohorts with better phenotyping of clinical and environmental characteristics, careful phenotyping approaches, evaluation of longitudinal exacerbation data, and combination or integration of different -omics layers of data will be crucial to identify accurate biomarkers of AEs for precision medicine.
Table 6.
Biological sample | RNA profiling | Subjects | Phenotype | Main findings | PMID [reference] |
---|---|---|---|---|---|
| |||||
Nasal blow | RNA-seq (NextSeq 500 platform) |
208 children with asthma from United States | OCS/Hospitalization | Increased SMAD3 expression among children with altered abundance of the two bacterial network was associated with increased exacerbation risk. | 35149044 [114] |
| |||||
Bronchial biopsies | GeneChip® Human Genome U133 Plus 2.0 Array (Affymetrix). | 317 participants with severe asthma from Europe | ≥2 events of systemic corticosteroids use vs (frequent exacerbators, FE) <2 events (infrequent exacerbators, IE) |
CEACAM5 expression was increased in FE compared with IE. Higher expression scores for viral infection gene signatures, type 1, T-helper type-17, and type 2 activation pathways in FE compared to IE. Higher expression scores of type 2, type 1 and steroid insensitivity pathway signatures in persistent FE compared to persistent IE. |
35474304 [116] |
Abbreviations: FE: Frequent exacerbators; IE: Infrequent exacerbators; OCS: Oral corticosteroids use; RNA-seq: RNA sequencing.
DEFINITIONS.
Candidate-gene association study: Statistical approach that interrogates the association of genetic variation with a trait of interest, analysing genomic regions selected based on a biological hypothesis.
Genome-wide association study (GWAS): Agnostic scan of genetic variation across the genome for association with a trait of interest.
Genome-wide interaction study (GWIS): Agnostic scan of the interaction between genetic variation across the genome and a factor of interest (e.g., environmental or genetic factors) on a phenotype under study.
Admixture mapping: gene mapping approach that investigates whether chromosomal ancestry (local ancestry) is associated with a trait of interest, allowing the detection of genomic regions harbouring genetic variants that exhibit ancestry differences.
Next-generation sequencing (NGS): high-throughput technology that allow determining the DNA sequence of single DNA molecules in parallel. These methods involve DNA fragmentation, DNA sequencing, and mapping to an organism’s reference genome to detect the genetic variation of a given sample.
Whole exome sequencing (WES): next-generation approach that allows detecting genetic variation in the genomic protein-coding regions (exons).
Whole genome sequencing (WGS): next-generation approach that determine the DNA sequence in the entire genome.
Epigenome-wide association studies (EWAS): agnostic scan of epigenetic markers, usually DNA methylation, across the genome for association with a trait of interest.
Transcriptome-wide association studies (TWAS): agnostic gene-based scan of gene expression across the whole genome for association with a trait of interest.
Proteome-wide association studies (PWAS): agnostic scan of protein expression for association with a trait of interest.
Metabolome-wide association studies (MWAS): agnostic scan of metabolite levels for association with a trait of interest.
Quantitative trait locus (QTL): position of the DNA sequence where genetic variation is associated with a quantitative trait, such as DNA methylation (methylation quantitative trait locus, meQTL), gene expression levels (expression quantitative trait locus, eQTL), splicing ratios of transcripts (splicing quantitative trait locus, sQTL), protein levels (protein quantitative trait locus, pQTL) or metabolite levels (metabolic quantitative trait locus, mQTL).
Expression quantitative methylation (eQTM): position of the DNA sequence where methylation levels are associated with gene expression levels.
FUNDING
This work was funded by the Spanish Ministry of Science and Innovation MCIN/AEI/10.13039/501100011033 (PID2020-116274RB-I00). MP-Y was funded by the Ramón y Cajal Program (RYC-2015-17205) by MCIN/AEI/10.13039/501100011033 and by the European Social Fund “ESF Investing in your future”. EH-L was supported by a fellowship awarded by MCIN/AEI/10.13039/501100011033 and by “ESF Investing in your future” (PRE2018-083837).
ABBREVIATIONS
- AEs
Area under the curve
- AUC
Asthma exacerbations
- ChIP-Seq
Chromatin immunoprecipitation sequencing
- COPD
Chronic obstructive pulmonary disease
- CpG
Cytosine-phosphate-guanine
- DMRs
Differentially methylated regions
- DNAm
DNA methylation
- eQTM
Expression quantitative trait methylation
- eQTL
Expression quantitative trait loci
- EWAS
Epigenome-wide association study
- FeNO
Fractional exhaled nitric oxide
- FEV1
Forced expiratory volume in one second
- GxE
Gene-environment
- GWAS
Genome-wide association study
- GWIS
Genome-wide interaction study
- ICS
Inhaled corticosteroids
- LABA
Long-acting beta2-agonists
- lncRNA
Long non-coding RNA
- miRNA
Micro-RNAs
- mRNA
Messenger RNA
- OCS
Oral corticosteroids
- PGWAS
Pharmacogenomics GWAS
- PGWIS
Pharmacogenomics GWIS
- PRS
Polygenic risk score
- TNF
Tumor necrosis factor
- UK
United Kingdom
- US
United States
- WGCNA
Weighted gene co-expression network analysis
Footnotes
CONFLICT OF INTEREST
The funding agencies had no role in the study design, data collection, and analysis, decision to publish, or preparation of the manuscript. EH-L, and MP-Y report funding from the Spanish Ministry of Science and Innovation (MCIN/AEI/10.13039/501100011033). MP-Y reports grant support CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Spain (CB/06/06/1088) and the European Regional Development Fund “ERDF A way of making Europe as well as from GlaxoSmithKline, Spain paid to Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC) for a project outside the submitted work. EH-L was supported by a fellowship awarded by MCIN/AEI/10.13039/501100011033 and by “ESF Investing in your future” (PRE2018-083837). EF reports funding from the U.S. National Institutes of Health (NIH, grant HL149693), the University of Pittsburgh Precision Medicine Institute, and the UPMC Children’s Hospital of Pittsburgh. JCC reports funding from the NIH (grants HL152475 and 150431), as well as receipt of research materials (inhaled corticosteroids) from Merck in order to provide medications free of cost to participants in an NIH-funded study (unrelated to this manuscript).
REFERENCES
- 1.Herrera-Luis E, Hernandez-Pacheco N, Vijverberg SJ, Flores C, Pino-Yanes M. Role of genomics in asthma exacerbations. Curr Opin Pulm Med. 2019;25(1):101–12. [DOI] [PubMed] [Google Scholar]
- 2.Martin MJ, Beasley R, Harrison TW. Towards a personalised treatment approach for asthma attacks. Thorax. 2020;75(12):1119–29. [DOI] [PubMed] [Google Scholar]
- 3.Asthma and Lung UK. Asthma data visualisations. Available at https://www.asthma.org.uk/support-us/campaigns/data-visualisations/
- 4.Mukherjee M, Stoddart A, Gupta RP, Nwaru BI, Farr A, Heaven M, et al. The epidemiology, healthcare and societal burden and costs of asthma in the UK and its member nations: analyses of standalone and linked national databases. BMC Med. 2016;14(1):113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Nurmagambetov T, Kuwahara R, Garbe P. The Economic Burden of Asthma in the United States, 2008–2013. Ann Am Thorac Soc. 2018;15(3):348–56. [DOI] [PubMed] [Google Scholar]
- 6.Luskin AT, Chipps BE, Rasouliyan L, Miller DP, Haselkorn T, Dorenbaum A. Impact of asthma exacerbations and asthma triggers on asthma-related quality of life in patients with severe or difficult-to-treat asthma. J allergy Clin Immunol Pract. 2(5):544–52.e1–2. [DOI] [PubMed] [Google Scholar]
- 7.Chipps BE, Haselkorn T, Rosén K, Mink DR, Trzaskoma BL, Luskin AT. Asthma Exacerbations and Triggers in Children in TENOR: Impact on Quality of Life. J allergy Clin Immunol Pract. 6(1):169–176.e2. [DOI] [PubMed] [Google Scholar]
- 8.Rastogi D, Madhok N, Kipperman S. Caregiver Asthma Knowledge, Aptitude, and Practice in High Healthcare Utilizing Children: Effect of an Educational Intervention. Pediatr Allergy Immunol Pulmonol. 2013;26(3):128–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Major S, Vézina K, Tse SM. Lung Function of Children Following an Intensive Care Unit Admission for Asthma. Pediatr Allergy Immunol Pulmonol. 2021;34(1):1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Briggs A, Nasser S, Hammerby E, Buchs S, Virchow JC. The impact of moderate and severe asthma exacerbations on quality of life: a post hoc analysis of randomised controlled trial data. J patient-reported outcomes. 2021;5(1):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.O’Brian AL, Lemanske RF, Evans MD, Gangnon RE, Gern JE, Jackson DJ. Recurrent severe exacerbations in early life and reduced lung function at school age. J Allergy Clin Immunol. 2012;129(4):1162–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.O’Byrne PM, Pedersen S, Lamm CJ, Tan WC, Busse WW, START Investigators Group. Severe exacerbations and decline in lung function in asthma. Am J Respir Crit Care Med. 2009;179(1):19–24. [DOI] [PubMed] [Google Scholar]
- 13.Bai TR, Vonk JM, Postma DS, Boezen HM. Severe exacerbations predict excess lung function decline in asthma. Eur Respir J. 2007;30(3):452–6. [DOI] [PubMed] [Google Scholar]
- 14.Gold DR, Sordillo JE, Coull BA. Lung Function Tracking throughout Childhood: Growth Trajectories May Not Be Set in Stone. J allergy Clin Immunol Pract. 2020;8(4):1272–4. [DOI] [PubMed] [Google Scholar]
- 15.Hallas HW, Chawes BL, Arianto L, Rasmussen MA, Kunøe A, Stokholm J, et al. Children with Asthma Have Fixed Airway Obstruction through Childhood Unaffected by Exacerbations. J allergy Clin Immunol Pract. 2020;8(4):1263–1271.e3. [DOI] [PubMed] [Google Scholar]
- 16.Winkler T, Frey U. Airway remodeling: Shifting the trigger point for exacerbations in asthma. J Allergy Clin Immunol. 2021;148(3):710–2. [DOI] [PubMed] [Google Scholar]
- 17.Krings JG, Goss CW, Lew D, Samant M, McGregor MC, Boomer J, et al. Quantitative CT metrics are associated with longitudinal lung function decline and future asthma exacerbations: Results from SARP-3. J Allergy Clin Immunol. 2021;148(3):752–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Navanandan N, Hatoun J, Celedón JC, Liu AH. Predicting Severe Asthma Exacerbations in Children: Blueprint for Today and Tomorrow. J allergy Clin Immunol Pract. 2021;9(7):2619–26. [DOI] [PubMed] [Google Scholar]
- 19.Bloom CI, Palmer T, Feary J, Quint JK, Cullinan P. Exacerbation Patterns in Adults with Asthma in England. A Population-based Study. Am J Respir Crit Care Med. 2019;199(4):446–53. [DOI] [PubMed] [Google Scholar]
- 20.Chipps BE, Zeiger RS, Borish L, Wenzel SE, Yegin A, Hayden M Lou, et al. Key findings and clinical implications from The Epidemiology and Natural History of Asthma: Outcomes and Treatment Regimens (TENOR) study. J Allergy Clin Immunol. 2012;130(2):332–42.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ramsahai JM, Hansbro PM, Wark PAB. Mechanisms and Management of Asthma Exacerbations. Am J Respir Crit Care Med. 2019;199(4):423–32. [DOI] [PubMed] [Google Scholar]
- 22.Ioachimescu OC, Desai NS. Nonallergic Triggers and Comorbidities in Asthma Exacerbations and Disease Severity. Clin Chest Med. 2019;40(1):71–85. [DOI] [PubMed] [Google Scholar]
- 23.di Palmo E, Cantarelli E, Catelli A, Ricci G, Gallucci M, Miniaci A, et al. The Predictive Role of Biomarkers and Genetics in Childhood Asthma Exacerbations. Int J Mol Sci. 2021;22(9):4651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Lee DS, Gross E, Hotz A, Rastogi D. Comparison of severity of asthma hospitalization between African American and Hispanic children in the Bronx. J Asthma. 2020;57(7):736–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Rosser FJ, Forno E, Cooper PJ, Celedón JC. Asthma in Hispanics. An 8-year update. Am J Respir Crit Care Med. 2014;189(11):1316–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Akinbami LJ, Moorman JE, Simon AE, Schoendorf KC. Trends in racial disparities for asthma outcomes among children 0 to 17 years, 2001–2010. J Allergy Clin Immunol. 2014;134(3):547–553.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Akinbami LJ, Moorman JE, Bailey C, Zahran HS, King M, Johnson CA, et al. Trends in asthma prevalence, health care use, and mortality in the United States, 2001–2010. NCHS Data Brief. 2012;(94):1–8. [PubMed] [Google Scholar]
- 28.Oraka E, Iqbal S, Flanders WD, Brinker K, Garbe P. Racial and ethnic disparities in current asthma and emergency department visits: findings from the National Health Interview Survey, 2001–2010. J Asthma. 2013;50(5):488–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Engelkes M, Baan EJ, de Ridder MAJ, Svensson E, Prieto-Alhambra D, Lapi F, et al. Incidence, risk factors and re-exacerbation rate of severe asthma exacerbations in a multinational, multidatabase pediatric cohort study. Pediatr Allergy Immunol. 2020;31(5):496–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Silber JH, Rosenbaum PR, Calhoun SR, Reiter JG, Hill AS, Guevara JP, et al. Racial Disparities in Medicaid Asthma Hospitalizations. Pediatrics. 2017;139(1):e20161221. [DOI] [PubMed] [Google Scholar]
- 31.Rumpel JA, Ahmedani BK, Peterson EL, Wells KE, Yang M, Levin AM, et al. Genetic ancestry and its association with asthma exacerbations among African American subjects with asthma. J Allergy Clin Immunol. 2012;130(6):1302–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Grossman NL, Ortega VE, King TS, Bleecker ER, Ampleford EA, Bacharier LB, et al. Exacerbation-prone asthma in the context of race and ancestry in Asthma Clinical Research Network trials. J Allergy Clin Immunol. 2019;144(6):1524–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Flores C, Ma S-FF, Pino-Yanes M, Wade MS, Pérez-Méndez L, Kittles RA, et al. African ancestry is associated with asthma risk in African Americans. PLoS One. 2012;7(1):e26807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Brehm JM, Acosta-Pérez E, Klei L, Roeder K, Barmada MM, Boutaoui N, et al. African ancestry and lung function in Puerto Rican children. J Allergy Clin Immunol. 2012;129(6):1484–1490.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Pino-Yanes M, Thakur N, Gignoux CR, Galanter JM, Roth LA, Eng C, et al. Genetic ancestry influences asthma susceptibility and lung function among Latinos. J Allergy Clin Immunol. 2015;135(1):228–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mersha TB, Qin K, Beck AF, Ding L, Huang B, Kahn RS. Genetic ancestry differences in pediatric asthma readmission are mediated by socioenvironmental factors. J Allergy Clin Immunol. 2021;148(5):1210–1218.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gautam Y, Johansson E, Mersha TB. Multi-Omics Profiling Approach to Asthma: An Evolving Paradigm. J Pers Med. 2022;12(1):66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.NCBI Resource Coordinators. Database resources of the National Center for Biotechnology Information. Nucleic Acids Res. 2016;44(D1):D7–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Duncan LE, Ostacher M, Ballon J. How genome-wide association studies (GWAS) made traditional candidate gene studies obsolete. Neuropsychopharmacology. 2019;44(9):1518–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.El-Husseini ZW, Gosens R, Dekker F, Koppelman GH. The genetics of asthma and the promise of genomics-guided drug target discovery. Lancet Respir Med. 2020;8(10):1045–56. [DOI] [PubMed] [Google Scholar]
- 41.Hernandez-Pacheco N, Flores C, Oh SS, Burchard EG, Pino-Yanes M. What Ancestry Can Tell Us About the Genetic Origins of Inter-Ethnic Differences in Asthma Expression. Curr Allergy Asthma Rep. 2016;16(8):53. [DOI] [PubMed] [Google Scholar]
- 42.Hernandez-Pacheco N, Pino-Yanes M, Flores C. Genomic Predictors of Asthma Phenotypes and Treatment Response. Front Pediatr. 2019;7:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Schoettler N, Rodríguez E, Weidinger S, Ober C. Advances in asthma and allergic disease genetics: Is bigger always better? J Allergy Clin Immunol. 2019;144(6):1495–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Shriner D Overview of Admixture Mapping. Curr Protoc Hum Genet. 2017;94:1.23.1–1.23.8. [DOI] [PubMed] [Google Scholar]
- 45.Gallagher MD, Chen-Plotkin AS. The Post-GWAS Era: From Association to Function. Am J Hum Genet. 2018;102(5):717–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ghoussaini M, Mountjoy E, Carmona M, Peat G, Schmidt EM, Hercules A, et al. Open Targets Genetics: systematic identification of trait-associated genes using large-scale genetics and functional genomics. Nucleic Acids Res. 2021;49(D1):D1311–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kamat MA, Blackshaw JA, Young R, Surendran P, Burgess S, Danesh J, et al. PhenoScanner V2: an expanded tool for searching human genotype-phenotype associations. Bioinformatics. 2019;35(22):4851–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Consortium GTEx. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science. 2020;369(6509):1318–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Min JL, Hemani G, Hannon E, Dekkers KF, Castillo-Fernandez J, Luijk R, et al. Genomic and phenotypic insights from an atlas of genetic effects on DNA methylation. Nat Genet. 2021;53(9):1311–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Tse SM, Krajinovic M, Chauhan BF, Zemek R, Gravel J, Chalut D, et al. Genetic determinants of acute asthma therapy response in children with moderate-to-severe asthma exacerbations. Pediatr Pulmonol. 2019;54(4):378–85. [DOI] [PubMed] [Google Scholar]
- 51.Uh S-T, Park J-S, Koo S-M, Kim Y-K, Kim KU, Kim M-A, et al. Association of Genetic Variants of NLRP4 with Exacerbation of Asthma: The Effect of Smoking. DNA Cell Biol. 2019;38(1):76–84. [DOI] [PubMed] [Google Scholar]
- 52.Cho SH, Jo A, Casale T, Jeong SJ, Hong S-J, Cho JK, et al. Soy isoflavones reduce asthma exacerbation in asthmatic patients with high PAI-1-producing genotypes. J Allergy Clin Immunol. 2019;144(1):109–117.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Tsai C-H, Wu AC, Chiang B-L, Yang Y-H, Hung S-P, Su M-W, et al. CEACAM3 decreases asthma exacerbations and modulates respiratory syncytial virus latent infection in children. Thorax. 2020;75(9):725–34. [DOI] [PubMed] [Google Scholar]
- 54.Leiter K, Franks K, Borland ML, Coleman L, Harris L, Le Souëf PN, et al. Vitamin D receptor polymorphisms are associated with severity of wheezing illnesses and asthma exacerbations in children. J Steroid Biochem Mol Biol. 2020;201:105692. [DOI] [PubMed] [Google Scholar]
- 55.Karimi L, Vijverberg SJ, Engelkes M, Hernandez-Pacheco N, Farzan N, Soares P, et al. ADRB2 haplotypes and asthma exacerbations in children and young adults: An individual participant data meta-analysis. Clin Exp Allergy. 2021;51(9):1157–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Herrera-Luis E, Lorenzo-Diaz F, Samedy-Bates LA, Eng C, Villar J, Rodriguez-Santana JR, et al. A deoxyribonuclease 1–like 3 genetic variant associates with asthma exacerbations. J Allergy Clin Immunol. 2021;147(3):1095–1097.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hernandez-Pacheco N, Gorenjak M, Jurgec S, Corrales A, Jorgensen A, Karimi L, et al. Combined analysis of transcriptomic and genetic data for the identification of loci involved in glucocorticosteroid response in asthma. Allergy. 2021;76(4):1238–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Maroteau C, Espuela-Ortiz A, Herrera-Luis E, Srinivasan S, Carr F, Tavendale R, et al. LTA4H rs2660845 association with montelukast response in early and late-onset asthma. PLoS One. 2021;16(9):e0257396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Li X, Christenson SA, Modena B, Li H, Busse WW, Castro M, et al. Genetic analyses identify GSDMB associated with asthma severity, exacerbations, and antiviral pathways. J Allergy Clin Immunol. 2021;147(3):894–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Dijk FN, Vijverberg SJ, Hernandez-Pacheco N, Repnik K, Karimi L, Mitratza M, et al. IL1RL1 gene variations are associated with asthma exacerbations in children and adolescents using inhaled corticosteroids. Allergy. 2020;75(4):984–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Edris A, de Roos EW, McGeachie MJ, Verhamme KMC, Brusselle GG, Tantisira KG, et al. Pharmacogenetics of inhaled corticosteroids and exacerbation risk in adults with asthma. Clin Exp Allergy. 2022;52(1):33–45. [DOI] [PubMed] [Google Scholar]
- 62.Kim M-H, Chang HS, Lee J-U, Shim J-S, Park J-S, Cho Y-J, et al. Association of genetic variants of oxidative stress responsive kinase 1 (OXSR1) with asthma exacerbations in non-smoking asthmatics. BMC Pulm Med. 2022;22(1):3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kan M, Diwadkar AR, Shuai H, Joo J, Wang AL, Ong M-S, et al. Multiomics analysis identifies BIRC3 as a novel glucocorticoid response-associated gene. J Allergy Clin Immunol. 2022;149(6):1981–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, et al. A large-scale, consortium-based genomewide association study of asthma. N Engl J Med. 2010;363(13):1211–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Bouzigon E, Corda E, Aschard H, Dizier M-H, Boland A, Bousquet J, et al. Effect of 17q21 variants and smoking exposure in early-onset asthma. N Engl J Med. 2008;359(19):1985–94. [DOI] [PubMed] [Google Scholar]
- 66.Flory JH, Sleiman PM, Christie JD, Annaiah K, Bradfield J, Kim CE, et al. 17q12–21 variants interact with smoke exposure as a risk factor for pediatric asthma but are equally associated with early-onset versus late-onset asthma in North Americans of European ancestry. J Allergy Clin Immunol. 2009;124(3):605–7. [DOI] [PubMed] [Google Scholar]
- 67.Hernandez-Pacheco N, Kere M, Melén E. Gene-environment interactions in childhood asthma revisited; expanding the interaction concept. Pediatr Allergy Immunol. 2022;33(5):e13780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Sunde RB, Thorsen J, Pedersen C-ET, Stokholm J, Bønnelykke K, Chawes B, et al. Prenatal tobacco exposure and risk of asthma and allergy outcomes in childhood. Eur Respir J. 2022;59(2):2100453. [DOI] [PubMed] [Google Scholar]
- 69.Poli G, Fabi C, Bellet MM, Costantini C, Nunziangeli L, Romani L, et al. Epigenetic Mechanisms of Inflammasome Regulation. Int J Mol Sci. 2020;21(16). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hernandez-Pacheco N, Gorenjak M, Jurgec S, Corrales A, Jorgensen A, Karimi L, et al. Combined analysis of transcriptomic and genetic data for the identification of loci involved in glucocorticosteroid response in asthma. Allergy. 2021;76(4):1238–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yan Q, Forno E, Herrera-Luis E, Pino-Yanes M, Yang G, Oh S, et al. A genome-wide association study of asthma hospitalizations in adults. J Allergy Clin Immunol. 2021;147(3):933–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Yan Q, Forno E, Herrera-Luis E, Pino-Yanes M, Qi C, Rios R, et al. A genome-wide association study of severe asthma exacerbations in Latino children and adolescents. Eur Respir J. 2021;57(4):2002693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Herrera-Luis E, Espuela-Ortiz A, Lorenzo-Diaz F, Keys KL, Mak ACY, Eng C, et al. Genome-wide association study reveals a novel locus for asthma with severe exacerbations in diverse populations. Pediatr Allergy Immunol. 2021;32(1):106–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ahluwalia TS, Eliasen AU, Sevelsted A, Pedersen C-ET, Stokholm J, Chawes B, et al. FUT2-ABO epistasis increases the risk of early childhood asthma and Streptococcus pneumoniae respiratory illnesses. Nat Commun. 2020;11(1):6398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Son J-H, Park J-S, Lee J-U, Kim MK, Min S-A, Park C-S, et al. A genome-wide association study on frequent exacerbation of asthma depending on smoking status. Respir Med. 2022;199:106877. [DOI] [PubMed] [Google Scholar]
- 76.Herrera-Luis E, Ortega VE, Ampleford EJ, Sio YY, Granell R, de Roos E, et al. Multi-ancestry genome-wide association study of asthma exacerbations. Pediatr Allergy Immunol. 2022;33(6):e13802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Hernandez-Pacheco N, Farzan N, Francis B, Karimi L, Repnik K, Vijverberg SJ, et al. Genome-wide association study of inhaled corticosteroid response in admixed children with asthma. Clin Exp Allergy. 2019;49(6):789–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Levin AM, Gui H, Hernandez-Pacheco N, Yang M, Xiao S, Yang JJ, et al. Integrative approach identifies corticosteroid response variant in diverse populations with asthma. J Allergy Clin Immunol. 2019;143(5):1791–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Slob EMA, Richards LB, Vijverberg SJH, Longo C, Koppelman GH, Pijnenburg MWH, et al. Genome-wide association studies of exacerbations in children using long-acting beta2-agonists. Pediatr Allergy Immunol. 2021;32(6):1197–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hernandez-Pacheco N, Vijverberg SJ, Herrera-Luis E, Li J, Sio YY, Granell R, et al. Genome-wide association study of asthma exacerbations despite inhaled corticosteroid use. Eur Respir J. 2021;57(5):2003388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hernandez-Pacheco N, Gorenjak M, Li J, Repnik K, Vijverberg SJ, Berce V, et al. Identification of ROBO2 as a Potential Locus Associated with Inhaled Corticosteroid Response in Childhood Asthma. J Pers Med. 2021;11(8):733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wang AL, Lahousse L, Dahlin A, Edris A, McGeachie M, Lutz SM, et al. Novel genetic variants associated with inhaled corticosteroid treatment response in older adults with asthma. Thorax. 2022; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Dahlin A, Sordillo JE, McGeachie M, Kelly RS, Tantisira KG, Lutz SM, et al. Genome-wide interaction study reveals age-dependent determinants of responsiveness to inhaled corticosteroids in individuals with asthma. PLoS One. 2020;15(3):e0229241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Forno E, Zhang R, Jiang Y, Kim S, Yan Q, Ren Z, et al. Transcriptome-wide and differential expression network analyses of childhood asthma in nasal epithelium. J Allergy Clin Immunol. 2020;146(3):671–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Repapi E, Sayers I, Wain LV., Burton PR, Johnson T, Obeidat M, et al. Genome-wide association study identifies five loci associated with lung function. Nat Genet. 2010;42(1):36–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Burk RD, Chen Z, Saller C, Tarvin K, Carvalho AL, Scapulatempo-Neto C, et al. Integrated genomic and molecular characterization of cervical cancer. Nature. 2017;543(7645):378–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Lutz SM, Cho MH, Young K, Hersh CP, Castaldi PJ, McDonald M-L, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Soler Artigas M, Wain LV, Miller S, Kheirallah AK, Huffman JE, Ntalla I, et al. Sixteen new lung function signals identified through 1000 Genomes Project reference panel imputation. Nat Commun. 2015;6:8658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Hardin M, Cho MH, McDonald M-LL, Wan E, Lomas DA, Coxson HO, et al. A genome-wide analysis of the response to inhaled β2-agonists in chronic obstructive pulmonary disease. Pharmacogenomics J. 2016;16(4):326–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Adcock IM, Tsaprouni L, Bhavsar P, Ito K. Epigenetic regulation of airway inflammation. Curr Opin Immunol. 2007;19(6):694–700. [DOI] [PubMed] [Google Scholar]
- 91.Adcock IM, Ito K, Barnes PJ. Histone deacetylation: an important mechanism in inflammatory lung diseases. COPD. 2005;2(4):445–55. [DOI] [PubMed] [Google Scholar]
- 92.Herrera-Luis E, Mak ACY, Perez-Garcia J, Martin-Gonzalez E, Eng C, Beckman KB, et al. Admixture mapping of severe asthma exacerbations in Hispanic/Latino children and youth. Thorax. 2022; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Ortega VE, Daya M, Szefler SJ, Bleecker ER, Chinchilli VM, Phipatanakul W, et al. Pharmacogenetic studies of long-acting beta agonist and inhaled corticosteroid responsiveness in randomised controlled trials of individuals of African descent with asthma. Lancet Child Adolesc Heal. 2021;5(12):862–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lee EY, Mak ACY, Hu D, Sajuthi S, White MJ, Keys KL, et al. Whole-Genome Sequencing Identifies Novel Functional Loci Associated with Lung Function in Puerto Rican Youth. Am J Respir Crit Care Med. 2020;202(7):962–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Sheikhpour M, Maleki M, Ebrahimi Vargoorani M, Amiri V. A review of epigenetic changes in asthma: methylation and acetylation. Clin Epigenetics. 2021;13(1):65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Alashkar Alhamwe B, Alhamdan F, Ruhl A, Potaczek DP, Renz H. The role of epigenetics in allergy and asthma development. Curr Opin Allergy Clin Immunol. 2020;20(1):48–55. [DOI] [PubMed] [Google Scholar]
- 97.Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13(7):484–92. [DOI] [PubMed] [Google Scholar]
- 98.Curtin JA, Simpson A, Belgrave D, Semic-Jusufagic A, Custovic A, Martinez FD. Methylation of IL-2 promoter at birth alters the risk of asthma exacerbations during childhood. Clin Exp Allergy. 2013;43(3):304–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wang AL, Gruzieva O, Qiu W, Kebede Merid S, Celedón JC, Raby BA, et al. DNA methylation is associated with inhaled corticosteroid response in persistent childhood asthmatics. Clin Exp Allergy. 2019;49(9):1225–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Bustamante J, Zhang S-Y, Boisson B, Ciancanelli M, Jouanguy E, Dupuis-Boisson S, et al. Immunodeficiencies at the Interface of Innate and Adaptive Immunity. In: Clinical Immunology. Elsevier; 2019. p. 509–522.e1. [Google Scholar]
- 101.Weidner J, Bartel S, Kılıç A, Zissler UM, Renz H, Schwarze J, et al. Spotlight on microRNAs in allergy and asthma. Allergy. 2021;76(6):1661–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Tubita V, Callejas-Díaz B, Roca-Ferrer J, Marin C, Liu Z, Wang DY, et al. Role of microRNAs in inflammatory upper airway diseases. Allergy. 2021;76(7):1967–80. [DOI] [PubMed] [Google Scholar]
- 103.Cañas JA, Rodrigo-Muñoz JM, Sastre B, Gil-Martinez M, Redondo N, Del Pozo V. MicroRNAs as Potential Regulators of Immune Response Networks in Asthma and Chronic Obstructive Pulmonary Disease. Front Immunol. 2020;11:608666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Midyat L, Gulen F, Karaca E, Ozkinay F, Tanac R, Demir E, et al. MicroRNA expression profiling in children with different asthma phenotypes. Pediatr Pulmonol. 2016;51(6):582–7. [DOI] [PubMed] [Google Scholar]
- 105.Tian M, Zhou Y, Jia H, Zhu X, Cui Y. The Clinical Significance of Changes in the Expression Levels of MicroRNA-1 and Inflammatory Factors in the Peripheral Blood of Children with Acute-Stage Asthma. Biomed Res Int. 2018;2018:7632487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Korde A, Ahangari F, Haslip M, Zhang X, Liu Q, Cohn L, et al. An endothelial microRNA-1-regulated network controls eosinophil trafficking in asthma and chronic rhinosinusitis. J Allergy Clin Immunol. 2020;145(2):550–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kho AT, McGeachie MJ, Moore KG, Sylvia JM, Weiss ST, Tantisira KG. Circulating microRNAs and prediction of asthma exacerbation in childhood asthma. Respir Res. 2018;19(1):128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Wardzyńska A, Pawełczyk M, Rywaniak J, Kurowski M, Makowska JS, Kowalski ML. Circulating MicroRNAs and T-Cell Cytokine Expression Are Associated With the Characteristics of Asthma Exacerbation. Allergy Asthma Immunol Res. 2020;12(1):125–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Gomez JL, Chen A, Diaz MP, Zirn N, Gupta A, Britto C, et al. A Network of Sputum MicroRNAs Is Associated with Neutrophilic Airway Inflammation in Asthma. Am J Respir Crit Care Med. 2020;202(1):51–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Tiwari A, Hobbs BD, Li J, Kho AT, Amr S, Celedón JC, et al. Blood miRNAs Are Linked to Frequent Asthma Exacerbations in Childhood Asthma and Adult COPD. Non-coding RNA. 2022;8(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Mattes J, Collison A, Plank M, Phipps S, Foster PS. Antagonism of microRNA-126 suppresses the effector function of TH2 cells and the development of allergic airways disease. Proc Natl Acad Sci U S A. 2009;106(44):18704–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kho AT, Sharma S, Davis JS, Spina J, Howard D, McEnroy K, et al. Circulating MicroRNAs: Association with Lung Function in Asthma. PLoS One. 2016;11(6):e0157998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Li H, Wang H, Sokulsky L, Liu S, Yang R, Liu X, et al. Single-cell transcriptomic analysis reveals key immune cell phenotypes in the lungs of patients with asthma exacerbation. J Allergy Clin Immunol. 2021;147(3):941–54. [DOI] [PubMed] [Google Scholar]
- 114.McCauley KE, Flynn K, Calatroni A, DiMassa V, LaMere B, Fadrosh DW, et al. Seasonal airway microbiome and transcriptome interactions promote childhood asthma exacerbations. J Allergy Clin Immunol. 2022;150(1):204–13. [DOI] [PubMed] [Google Scholar]
- 115.Klaile E, Klassert TE, Scheffrahn I, Müller MM, Heinrich A, Heyl KA, et al. Carcinoembryonic antigen (CEA)-related cell adhesion molecules are co-expressed in the human lung and their expression can be modulated in bronchial epithelial cells by non-typable Haemophilus influenzae, Moraxella catarrhalis, TLR3, and type I and II int. Respir Res. 2013;14:85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Hoda U, Pavlidis S, Bansal AT, Takahashi K, Hu S, Ng Kee Kwong F, et al. Clinical and transcriptomic features of persistent exacerbation-prone severe asthma in U-BIOPRED cohort. Clin Transl Med. 2022;12(4):e816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Cho MH, Hobbs BD, Silverman EK. Genetics of chronic obstructive pulmonary disease: understanding the pathobiology and heterogeneity of a complex disorder. Lancet Respir Med. 2022;10(5):485–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.DeMeo DL. Sex and Gender Omic Biomarkers in Men and Women With COPD: Considerations for Precision Medicine. Chest. 2021;160(1):104–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Regan EA, Hersh CP, Castaldi PJ, DeMeo DL, Silverman EK, Crapo JD, et al. Omics and the Search for Blood Biomarkers in Chronic Obstructive Pulmonary Disease. Insights from COPDGene. Am J Respir Cell Mol Biol. 2019;61(2):143–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Bønnelykke K, Sleiman P, Nielsen K, Kreiner-Møller E, Mercader JM, Belgrave D, et al. A genome-wide association study identifies CDHR3 as a susceptibility locus for early childhood asthma with severe exacerbations. Nat Genet. 2014;46(1):51–5. [DOI] [PubMed] [Google Scholar]
- 121.Atkinson EG, Maihofer AX, Kanai M, Martin AR, Karczewski KJ, Santoro ML, et al. Tractor uses local ancestry to enable the inclusion of admixed individuals in GWAS and to boost power. Nat Genet. 2021;53(2):195–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Dahlin A, Denny J, Roden DM, Brilliant MH, Ingram C, Kitchner TE, et al. CMTR1 is associated with increased asthma exacerbations in patients taking inhaled corticosteroids. Immunity, Inflamm Dis. 2015;3(4):350–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Sordillo JE, Lutz SM, Jorgenson E, Iribarren C, McGeachie M, Dahlin A, et al. A polygenic risk score for asthma in a large racially diverse population. Clin Exp Allergy. 2021;51(11):1410–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Namjou B, Lape M, Malolepsza E, DeVore SB, Weirauch MT, Dikilitas O, et al. Multiancestral polygenic risk score for pediatric asthma. J Allergy Clin Immunol. 2022;S0091–6749(22)00660–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Kothalawala DM, Kadalayil L, Curtin JA, Murray CS, Simpson A, Custovic A, et al. Integration of Genomic Risk Scores to Improve the Prediction of Childhood Asthma Diagnosis. J Pers Med. 2022;12(1):75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Dijk FN, Folkersma C, Gruzieva O, Kumar A, Wijga AH, Gehring U, et al. Genetic risk scores do not improve asthma prediction in childhood. J Allergy Clin Immunol. 2019;144(3):857–860.e7. [DOI] [PubMed] [Google Scholar]
- 127.Wu C, Zhu J, King A, Tong X, Lu Q, Park JY, et al. Novel strategy for disease risk prediction incorporating predicted gene expression and DNA methylation data: a multi-phased study of prostate cancer. Cancer Commun (London, England). 2021;41(12):1387–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Hu X, Qiao D, Kim W, Moll M, Balte PP, Lange LA, et al. Polygenic transcriptome risk scores for COPD and lung function improve cross-ethnic portability of prediction in the NHLBI TOPMed program. Am J Hum Genet. 2022;109(5):857–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Kilanowski A, Chen J, Everson T, Thiering E, Wilson R, Gladish N, et al. Methylation risk scores for childhood aeroallergen sensitization: Results from the LISA birth cohort. Allergy. 2022;77(9):2803–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Montesinos López OA, Montesinos López A, Crossa J. Overfitting, Model Tuning, and Evaluation of Prediction Performance. In: Montesinos López OA, Montesinos López A, Crossa J, editors. Multivariate Statistical Machine Learning Methods for Genomic Prediction. 1st ed. Springer; Cham; 2022. p. 109–139. [PubMed] [Google Scholar]
- 131.Lee TY, Petkau J, Sadatsafavi M. Long-Term Natural History of Severe Asthma Exacerbations and Their Impact on the Disease Course . Ann Am Thorac Soc. 2022;19(6):907–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Bélanger É, Laprise C. Could the Epigenetics of Eosinophils in Asthma and Allergy Solve Parts of the Puzzle? Int J Mol Sci. 2021;22(16). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Hudon Thibeault A-A, Laprise C. Cell-Specific DNA Methylation Signatures in Asthma. Genes (Basel). 2019;10(11):932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Rahmani E, Schweiger R, Rhead B, Criswell LA, Barcellos LF, Eskin E, et al. Cell-type-specific resolution epigenetics without the need for cell sorting or single-cell biology. Nat Commun. 2019;10(1):3417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Helling BA, Sobreira DR, Hansen GT, Sakabe NJ, Luo K, Billstrand C, et al. Altered transcriptional and chromatin responses to rhinovirus in bronchial epithelial cells from adults with asthma. Commun Biol. 2020;3(1):678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Kim SH, MacIntyre DA, Sykes L, Arianoglou M, Bennett PR, Terzidou V. Whole Blood Holding Time Prior to Plasma Processing Alters microRNA Expression Profile. Front Genet. 2021;12:818334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Chorley BN, Atabakhsh E, Doran G, Gautier J-C, Ellinger-Ziegelbauer H, Jackson D, et al. Methodological considerations for measuring biofluid-based microRNA biomarkers. Crit Rev Toxicol. 2021;51(3):264–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Taka S, Tzani-Tzanopoulou P, Wanstall H, Papadopoulos NG. MicroRNAs in Asthma and Respiratory Infections: Identifying Common Pathways. Allergy Asthma Immunol Res. 2020;12(1):4–23. [DOI] [PMC free article] [PubMed] [Google Scholar]