Abstract
Asthma is a common complex respiratory disease characterized by chronic airway inflammation and partially reversible airflow obstruction resulting from genetic and environmental determinants. Since epigenetic marks influence gene expression and can be modified by both environmental exposures and genetic variation they are increasingly recognized as relevant to the pathogenesis of asthma and may be a key link between environmental exposures and asthma susceptibility. Unlike changes to DNA sequence, epigenetic signatures are dynamic and reversible, creating an opportunity for not only therapeutic targets but may serve as biomarkers to follow disease course and identify molecular subtypes in heterogeneous diseases such as asthma. In this review, we will examine the relationship between asthma and three key epigenetic processes that modify gene expression: DNA methylation, modification of histone tails, and non-coding RNAs. In addition to presenting a comprehensive assessment of the existing epigenetic studies focusing on immune regulation in asthma, we will also discuss future directions for epigenetic investigation in allergic airway disease.
Keywords: Asthma, Epigenetics, DNA methylation, histone modification, non-coding RNA, microRNA
Introduction
Asthma is a heterogeneous disease characterized by airway inflammation and partially reversible airflow obstruction1. It is a common chronic respiratory disease defined by a history of shortness of breath, wheeze, chest tightness and cough that vary over time and intensity1. Asthma is a complex disease resulting from genetic, epigenetic, and environmental determinants. Genetic studies have identified numerous genetic polymorphisms associated with asthma susceptibility, but these genetic variants alone are unable to explain a large proportion of the variability in asthma risk2–4. Environmental exposures are thought to contribute to the increased prevalence of asthma, however, environmental exposures further compound the complexity of this disease by causing airway inflammation and obstruction through a variety of mechanisms. Notably, epigenetic modifications are increasingly recognized as critical in the pathogenesis of asthma and may be a key link between environmental exposures and asthma endotypes5, 6. In this review, we will present a comprehensive assessment of the existing epigenetic studies focusing on immune regulation in asthma and discuss future directions for epigenetic investigation in allergic airway disease.
Epigenetics
Epigenetics refers to potentially heritable changes in gene expression that do not involve specific changes in nucleotide sequence. The epigenome, the collection of epigenetic modifications across an individual’s genome, is influenced primarily by environmental exposures and genetic variation. Environmental exposures of high relevance to asthma, including specific allergens7, cigarette smoke8, and air pollution9, have all been associated with changes in epigenetic modifications. This is also true of dietary factors that have been associated with asthma, such as folate and vitamin D10. Environmental influences on the epigenome have been documented both prenatally and postnatally11, 12. An individual’s genetic background can impact epigenetic modifications by genetic variants that are directly associated with changes in epigenetic marks11, 13–16. Importantly, epigenetic modifications can also be inherited, classically involving imprinted loci17. While there is a global loss of the majority of known epigenetic modifications on parental chromosomes during primordial germ cell development and after fertilization, potential mechanisms of transgenerational epigenetic inheritance are being unraveled18. Recently transcription factors (TFs) were identified to be carriers of epigenetic information during germ cell and pre-implantation development19. Through this, epigenetic modifications are likely to be a key mechanism by which traits are passed from parent or grandparent to offspring, such as the influence of maternal and grandmaternal cigarette smoking on risk of asthma in the offspring20. Unlike changes to DNA sequence, epigenetic marks are dynamic and can be reversible, creating an opportunity for not only therapeutic targets but also biomarkers to follow disease course and identify molecular subtypes in heterogeneous diseases. We will examine the relationship between asthma and three key epigenetic processes that modify gene expression: DNA methylation, modification of histone tails, and non-coding RNAs (Figure 1).
Figure 1.
demonstrates the relationship between asthma and three key epigenetic processes that modify gene expression: DNA methylation, modification of histone tails, and non-coding RNAs.
DNA Methylation
Addition of a methyl group to cytosine bases in DNA, most often in the context of cytosine followed by guanine (CpG), alters DNA accessibility by transcriptional machinery and results in recruitment of downstream proteins for chromatin remodeling21. In the human genome, gene bodies, repeat sequences, and intergenic regions are generally methylated while promoters of housekeeping genes are not methylated to allow for gene transcription to occur22. The extent of DNA methylation at gene promoters and enhancers is associated with the level of gene expression, with increased DNA methylation often associated with decreased accessibility and repressed transcription. However, recent studies have identified TFs that bind methylated DNA23 and suggest that this canonical relationship of DNA methylation and gene expression is not always followed. While DNA methyl transferase (DNMT) enzymes catalyze addition of methyl group to CpGs, demethylation of CpGs can occur either actively through successive oxidation reactions catalyzed by the ten-eleven translocase (TET) family of enzymes and 5-hydroxymethylcytosine intermediate, resulting in eventual replacement with cytosine through base excision repair24, or passively, in which newly synthesized DNA is not methylated by DNMTs. Sodium bisulfite conversion is the key technique used to assess the level of CpG methylation. DNA treatment with bisulfite results in deamination of cytosine to uracil (which becomes thymine during DNA amplification by PCR) but a methyl group protects this from occurring so that the ratio of C to T at any CpG reflects the extent of DNA methylation25. Next-generation sequencing and array-based genotyping can then be performed.
Investigation of DNA methylation changes are the most common epigenetic studies in asthma. Identification of DNA methylation profiles using both candidate gene and epigenome-wide association studies (EWAS) in asthmatic populations are being used to elucidate biologic mechanisms that influence asthma susceptibility and severity. Importantly, DNA methylation patterns are cell and tissue-specific and reveal how methylation changes are distinct based on the biological site being investigated.
DNA Methylation Studies in the Developmental Origin of Asthma
Epigenetic modifications have been studied in lung development and may provide mechanistic insight into the impact of intrauterine environmental perturbations on subsequent disease risk. The developmental origins of health and disease (DOHaD) hypothesis proposes that environmental exposures during critical periods of development can induce permanent changes in fetal physiology and metabolism that impact disease susceptibility later in life26. Alterations in the epigenome are being increasingly recognized as mechanisms by which intrauterine exposures enhance postnatal disease risk27. Initial studies in the agouti mouse model demonstrated that prenatal nutrition could result in transgenerational changes in DNA methylation altering the phenotype of subsequent generations28. We and others have shown in murine models that maternal diet supplemented with methyl donors and alterations in folate metabolism enhance the severity of allergic airway disease that is inherited in a transgenerational fashion29, 30. These data suggest a causal link between maternal diet and fetal epigenetic changes that subsequently influence postnatal disease.
Other common prenatal exposures including in utero exposure to maternal smoking has been shown to result in differential methylation of genes in children31. We have previously shown that in utero smoke exposure results in differential fetal lung tissue and placental methylation32 and others have demonstrated widespread DNA methylation changes in the blood of children exposed to maternal smoking during pregnancy31, 33. Previous studies have also identified the intrauterine period as a key developmental timeframe during which preventive strategies can be implemented. For example, vitamin C supplementation in pregnant smokers was found to decrease the impact of maternal smoking on offspring pulmonary function and incidence of wheeze at 1 year of age34. Improvements in lung function were mediated by the resolution of DNA methylation changes identified in the placenta of the treated individuals. The reduction in DNA methylation in the placenta occurred at loci in pathways associated with oxidative stress, fetal development, and lung function35. Furthermore, normalization of in utero smoke-induced methylation changes were noted across multiple additional tissues in the offspring suggesting a more systemic impact of vitamin C supplementation on blunting the impact of maternal smoke exposure36. These data demonstrate the benefit of understanding the epigenetic impact of environmental exposures during human lung development to identify novel preventive and therapeutic targets.
Intrauterine exposures have also been shown to impact post-natal susceptibility to allergic disease through regulation of Th1 and Th2 polarization. A study of prenatal exposure to traffic-related airborne polycyclic aromatic hydrocarbons demonstrated an association between methylation of acyl-CoA synthetase long-chain family member 3 (ASCL3) and asthma symptoms before age 537. These changes resulted in increased Th1 polarization. Interestingly, animal models have demonstrated that a combination of other environmental exposures including diesel exhaust particles and allergen exposure differentially modulate Th1 and Th2 gene methylation, gene expression, and subsequent IgE production38. These data suggest that asthma endotypes may be influenced by epigenetic changes that result from environmental exposures during fetal development.
DNA Methylation Studies in Childhood Asthma
Initial studies of DNA methylation in human subjects identified association of DNA methylation with candidate genes in the blood39, buccal mucosa40, and the nasal epithelium41 of asthmatic subjects. For example, increased methylation of the beta-2-adrenergic receptor (ADRB2) in the blood of childhood asthmatics was associated with increased asthma severity,42 while hypomethylation of genes in the lipoxygenase pathway in the blood of childhood asthmatics was associated with increased wheeze39.
EWAS investigations identified DNA methylation signatures associated with markers of atopy including serum Immunoblobulin E (IgE) and peripheral blood eosinophils. A study utilizing data from the Avon Longitudinal Study of Parents and Children (ALSPAC) demonstrated that DNA methylation changes that were associated with asthma were largely driven by higher eosinophil counts in asthma cases43.
The Pregnancy and Childhood Epigenetics (PACE) consortium investigated the association of DNA methylation changes identified at birth with the development of childhood asthma. DNA methylation in newborn blood and measured during childhood identified differentially methylated regions associated with childhood asthma44. The genes identified as dysregulated included genes in the endothelial nitric oxide synthase pathways and biological processes associated with immune function44.
We have previously identified 81 differentially methylated regions in peripheral blood mononuclear cells (PBMCs) associated with allergic asthma in African-American inner-city children45. We found that several immune genes were hypomethylated resulting in overexpression of IL-4 and IL-13 in asthma45. These findings demonstrate the potential importance of DNA methylation on the Th2 immune response in allergic asthma and the impact of epigenetic regulation of T cell differentiation.
Nasal epithelial DNA methylation changes have also been used to identify additional biological pathways that influence asthma susceptibility and severity. Previous investigation of differential DNA methylation in the nasal epithelium between children with and without atopic asthma, identified methylation changes in genes involved in epithelial barrier function and immune regulation. We have previously identified extensive methylation changes in the nasal epithelium of inner-city African American Children with asthma46. Notably, many of the DNA methylation changes that were identified in this cohort influenced the expression of numerous genes46 including many that were also identified in the IL-13 DNA methylome signature of cultured airway epithelial cells of asthmatics suggesting a potential mechanistic link to allergic asthma47. Importantly, replication of nasal epithelial methylation profiles across cohorts demonstrates consistency across top loci and similar effect sizes46, 48.
Notably, DNA methylation studies have also been used to classify subjects based on atopic status demonstrating their potential as predictive biomarkers of asthma. Previous studies have demonstrated that nasal epithelial methylation changes can be used to classify atopic asthmatics from non-asthmatics with a high level of accuracy (area under the curve=0.93)48.
DNA methylation studies in childhood asthma have also established a link between early-life environmental exposures and asthma susceptibility and disease severity. Emerging literature suggests that air pollution exposure modulates DNA methylation during childhood demonstrating the importance of investigation of the impact of environmental exposures throughout the life course. Prunicki and colleagues have demonstrated differential methylation of the Foxp3 gene in asthmatic subjects with exposure to ambient air pollution compared to non-asthmatic control subjects with similar levels of ambient air pollution exposure49. The DNA methylation changes identified in the promoter of the Foxp3 gene were also shown to be associated with impairment in T-regulatory cell (T-reg) function49. Foxp3 has been shown to be involved in immune regulation50 and has been implicated in asthma pathogenesis51. In addition, exposure to traffic-related air pollution including exposure to black carbon has been associated with changes in DNA methylation in the nitric oxide synthase 3 (NOS3) gene in the buccal mucosa and is also associated with increased fraction of expired nitric oxide (FeNO) in asthmatic children52.
DNA Methylation Studies in Adult Asthma
Epigenome-wide studies of DNA methylation support a role for epigenetic mechanisms in childhood asthma, but DNA methylation studies in adults remain limited. Liang and colleagues identified an association of serum IgE and hypomethylation at 36 loci that were identified in three independent adult cohorts53. DNA methylation patterns demonstrate biological differences between remitting and persistent asthma in adults. DNA methylation patterns in bronchial biopsy specimens between subjects with asthma in remission were compared to those with persistent disease and healthy controls. Differentially methylated regions associated with remission include genes involved in the resolution of inflammation54.
Using EWAS from the peripheral blood of adult participants in the Agricultural Lung Health Study (ALHS), 524 differentially methylated sites were identified in adult subjects with non-atopic asthma compared to controls55. These differentially methylated CpG sites were enriched for genes in the asthma and sphingolipid metabolism pathways55. In this cohort, 1086 DNA methylation sites were identified to be differentially methylated in subjects with atopic asthma compared to non-asthmatic controls at a false discovery rate (FDR<0.05) including genes involved in insulin signaling. Notably, more CpG sites were differentially methylated in atopic asthmatics compared to non-atopic asthmatics with approximately 10% overlapping between the two conditions. These results highlight the need to differentiate between atopic and non-atopic asthma. Furthermore, these results highlight the need to investigate epigenetic mechanisms across the life course as asthma risk factors and pathobiological mechanisms may differ during development, in childhood, and into adulthood.
Histone Modifications
Chromatin accessibility is directly regulated by chemical modifications to tails of histone proteins that form octamers around which DNA is wrapped to form nucleosomes56. Histone tails can be modified in many ways, including methylation, acetylation, ubiquitylation, and SUMOylation57. As an example, acetylation of histone tails is often associated with gene activation while deacetylation is associated with gene repression. A common acetylation mark, lysine 27 on the histone H3 (H3K27ac), is a mark of active enhancers and active promoters while trimethylation of that same lysine residue (H3K27me3) is a repressive mark on both promoters and enhancers58. Histone acetyltransferases (HATs) acetylate histone tails, histone deacetylases (HDACs) remove acetyl groups from histone tails, and bromodomain (Brd) proteins are chromatin readers that recognize and bind acetylated histones and play a key role in transmission of epigenetic memory across cell divisions and transcription regulation59. Similarly, histone methyltransferases (HMTs) add the methyl groups to histone tails while histone demethylases (HDMs) remove them. The Encyclopedia of DNA Elements (ENCODE), Roadmap Epigenomics (RE), and the International Human Epigenome Consortium (IHEC) projects have created maps of histone modifications in cell lines and primary cells/tissues, providing important public resources for the research community60–62.
Histone Modifications in Asthma
Although studied less frequently than DNA methylation, histone modifications have been implicated in asthma susceptibility, disease severity, and treatment response. The level of histone acetylation, which is associated with increased gene expression, is dependent on the balance of histone deacetylase (HDAC) and histone acetyltransferase (HAT activity). Evidence demonstrates a role for histone modifications in immune cell differentiation including those involved in Th2 mediated inflammation that are crucial to the development of asthma7, 63. Acetylation of histones has been implicated in the differentiation and function of T-helper cells with increased histone acetylation being identified at Th2 cytokine loci in response to Th2 differentiation resulting in increased Th2 cytokine production64. Previous animal models have also demonstrated the impact of histone acetylation on asthma by demonstrating that a loss of histone acetylation at the interleukin 4 (IL-4)/Il-13 locus greatly reduces Th2 effector functions65. A study characterizing genome-wide histone modifications in T cell subsets from patients with asthma and healthy controls showed asthma-specific differences in enhancers involved in T cell differentiation66.
There is also increasing evidence for the role of HDACs in asthma severity and treatment response. Glucocorticoids remain the most effective anti-inflammatory medications for the treatment of chronic airway inflammation in asthma. Glucocorticoids are known to cause activation of anti-inflammatory gene expression through acetylation of histone H4. Furthermore, activated glucocorticoid receptors reduce histone acetylation through recruitment of HDAC2 resulting in suppression of activated inflammatory genes67. Notably, HDAC2 deficiency results in decreased corticosteroid treatment response in severe asthmatics68, 69. Furthermore, exposure to passive smoke diminished corticosteroid sensitivity of alveolar macrophages obtained from children with severe asthma due to decreased HDAC2 expression70.
HDAC1 is integral to airway epithelial repair and remodeling after injury. Increased HDAC1 has been previously associated with severe asthma and airway remodeling in severe asthmatics compared to those with mild disease71, 72 HDAC1 is associated with T-cell development and previous murine models have demonstrated that HDAC1-deficient T-cells result in increased eosinophil recruitment and enhancing Th2 polarization73. The data linking histone modifications to asthma remain limited. Given the importance of these mechanisms in glucocorticoid handling, additional investigation of the role of histone modifications in asthma susceptibility and treatment response are warranted.
Non-Coding RNAs
Noncoding RNAs (ncRNAs) comprise a much larger portion of the human genome than protein-coding RNAs. They are broadly classified into long non-coding RNAs (lncRNAs) and small RNAs (<200 nucleotides). lncRNAs comprise a large family consisting of long intergenic noncoding RNAs (linkRNAs), enhancer RNAs (eRNAs), and antisense RNAs, to name a few. They perform a variety of functions, including regulation of transcriptional, post-transcriptional control, structural function and genome integrity74. The majority of lncRNAs are polyadenylated and captured with standard mRNA library preparation; however, lncRNAs are less abundant and more cell/tissue specific, therefore requiring deeper sequencing than mRNAs. As an alternative to mRNA sequencing, library preparation protocols that remove ribosomal RNA (rRNA depletion) capture all lncRNAs. The most well-studied class of small RNAs is microRNAs (miRNAs), which are ~22 nucleotide long regulatory RNAs that control gene expression by binding to the 3’ untranslated regions (UTRs) of messenger RNA (mRNA), which leads to either mRNA degradation or inhibition of protein translation75. While immature forms of miRNAs are detectable in standard RNA-seq protocols, the fully mature form can only be detected by doing small RNA-seq library preparation.
Non-Coding RNAs in Asthma
While ncRNAs are known to regulate gene transcription, an understanding of their role in asthma susceptibility remains limited. Murine models have identified the role of several lncRNAs with biological mechanisms tied to asthma pathogenesis. For example, lncRNA PTPRE-AS1 has been shown to modulate M2 macrophage activation and has been associated with allergic inflammation76. Additionally, the expression of PTPRE was found to be significantly lower in PBMCs from patients with allergic asthma suggesting a role in asthma pathogenesis76. LncRNA MALAT1 has also been previously shown to be up regulated in airway smooth muscle cells of asthmatics and has been shown to promote proliferation of airway smooth muscle cells by downregulating miR-216a77. Together these data demonstrate the cell-specific nature of the impact of noncoding RNAs and demonstrates that these long non-coding RNAs impact under lying pathobiology through complex mechanisms that involve microRNAs and other epigenetic and genomic features78. Further research using integrative methods to investigate these epigenetic mechanisms are necessary to elucidate the full impact of non-coding RNAs on asthma pathobiology.
Role of microRNAs in Asthma
The role of miRNAs in asthma pathogenesis has been more widely investigated. miRNAs are integral to normal lung development and murine models have shown that aberrant miRNA expression during lung development has been implicated in subsequent postnatal respiratory disease. We have previously demonstrated the involvement of miRNA-15a with the developmental origin of asthma through its differential expression by intrauterine smoke exposure in our murine model, demonstrating that miR-15a was associated with the intrauterine expression of gasdermin B (GSDMB) a known childhood asthma gene in the human fetal lung, and demonstrating and association of miR-15a expression with asthma exacerbations in children participating in the Childhood Asthma Management Program (CAMP)79. These results suggest that miRNAs impact the expression of known asthma susceptibility loci impacting subsequent disease development.
Previous studies have also demonstrated a link between miRNAs and allergic asthma by altering Th1/Th2 polarization. The let-7 family of miRNAs has been associated with Th2 inflammation80, 81, which is critical to the development of allergic asthma. Two additional miRNAs, mir-155 and miR-221 have been associated with Th2 response and eosinophilia in asthma82. miR-155 targets CTLA-4 expression and enhances the secretion of interleukin-4 (IL-4), IL-5, and IL-13 increasing Th2 mediated inflammation83. miR1 has been shown to have an opposite effect resulting in the inhibition of IL-4 and IL-5 secretion thereby regulating the Th1/Th2 balance in asthma84.
miRNAs are also involved in airway smooth muscle proliferation in asthma. miR-221 has also been shown to play a role in smooth muscle activation in asthma by increasing proliferation of smooth muscle cells in patients with severe disease85. Similarly, miR26 promotes airway smooth muscle hypertrophy though regulation of the expression of GSK-3β 86. These miRNAs are involved in smooth muscle hypertrophy and contraction seen in the airway of asthmatics.
miRNAs have been previously associated with asthma susceptibility and respiratory outcomes in asthmatic subjects. Studies in children with mild-moderate persistent asthma participating in the CAMP study, identified 74 serum microRNAs to be associated with asthma susceptibility and 34 miRNAs to be associated with lung function in asthma87. Eight serum miRNAs were also associated with airway responsiveness in the CAMP cohort88. While these data demonstrate the association of miRNAs with asthma and asthma outcomes, additional functional validation of these loci are necessary to understand the mechanism of action of these miRNAs in asthma pathobiology.
miRNAs have also been implicated in asthma treatment response. Budesonide, an inhaled corticosteroid used in the treatment of asthma, has also been shown to impact the miRNA signature in the bronchial epithelium of asthmatics. Solberg et al showed that inhaled budesonide had modest effect on the miRNA profile in the bronchial epithelium of steroid-naïve patients after initiation of inhaled corticosteroid treatment89. Additionally, several circulating miRNAs have been associated with inhaled budesonide treatment response in CAMP90. Together, these observations demonstrate a role for miRNAs in asthma susceptibility and treatment response by highlighting their impact on the control of inflammatory responses, Th1/Th2 polarization, disease outcomes, and response to therapy.
Future Directions in Epigenetic Investigation
Cross-sectional studies of epigenetic signatures in asthmatic subjects have identified some of the biologic mechanisms underlying asthma susceptibility and disease heterogeneity (Table 1). However, epigenetic studies in extensively phenotyped subjects with asthma followed prospectively remain limited. In addition to better characterization of environmental exposures that impact tissue-specific epigenetic signatures, systems-based approaches that incorporate additional genomic datatypes in asthma cohorts will identify additional biological pathways that impact disease susceptibility and help to identify novel therapeutic targets for this common disease91–93.
Table 1:
Human Epigenetic Studies in Asthma
Epigenetic Mark | Age | Tissue Type | Trait Association | Reference* |
---|---|---|---|---|
DNA Methylation | Fetal | Fetal Lung | Asthma | 32 |
Children | Blood | Asthma | 39,42,43,44 | |
Buccal Mucosa | Asthma | 40 | ||
Nasal Epithelium | Asthma | 41,46,48 | ||
PBMC | Asthma | 45 | ||
Adult | Blood | Asthma | 53,54 | |
Atopy/IgE | 53 | |||
Atopic Asthma | 55 | |||
Histone Modifications | Adult | Blood | Asthma | 68,69 |
MicroRNA | Fetal | Fetal Lung | Asthma | 79 |
Children | Blood | Asthma | 80,81 | |
Asthma Treatment Response | 90 | |||
Asthma/Lung Function | 87 | |||
Adult | Blood | Asthma | 85 | |
Asthma/Treatment Response | 89 |
these studies highlighted in the review.
Furthermore, technological advances including the assessment of chromatin accessibility94 using the Assay for Transposase-Accessible Chromatin using sequencing (ATAC-Seq), Cleavage Under Targets & Tagmentation (CUT&Tag), and single cell sequencing technologies have yet to be extensively applied in asthma. Additionally, recent advances in molecular imaging provide novel non-invasive methods for visualization of epigenetic changes in vivo in a tissue specific manner95. Application of these novel technologies in at-risk and asthmatic cohorts is necessary to elucidate new biological mechanisms that impact asthma susceptibility.
Abbreviations:
- DNMT
DNA methyl transferase
- TFs
transcription factors
- TET
ten-eleven translocase
- EWAS
epigenome-wide association studies
- DOHaD
developmental origins of health and disease
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosures: The authors have nothing to disclose
References:
- 1.Global Strategy for Asthma Management and Prevention (2021 Update). 2021.] Available from https://ginasthma.org/reports/.
- 2.Weiss ST, Silverman EK. Pro: Genome-wide association studies (GWAS) in asthma. Am J Respir Crit Care Med 2011; 184:631–3. [DOI] [PubMed] [Google Scholar]
- 3.Moffatt MF, Kabesch M, Liang L, Dixon AL, Strachan D, Heath S, et al. Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature 2007; 448:470–3. [DOI] [PubMed] [Google Scholar]
- 4.Eichler EE, Flint J, Gibson G, Kong A, Leal SM, Moore JH, et al. Missing heritability and strategies for finding the underlying causes of complex disease. Nat Rev Genet 2010; 11:446–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 2003; 33 Suppl:245–54. [DOI] [PubMed] [Google Scholar]
- 6.Yang IV, Schwartz DA. Epigenetic mechanisms and the development of asthma. J Allergy Clin Immunol 2012; 130:1243–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Potaczek DP, Harb H, Michel S, Alhamwe BA, Renz H, Tost J. Epigenetics and allergy: from basic mechanisms to clinical applications. Epigenomics 2017; 9:539–71. [DOI] [PubMed] [Google Scholar]
- 8.Zong D, Liu X, Li J, Ouyang R, Chen P. The role of cigarette smoke-induced epigenetic alterations in inflammation. Epigenetics Chromatin 2019; 12:65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Rider CF, Carlsten C. Air pollution and DNA methylation: effects of exposure in humans. Clin Epigenetics 2019; 11:131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zhang Y, Kutateladze TG. Diet and the epigenome. Nat Commun 2018; 9:3375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet 2007; 8:253–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Perera BPU, Faulk C, Svoboda LK, Goodrich JM, Dolinoy DC. The role of environmental exposures and the epigenome in health and disease. Environ Mol Mutagen 2020; 61:176–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.McRae AF, Marioni RE, Shah S, Yang J, Powell JE, Harris SE, et al. Identification of 55,000 Replicated DNA Methylation QTL. Sci Rep 2018; 8:17605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hannon E, Gorrie-Stone TJ, Smart MC, Burrage J, Hughes A, Bao Y, et al. Leveraging DNA-Methylation Quantitative-Trait Loci to Characterize the Relationship between Methylomic Variation, Gene Expression, and Complex Traits. Am J Hum Genet 2018; 103:654–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kilpinen H, Waszak SM, Gschwind AR, Raghav SK, Witwicki RM, Orioli A, et al. Coordinated effects of sequence variation on DNA binding, chromatin structure, and transcription. Science 2013; 342:744–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.McVicker G, van de Geijn B, Degner JF, Cain CE, Banovich NE, Raj A, et al. Identification of genetic variants that affect histone modifications in human cells. Science 2013; 342:747–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hanna CW, Kelsey G. Genomic imprinting beyond DNA methylation: a role for maternal histones. Genome Biol 2017; 18:177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Boskovic A, Rando OJ. Transgenerational Epigenetic Inheritance. Annu Rev Genet 2018; 52:21–41. [DOI] [PubMed] [Google Scholar]
- 19.Kremsky I, Corces VG. Protection from DNA re-methylation by transcription factors in primordial germ cells and pre-implantation embryos can explain trans-generational epigenetic inheritance. Genome Biol 2020; 21:118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Mahon GM, Koppelman GH, Vonk JM. Grandmaternal smoking, asthma and lung function in the offspring: the Lifelines cohort study. Thorax 2021; 76:441–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Luo C, Hajkova P, Ecker JR. Dynamic DNA methylation: In the right place at the right time. Science 2018; 361:1336–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chen ZX, Riggs AD. DNA methylation and demethylation in mammals. J Biol Chem 2011; 286:18347–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zhu H, Wang G, Qian J. Transcription factors as readers and effectors of DNA methylation. Nat Rev Genet 2016; 17:551–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Branco MR, Ficz G, Reik W. Uncovering the role of 5-hydroxymethylcytosine in the epigenome. Nat Rev Genet 2011; 13:7–13. [DOI] [PubMed] [Google Scholar]
- 25.Frommer M, McDonald LE, Millar DS, Collis CM, Watt F, Grigg GW, et al. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci U S A 1992; 89:1827–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gillman MW, Barker D, Bier D, Cagampang F, Challis J, Fall C, et al. Meeting report on the 3rd International Congress on Developmental Origins of Health and Disease (DOHaD). Pediatr Res 2007; 61:625–9. [DOI] [PubMed] [Google Scholar]
- 27.Waterland RA, Michels KB. Epigenetic epidemiology of the developmental origins hypothesis. Annu Rev Nutr 2007; 27:363–88. [DOI] [PubMed] [Google Scholar]
- 28.Martin DI, Cropley JE, Suter CM. Environmental influence on epigenetic inheritance at the Avy allele. Nutr Rev 2008; 66 Suppl 1: S12–4. [DOI] [PubMed] [Google Scholar]
- 29.Hollingsworth JW, Maruoka S, Boon K, Garantziotis S, Li Z, Tomfohr J, et al. In utero supplementation with methyl donors enhances allergic airway disease in mice. J Clin Invest 2008; 118:3462–9. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 30.Eyring KR, Pedersen BS, Maclean KN, Stabler SP, Yang IV, Schwartz DA. Methylenetetrahydrofolate reductase contributes to allergic airway disease. PLoS One 2018; 13:e0190916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Breton CV, Byun HM, Wenten M, Pan F, Yang A, Gilliland FD. Prenatal tobacco smoke exposure affects global and gene-specific DNA methylation. Am J Respir Crit Care Med 2009; 180:462–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chhabra D, Sharma S, Kho AT, Gaedigk R, Vyhlidal CA, Leeder JS, et al. Fetal lung and placental methylation is associated with in utero nicotine exposure. Epigenetics 2014; 9:1473–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Joubert BR, Felix JF, Yousefi P, Bakulski KM, Just AC, Breton C, et al. DNA Methylation in Newborns and Maternal Smoking in Pregnancy: Genome-wide Consortium Meta-analysis. Am J Hum Genet 2016; 98:680–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.McEvoy CT, Schilling D, Clay N, Jackson K, Go MD, Spitale P, et al. Vitamin C supplementation for pregnant smoking women and pulmonary function in their newborn infants: a randomized clinical trial. JAMA 2014; 311:2074–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Shorey-Kendrick LE, McEvoy CT, O’Sullivan SM, Milner K, Vuylsteke B, Tepper RS, et al. Impact of vitamin C supplementation on placental DNA methylation changes related to maternal smoking: association with gene expression and respiratory outcomes. Clin Epigenetics 2021; 13:177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Shorey-Kendrick LE, McEvoy CT, Ferguson B, Burchard J, Park BS, Gao L, et al. Vitamin C Prevents Offspring DNA Methylation Changes Associated with Maternal Smoking in Pregnancy. Am J Respir Crit Care Med 2017; 196:745–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Perera F, Tang WY, Herbstman J, Tang D, Levin L, Miller R, et al. Relation of DNA methylation of 5′-CpG island of ACSL3 to transplacental exposure to airborne polycyclic aromatic hydrocarbons and childhood asthma. PLoS One 2009; 4:e4488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Liu J, Ballaney M, Al-alem U, Quan C, Jin X, Perera F, et al. Combined inhaled diesel exhaust particles and allergen exposure alter methylation of T helper genes and IgE production in vivo. Toxicol Sci 2008; 102:76–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Morales E, Bustamante M, Vilahur N, Escaramis G, Montfort M, de Cid R, et al. DNA hypomethylation at ALOX12 is associated with persistent wheezing in childhood. Am J Respir Crit Care Med 2012; 185:937–43. [DOI] [PubMed] [Google Scholar]
- 40.Breton CV, Byun HM, Wang X, Salam MT, Siegmund K, Gilliland FD. DNA methylation in the arginase-nitric oxide synthase pathway is associated with exhaled nitric oxide in children with asthma. Am J Respir Crit Care Med 2011; 184:191–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Baccarelli A, Rusconi F, Bollati V, Catelan D, Accetta G, Hou L, et al. Nasal cell DNA methylation, inflammation, lung function and wheezing in children with asthma. Epigenomics 2012; 4:91–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Fu A, Leaderer BP, Gent JF, Leaderer D, Zhu Y. An environmental epigenetic study of ADRB2 5′-UTR methylation and childhood asthma severity. Clin Exp Allergy 2012; 42:1575–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Arathimos R, Suderman M, Sharp GC, Burrows K, Granell R, Tilling K, et al. Epigenome-wide association study of asthma and wheeze in childhood and adolescence. Clin Epigenetics 2017; 9:112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Reese SE, Xu CJ, den Dekker HT, Lee MK, Sikdar S, Ruiz-Arenas C, et al. Epigenome-wide meta-analysis of DNA methylation and childhood asthma. J Allergy Clin Immunol 2019; 143:2062–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yang IV, Pedersen BS, Liu A, O’Connor GT, Teach SJ, Kattan M, et al. DNA methylation and childhood asthma in the inner city. J Allergy Clin Immunol 2015; 136:69–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yang IV, Pedersen BS, Liu AH, O’Connor GT, Pillai D, Kattan M, et al. The nasal methylome and childhood atopic asthma. J Allergy Clin Immunol 2017; 139:1478–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Nicodemus-Johnson J, Naughton KA, Sudi J, Hogarth K, Naurekas ET, Nicolae DL, et al. Genome-Wide Methylation Study Identifies an IL-13-induced Epigenetic Signature in Asthmatic Airways. Am J Respir Crit Care Med 2016; 193:376–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Forno E, Wang T, Qi C, Yan Q, Xu CJ, Boutaoui N, et al. DNA methylation in nasal epithelium, atopy, and atopic asthma in children: a genome-wide study. Lancet Respir Med 2019; 7:336–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Prunicki M, Stell L, Dinakarpandian D, de Planell-Saguer M, Lucas RW, Hammond SK, et al. Exposure to NO2, CO, and PM2.5 is linked to regional DNA methylation differences in asthma. Clin Epigenetics 2018; 10:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Akdis CA, Akdis M. Mechanisms of immune tolerance to allergens: role of IL-10 and Tregs. J Clin Invest 2014; 124:4678–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zhu X, Chen Q, Liu Z, Luo D, Li L, Zhong Y. Low expression and hypermethylation of FOXP3 in regulatory T cells are associated with asthma in children. Exp Ther Med 2020; 19:2045–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ji N, Fang M, Baptista A, Cepeda C, Greenberg M, Mincey IC, et al. Exposure to traffic-related air pollution and changes in exhaled nitric oxide and DNA methylation in arginase and nitric oxide synthase in children with asthma. Environ Health 2021; 20:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Liang L, Willis-Owen SAG, Laprise C, Wong KCC, Davies GA, Hudson TJ, et al. An epigenome-wide association study of total serum immunoglobulin E concentration. Nature 2015; 520:670–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Vermeulen CJ, Xu CJ, Vonk JM, Ten Hacken NHT, Timens W, Heijink IH, et al. Differential DNA methylation in bronchial biopsies between persistent asthma and asthma in remission. Eur Respir J 2020; 55. [DOI] [PubMed] [Google Scholar]
- 55.Hoang TT, Sikdar S, Xu CJ, Lee MK, Cardwell J, Forno E, et al. Epigenome-wide association study of DNA methylation and adult asthma in the Agricultural Lung Health Study. Eur Respir J 2020; 56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Huang RC, Bonner J. Histone, a suppressor of chromosomal RNA synthesis. Proc Natl Acad Sci U S A 1962; 48:1216–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res 2011; 21:381–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zhou VW, Goren A, Bernstein BE. Charting histone modifications and the functional organization of mammalian genomes. Nat Rev Genet 2011; 12:7–18. [DOI] [PubMed] [Google Scholar]
- 59.Tarakhovsky A. Tools and landscapes of epigenetics. Nat Immunol 2010; 11:565–8. [DOI] [PubMed] [Google Scholar]
- 60.Roadmap Epigenomics C, Kundaje A, Meuleman W, Ernst J, Bilenky M, Yen A, et al. Integrative analysis of 111 reference human epigenomes. Nature 2015; 518:317–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Stunnenberg HG, International Human Epigenome C, Hirst M. The International Human Epigenome Consortium: A Blueprint for Scientific Collaboration and Discovery. Cell 2016; 167:1145–9. [DOI] [PubMed] [Google Scholar]
- 62.Consortium EP. An integrated encyclopedia of DNA elements in the human genome. Nature 2012; 489:57–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Tripathi SK, Lahesmaa R. Transcriptional and epigenetic regulation of T-helper lineage specification. Immunol Rev 2014; 261:62–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Naito T, Taniuchi I. Roles of repressive epigenetic machinery in lineage decision of T cells. Immunology 2013; 139:151–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Koh BH, Hwang SS, Kim JY, Lee W, Kang MJ, Lee CG, et al. Th2 LCR is essential for regulation of Th2 cytokine genes and for pathogenesis of allergic asthma. Proc Natl Acad Sci U S A 2010; 107:10614–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Seumois G, Chavez L, Gerasimova A, Lienhard M, Omran N, Kalinke L, et al. Epigenomic analysis of primary human T cells reveals enhancers associated with TH2 memory cell differentiation and asthma susceptibility. Nat Immunol 2014; 15:777–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ito K, Barnes PJ, Adcock IM. Glucocorticoid receptor recruitment of histone deacetylase 2 inhibits interleukin-1beta-induced histone H4 acetylation on lysines 8 and 12. Mol Cell Biol 2000; 20:6891–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hew M, Bhavsar P, Torrego A, Meah S, Khorasani N, Barnes PJ, et al. Relative corticosteroid insensitivity of peripheral blood mononuclear cells in severe asthma. Am J Respir Crit Care Med 2006; 174:134–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Matthews JG, Ito K, Barnes PJ, Adcock IM. Defective glucocorticoid receptor nuclear translocation and altered histone acetylation patterns in glucocorticoid-resistant patients. J Allergy Clin Immunol 2004; 113:1100–8. [DOI] [PubMed] [Google Scholar]
- 70.Kobayashi Y, Bossley C, Gupta A, Akashi K, Tsartsali L, Mercado N, et al. Passive smoking impairs histone deacetylase-2 in children with severe asthma. Chest 2014; 145:305–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Butler CA, McQuaid S, Taggart CC, Weldon S, Carter R, Skibinski G, et al. Glucocorticoid receptor beta and histone deacetylase 1 and 2 expression in the airways of severe asthma. Thorax 2012; 67:392–8. [DOI] [PubMed] [Google Scholar]
- 72.Kim MH, Kim SH, Kim YK, Hong SJ, Min KU, Cho SH, et al. A polymorphism in the histone deacetylase 1 gene is associated with the response to corticosteroids in asthmatics. Korean J Intern Med 2013; 28:708–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Grausenburger R, Bilic I, Boucheron N, Zupkovitz G, El-Housseiny L, Tschismarov R, et al. Conditional deletion of histone deacetylase 1 in T cells leads to enhanced airway inflammation and increased Th2 cytokine production. J Immunol 2010; 185:3489–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Statello L, Guo CJ, Chen LL, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol 2021; 22:96–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Flynt AS, Lai EC. Biological principles of microRNA-mediated regulation: shared themes amid diversity. Nat Rev Genet 2008; 9:831–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Han X, Huang S, Xue P, Fu J, Liu L, Zhang C, et al. LncRNA PTPRE-AS1 modulates M2 macrophage activation and inflammatory diseases by epigenetic promotion of PTPRE. Sci Adv 2019; 5:eaax9230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Huang J, Wang FH, Wang L, Li Y, Lu J, Chen J. LncRNA MALAT1 promotes proliferation and migration of airway smooth muscle cells in asthma by downregulating microRNA-216a. Saudi J Biol Sci 2021; 28:4124–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wang X, Chen H, Liu J, Gai L, Yan X, Guo Z, et al. Emerging Advances of Non-coding RNAs and Competitive Endogenous RNA Regulatory Networks in Asthma. Bioengineered 2021; 12:7820–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Sharma S, Kho AT, Chhabra D, Haley K, Vyhlidal C, Gaedigk R, et al. Effect of Intrauterine Smoke Exposure on microRNA-15a Expression in Human Lung Development and Subsequent Asthma Risk. Healthcare (Basel) 2020; 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Levanen B, Bhakta NR, Torregrosa Paredes P, Barbeau R, Hiltbrunner S, Pollack JL, et al. Altered microRNA profiles in bronchoalveolar lavage fluid exosomes in asthmatic patients. J Allergy Clin Immunol 2013; 131:894–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kumar M, Ahmad T, Sharma A, Mabalirajan U, Kulshreshtha A, Agrawal A, et al. Let-7 microRNA-mediated regulation of IL-13 and allergic airway inflammation. J Allergy Clin Immunol 2011; 128:1077–85 e1-10. [DOI] [PubMed] [Google Scholar]
- 82.Malmhall C, Alawieh S, Lu Y, Sjostrand M, Bossios A, Eldh M, et al. MicroRNA-155 is essential for T(H)2-mediated allergen-induced eosinophilic inflammation in the lung. J Allergy Clin Immunol 2014; 133:1429–38, 38 e1-7. [DOI] [PubMed] [Google Scholar]
- 83.Zhou H, Li J, Gao P, Wang Q, Zhang J. miR-155: A Novel Target in Allergic Asthma. Int J Mol Sci 2016; 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.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]
- 85.Perry MM, Baker JE, Gibeon DS, Adcock IM, Chung KF. Airway smooth muscle hyperproliferation is regulated by microRNA-221 in severe asthma. Am J Respir Cell Mol Biol 2014; 50:7–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Mohamed JS, Lopez MA, Boriek AM. Mechanical stretch up-regulates microRNA-26a and induces human airway smooth muscle hypertrophy by suppressing glycogen synthase kinase-3beta. J Biol Chem 2010; 285:29336–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.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:e0157998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Davis JS, Sun M, Kho AT, Moore KG, Sylvia JM, Weiss ST, et al. Circulating microRNAs and association with methacholine PC20 in the Childhood Asthma Management Program (CAMP) cohort. PLoS One 2017; 12:e0180329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Solberg OD, Ostrin EJ, Love MI, Peng JC, Bhakta NR, Hou L, et al. Airway epithelial miRNA expression is altered in asthma. Am J Respir Crit Care Med 2012; 186:965–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Li J, Panganiban R, Kho AT, McGeachie MJ, Farnam L, Chase RP, et al. Circulating MicroRNAs and Treatment Response in Childhood Asthma. Am J Respir Crit Care Med 2020; 202:65–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.McGeachie MJ, Dahlin A, Qiu W, Croteau-Chonka DC, Savage J, Wu AC, et al. The metabolomics of asthma control: a promising link between genetics and disease. Immun Inflamm Dis 2015; 3:224–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lefaudeux D, De Meulder B, Loza MJ, Peffer N, Rowe A, Baribaud F, et al. U-BIOPRED clinical adult asthma clusters linked to a subset of sputum omics. J Allergy Clin Immunol 2017; 139:1797–807. [DOI] [PubMed] [Google Scholar]
- 93.Dong Z, Ma Y, Zhou H, Shi L, Ye G, Yang L, et al. Integrated genomics analysis highlights important SNPs and genes implicated in moderate-to-severe asthma based on GWAS and eQTL datasets. BMC Pulm Med 2020; 20:270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Meyer CA, Liu XS. Identifying and mitigating bias in next-generation sequencing methods for chromatin biology. Nat Rev Genet 2014; 15:709–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Wang C, Schroeder FA, Hooker JM. Visualizing epigenetics: current advances and advantages in HDAC PET imaging techniques. Neuroscience 2014; 264:186–97. [DOI] [PMC free article] [PubMed] [Google Scholar]