5,660
Views
30
CrossRef citations to date
0
Altmetric
Review

The role of epigenetics in the development of childhood asthma

, &
Pages 1287-1302 | Received 17 Jul 2019, Accepted 28 Oct 2019, Published online: 10 Nov 2019

References

  • Lambrecht BN, Hammad H. The airway epithelium in asthma. Nat Med. 2012;18:684–692.
  • Lambrecht BN, Hammad H. The immunology of asthma. Nat Immunol. 2015;16:45–56.
  • Eder W, Ege MJ, von Mutius E. The asthma epidemic. N Engl J Med. 2006;355:2226–2235.
  • de Nijs SB, Venekamp LN, Bel EH. Adult-onset asthma: is it really different? Eur Respir Rev. 2013;22:44–52.
  • Ferreira MAR, Mathur R, Vonk JM, et al. Genetic architectures of childhood- and adult-onset asthma are partly distinct. Am J Hum Genet. 2019;104:665–684.
  • Meyers DA, Bleecker ER, Holloway JW, et al. Asthma genetics and personalised medicine. Lancet Respir Med. 2014;2:405–415.
  • Thomsen SF, Van Der Sluis S, Kyvik KO, et al. Estimates of asthma heritability in a large twin sample: estimates of asthma heritability. Clin Exp Allergy. 2010;40:1054–1061.
  • Moffatt MF, Kabesch M, Liang L, et al. Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature. 2007;448:470–473.
  • Demenais F, Margaritte-Jeannin P, Australian Asthma Genetics Consortium (AAGC) collaborators, et al. Multiancestry association study identifies new asthma risk loci that colocalize with immune-cell enhancer marks. Nat Genet. 2018;50:42–53.
  • Stein MM, Hrusch CL, Gozdz J, et al. Innate immunity and asthma risk in amish and hutterite farm children. N Engl J Med. 2016;375:411–421.
  • Thacher JD, Gruzieva O, Pershagen G, et al. Mold and dampness exposure and allergic outcomes from birth to adolescence: data from the BAMSE cohort. Allergy. 2017;72:967–974.
  • O’Connor GT, Lynch SV, Bloomberg GR, et al. Early-life home environment and risk of asthma among inner-city children. J Allergy Clin Immunol. 2018;141:1468–1475.
  • Kelsey G, Stegle O, Reik W. Single-cell epigenomics: recording the past and predicting the future. Science. 2017;358:69–75.
  • Forno E, Celedón JC. Epigenomics and transcriptomics in the prediction and diagnosis of childhood asthma: are we there yet? Front Pediatr. 2019;7:115.
  • Gibney ER, Nolan CM. Epigenetics and gene expression. Heredity (Edinb). 2010;105:4–13.
  • Gardiner-Garden M, Frommer M. CpG Islands in vertebrate genomes. J Mol Biol. 1987;196:261–282.
  • Schübeler D. Function and information content of DNA methylation. Nature. 2015;517:321–326.
  • Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33:245–254.
  • Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13:484–492.
  • Cedar H, Bergman Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet. 2009;10:295–304.
  • Portela A, Esteller M. Epigenetic modifications and human disease. Nat Biotechnol. 2010;28:1057–1068.
  • Potaczek DP, Harb H, Michel S, et al. Epigenetics and allergy: from basic mechanisms to clinical applications. Epigenomics. 2017;9:539–571.
  • Holoch D, Moazed D. RNA-mediated epigenetic regulation of gene expression. Nat Rev Genet. 2015;16:71–84.
  • Lin S, Gregory RI. MicroRNA biogenesis pathways in cancer. Nat Rev Cancer. 2015;15:321–333.
  • Wei J-W, Huang K, Yang C, et al. Non-coding RNAs as regulators in epigenetics. Oncol Rep. 2017;37:3–9.
  • Gruzieva O, Merid SK, Melén E. An update on epigenetics and childhood respiratory diseases. Paediatr Respir Rev. 2014;15:348–354.
  • Teschendorff AE, Relton CL. Statistical and integrative system-level analysis of DNA methylation data. Nat Rev Genet. 2018;19:129–147.
  • Houseman E, Accomando WP, Koestler DC, et al. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics. 2012;13:86.
  • Xu C-J, Söderhäll C, Bustamante M, et al. DNA methylation in childhood asthma: an epigenome-wide meta-analysis. Lancet Respir Med. 2018;6:379–388.
  • Reese SE, Xu C-J, den Dekker HT, et al. Epigenome-wide meta-analysis of DNA methylation and childhood asthma. J Allergy Clin Immunol [Internet]. 2018 [cited 2019 Jan 22]. Available from: https://linkinghub.elsevier.com/retrieve/pii/S009167491832788X
  • Felix JF, Joubert BR, Baccarelli AA, et al. Cohort profile: Pregnancy And Childhood Epigenetics (PACE) consortium. Int J Epidemiol. 2018;47:22–23u.
  • Arathimos R, Suderman M, Sharp GC, et al. Epigenome-wide association study of asthma and wheeze in childhood and adolescence. Clin Epigenet. 2017;9:112.
  • Roth HM, Wadsworth SJ, Kahn M, et al. The airway epithelium in asthma: developmental issues that scar the airways for life? Pulm Pharmacol Ther. 2012;25:420–426.
  • Murphy TM, Wong CCY, Arseneault L, et al. Methylomic markers of persistent childhood asthma: a longitudinal study of asthma-discordant monozygotic twins. Clin Epigenet. 2015;7:130.
  • Brugha R, Lowe R, Henderson AJ, et al. DNA methylation profiles between airway epithelium and proxy tissues in children. Acta Paediatrica. 2017;106:2011–2016.
  • Yang IV, Pedersen BS, Liu AH, et al. The nasal methylome and childhood atopic asthma. J Allergy Clin Immunol. 2017;139:1478–1488.
  • Forno E, Wang T, Qi C, et al. DNA methylation in nasal epithelium, atopy, and atopic asthma in children: a genome-wide study. Lancet Respir Med. 2019;7:336–346.
  • Cardenas A, Sordillo JE, Rifas-Shiman SL, et al. The nasal methylome as a biomarker of asthma and airway inflammation in children. Nat Commun. 2019;10:3095.
  • Kidd CDA, Thompson PJ, Barrett L, et al. Histone modifications and asthma. the interface of the epigenetic and genetic landscapes. Am J Respir Cell Mol Biol. 2016;54:3–12.
  • Seumois G, Chavez L, Gerasimova A, et al. Epigenomic analysis of primary human T cells reveals enhancers associated with TH2 memory cell differentiation and asthma susceptibility. Nat Immunol. 2014;15:777–788.
  • Harb H, Raedler D, Ballenberger N, et al. Childhood allergic asthma is associated with increased IL-13 and FOXP3 histone acetylation. J Allergy Clin Immunol. 2015;136:200–202.
  • Wawrzyniak P, Wawrzyniak M, Wanke K, et al. Regulation of bronchial epithelial barrier integrity by type 2 cytokines and histone deacetylases in asthmatic patients. J Allergy Clin Immunol. 2017;139:93–103.
  • Stefanowicz D, Lee JY, Lee K, et al. Elevated H3K18 acetylation in airway epithelial cells of asthmatic subjects. Respir Res. 2015;16:95.
  • Specjalski K, Jassem E. MicroRNAs: potential biomarkers and targets of therapy in allergic diseases? Arch Immunol Ther Exp (Warsz.). 2019;67:213–223.
  • Narożna B, Langwiński W, Szczepankiewicz A. Non-coding RNAs in pediatric airway diseases. Genes (Basel). 2017;8:348.
  • Kärner J, Wawrzyniak M, Tankov S, et al. Increased microRNA-323-3p in IL-22/IL-17-producing T cells and asthma: a role in the regulation of the TGF-β pathway and IL-22 production. Allergy. 2017;72:55–65.
  • Sawant DV, Yao W, Wright Z, et al. Serum MicroRNA-21 as a biomarker for allergic inflammatory disease in children. Microrna. 2015;4:36–40.
  • Elbehidy RM, Youssef DM, El-Shal AS, et al. MicroRNA-21 as a novel biomarker in diagnosis and response to therapy in asthmatic children. Mol Immunol. 2016;71:107–114.
  • Hammad Mahmoud Hammad R, Hamed DHED, Eldosoky MAER, et al. Plasma microRNA-21, microRNA-146a and IL-13 expression in asthmatic children. Innate Immun. 2018;24:171–179.
  • Karam RA, Abd Elrahman DM. Differential expression of miR-155 and Let-7a in the plasma of childhood asthma: potential biomarkers for diagnosis and severity. Clin Biochem. 2019;68:30–36.
  • Johansson K, Malmhäll C, Ramos-Ramírez P, et al. MicroRNA-155 is a critical regulator of type 2 innate lymphoid cells and IL-33 signaling in experimental models of allergic airway inflammation. J Allergy Clin Immunol. 2017;139:1007–1016.e9.
  • Kumar M, Ahmad T, Sharma A, et al. Let-7 microRNA-mediated regulation of IL-13 and allergic airway inflammation. J Allergy Clin Immunol. 2011;128:1077–1085.e1–10.
  • Saulyte J, Regueira C, Montes-Martínez A, et al. Active or passive exposure to tobacco smoking and allergic rhinitis, allergic dermatitis, and food allergy in adults and children: a systematic review and meta-analysis. Novotny TE, editor. PLoS Med. 2014;11:e1001611.
  • Murrison LB, Brandt EB, Myers JB, et al. Environmental exposures and mechanisms in allergy and asthma development. J Clin Investig. 2019;129:1504–1515.
  • Burbank AJ, Sood AK, Kesic MJ, et al. Environmental determinants of allergy and asthma in early life. J Allergy Clin Immunol. 2017;140:1–12.
  • von Mutius E. The microbial environment and its influence on asthma prevention in early life. J Allergy Clin Immunol. 2016;137:680–689.
  • Miliku K, Azad M. Breastfeeding and the developmental origins of asthma: current evidence, possible mechanisms, and future research priorities. Nutrients. 2018;10:995.
  • Beckhaus AA, Garcia-Marcos L, Forno E, et al. Maternal nutrition during pregnancy and risk of asthma, wheeze, and atopic diseases during childhood: a systematic review and meta-analysis. Allergy. 2015;70:1588–1604.
  • Rusconi F, Popovic M. Maternal obesity and childhood wheezing and asthma. Paediatr Respir Rev. 2017;22:66–71.
  • DeVries A, Wlasiuk G, Miller SJ, et al. Epigenome-wide analysis links SMAD3 methylation at birth to asthma in children of asthmatic mothers. J Allergy Clin Immunol. 2017;140:534–542.
  • Joubert BR, Felix JF, Yousefi P, et al. DNA methylation in newborns and maternal smoking in pregnancy: genome-wide consortium meta-analysis. Am J Hum Genet. 2016;98:680–696.
  • Dijkstra A, Howard TD, Vonk JM, et al. Estrogen receptor 1 polymorphisms are associated with airway hyperresponsiveness and lung function decline, particularly in female subjects with asthma. J Allergy Clin Immunol. 2006;117:604–611.
  • Meyer N, Christoph J, Makrinioti H, et al. Inhibition of angiogenesis by IL-32: possible role in asthma. J Allergy Clin Immunol. 2012;129:964–973.e7.
  • Bauer T, Trump S, Ishaque N, et al. Environment-induced epigenetic reprogramming in genomic regulatory elements in smoking mothers and their children. Mol Syst Biol. 2016;12:861.
  • Joetham A, Schedel M, Takeda K, et al. JNK2 regulates the functional plasticity of naturally occurring T regulatory cells and the enhancement of lung allergic responses. J.I. 2014;193:2238–2247.
  • Buro-Auriemma LJ, Salit J, Hackett NR, et al. Cigarette smoking induces small airway epithelial epigenetic changes with corresponding modulation of gene expression. Hum Mol Genet. 2013;22:4726–4738.
  • Alipoor SD, Adcock IM, Garssen J, et al. The roles of miRNAs as potential biomarkers in lung diseases. Eur J Pharmacol. 2016;791:395–404.
  • Andersson B-Å, Sayardoust S, Löfgren S, et al. Cigarette smoking affects microRNAs and inflammatory biomarkers in healthy individuals and an association to single nucleotide polymorphisms is indicated. Biomarkers. 2019;24:180–185.
  • Osei ET, Florez-Sampedro L, Timens W, et al. Unravelling the complexity of COPD by microRNAs: it’s a small world after all. Eur Respir J. 2015;46:807–818.
  • Guarnieri M, Balmes JR. Outdoor air pollution and asthma. Lancet. 2014;383:1581–1592.
  • Sofer T, Baccarelli A, Cantone L, et al. Exposure to airborne particulate matter is associated with methylation pattern in the asthma pathway. Epigenomics. 2013;5:147–154.
  • Prunicki M, Stell L, Dinakarpandian D, et al. Exposure to NO2, CO, and PM2.5 is linked to regional DNA methylation differences in asthma. Clin Epigenet. 2018;10:2.
  • Lichtenfels DFCAJ, van der Plaat DA, de Jong K, et al. Long-term air pollution exposure, genome-wide DNA methylation and lung function in the lifelines cohort study. Environ Health Perspect. 2018;126:027004.
  • Gruzieva O, Xu C-J, Yousefi P, et al. Prenatal particulate air pollution and DNA methylation in newborns: an epigenome-wide meta-analysis. Environ Health Perspect. 2019;127:057012.
  • Heßelbach K, Kim G-J, Flemming S, et al. Disease relevant modifications of the methylome and transcriptome by particulate matter (PM 2.5) from biomass combustion. Epigenetics. 2017;12:779–792.
  • Somineni HK, Zhang X, Biagini Myers JM, et al. Ten-eleven translocation 1 (TET1) methylation is associated with childhood asthma and traffic-related air pollution. J Allergy Clin Immunol. 2016;137:797–805.e5.
  • Liu AH. Revisiting the hygiene hypothesis for allergy and asthma. J Allergy Clin Immunol. 2015;136:860–865.
  • Braun-Fahrländer C, Riedler J, Herz U, et al. Environmental exposure to endotoxin and its relation to asthma in school-age children. N Engl J Med. 2002;347:869–877.
  • Woo V, Alenghat T. Host–microbiota interactions: epigenomic regulation. Curr Opin Immunol. 2017;44:52–60.
  • Obata Y, Furusawa Y, Endo TA, et al. The epigenetic regulator Uhrf1 facilitates the proliferation and maturation of colonic regulatory T cells. Nat Immunol. 2014;15:571–579.
  • Lu Y, Guo Y, Xu L, et al. Foxp3 regulates ratio of Treg and NKT cells in a mouse model of asthma. Mol Cell Biochem. 2015;403:25–31.
  • Schaub B, Liu J, Höppler S, et al. Maternal farm exposure modulates neonatal immune mechanisms through regulatory T cells. J Allergy Clin Immunol. 2009;123:774–782.e5.
  • Blanken MO, Rovers MM, Molenaar JM, et al. Respiratory syncytial virus and recurrent wheeze in healthy preterm infants. N Engl J Med. 2016;374:2406.
  • Scheltema NM, Nibbelke EE, Pouw J, et al. Respiratory syncytial virus prevention and asthma in healthy preterm infants: a randomised controlled trial. Lancet Respir Med. 2018;6:257–264.
  • Lund RJ, Osmala M, Malonzo M, et al. Atopic asthma after rhinovirus-induced wheezing is associated with DNA methylation change in the SMAD3 gene promoter. Allergy. 2018;73:1735–1740.
  • Ptaschinski C, Mukherjee S, Moore ML, et al. RSV-Induced H3K4 demethylase KDM5B leads to regulation of dendritic cell-derived innate cytokines and exacerbates pathogenesis in vivo. Heise MT, editor. PLoS Pathog. 2015;11:e1004978.
  • Kroetz DN, Allen RM, Schaller MA, et al. Type I interferon induced epigenetic regulation of macrophages suppresses innate and adaptive immunity in acute respiratory viral infection. Thomas PG, editor. PLoS Pathog. 2015;11:e1005338.
  • Inchley CS, Sonerud T, Fjærli HO, et al. Nasal mucosal microRNA expression in children with respiratory syncytial virus infection. BMC Infect Dis. 2015;15:150.
  • Duijts L. Fetal and infant origins of asthma. Eur J Epidemiol. 2012;27:5–14.
  • Popovic M, Fiano V, Fasanelli F, et al. Differentially methylated DNA regions in early childhood wheezing: an epigenome‐wide study using saliva. Genuneit J, editor. Pediatr Allergy Immunol. 2019;30:305–314.
  • Gunawardhana LP, Baines KJ, Mattes J, et al. Differential DNA methylation profiles of infants exposed to maternal asthma during pregnancy: maternal Asthma and Infant DNA methylation. Pediatr Pulmonol. 2014;49:852–862.
  • Chen C-M, Tischer C, Schnappinger M, et al. The role of cats and dogs in asthma and allergy–a systematic review. Int J Hyg Environ Health. 2010;213:1–31.
  • Takkouche B, González-Barcala F-J, Etminan M, et al. Exposure to furry pets and the risk of asthma and allergic rhinitis: a meta-analysis: risk of asthma and allergic rhinitis. Allergy. 2008;63:857–864.
  • Zhang X, Chen X, Weirauch MT, et al. Diesel exhaust and house dust mite allergen lead to common changes in the airway methylome and hydroxymethylome. Baccarelli A, editor. Environ Epigenet [Internet]. 2018 [cited 2019 Sep 19]; 4. Available from: https://academic.oup.com/eep/article/doi/10.1093/eep/dvy020/5060796
  • Miller RL, Zhang H, Jezioro J, et al. Reduced mouse allergen is associated with epigenetic changes in regulatory genes, but not mouse sensitization, in asthmatic children. Environ Res. 2017;156:619–624.
  • Qiu L, Zhang Y, Do DC, et al. miR-155 modulates cockroach allergen- and oxidative stress-induced cyclooxygenase-2 in asthma. J Immunol. 2018;201:916–929.
  • Bonder MJ, Luijk R, The BIOS Consortium, et al. Disease variants alter transcription factor levels and methylation of their binding sites. Nat Genet. 2017;49:131–138.
  • Dijk FN, Xu C, Melén E, et al. Genetic regulation of IL1RL1 methylation and IL1RL1-a protein levels in asthma. Eur Respir J. 2018;51:1701377.
  • Soliai MM, Kato A, Stanhope CT, et al. Multi-omics co-localization with genome-wide association studies reveals context-specific mechanisms of asthma risk variants [Internet]. Genetics. 2019 [cited 2019 Jun 26]. Available from: http://biorxiv.org/lookup/doi/10.1101/593558
  • Guerra S, Melén E, Sunyer J, et al. Genetic and epigenetic regulation of YKL-40 in childhood. J Allergy Clin Immunol. 2018;141:1105–1114.
  • Lee CG, Hartl D, Lee GR, et al. Role of breast regression protein 39 (BRP-39)/chitinase 3-like-1 in Th2 and IL-13-induced tissue responses and apoptosis. J Exp Med. 2009;206:1149–1166.
  • Kothari PH, Qiu W, Croteau-Chonka DC, et al. Role of local CpG DNA methylation in mediating the 17q21 asthma susceptibility gasdermin B (GSDMB)/ORMDL sphingolipid biosynthesis regulator 3 (ORMDL3) expression quantitative trait locus. J Allergy Clin Immunol. 2018;141:2282–2286.e6.
  • McVicker G, van de Geijn B, Degner JF, et al. Identification of genetic variants that affect histone modifications in human cells. Science. 2013;342:747–749.
  • Huan T, Rong J, Liu C, et al. Genome-wide identification of microRNA expression quantitative trait loci. Nat Commun. 2015;6:6601.
  • Rutledge H, Baran-Gale J, de Villena FP-M, et al. Identification of microRNAs associated with allergic airway disease using a genetically diverse mouse population. BMC Genomics. 2015;16:633.
  • Weidner J, Malmhäll C, Rådinger M microRNAs in asthma pathogenesis - from mouse to man. JTGG [Internet]. 2019 [cited 2019 Jul 9]; Available from: https://jtggjournal.com/article/view/2951.
  • Pacis A, Mailhot-Léonard F, Tailleux L, et al. Gene activation precedes DNA demethylation in response to infection in human dendritic cells. Proc Natl Acad Sci USA. 2019;116:6938–6943.
  • Nicodemus-Johnson J, Naughton KA, Sudi J, et al. Genome-wide methylation study identifies an IL-13-induced epigenetic signature in asthmatic airways. Am J Respir Crit Care Med. 2016;193:376–385.
  • Nicodemus-Johnson J, Myers RA, Sakabe NJ, et al. DNA methylation in lung cells is associated with asthma endotypes and genetic risk. JCI Insight. 2016;1:e90151.
  • Pal S, Tyler JK. Epigenetics and aging. Sci Adv. 2016;2:e1600584.
  • Xu C-J, Bonder MJ, Söderhäll C, et al. The emerging landscape of dynamic DNA methylation in early childhood. BMC Genomics. 2017;18:25.
  • Bauer M. Cell-type-specific disturbance of DNA methylation pattern: a chance to get more benefit from and to minimize cohorts for epigenome-wide association studies. Int J Epidemiol. 2018;47(3):917–927.
  • Tumes DJ, Papadopoulos M, Endo Y, et al. Epigenetic regulation of T-helper cell differentiation, memory, and plasticity in allergic asthma. Immunol Rev. 2017;278:8–19.
  • Vieira Braga FA, Kar G, Berg M, et al. A cellular census of human lungs identifies novel cell states in health and in asthma. Nat Med [Internet]. 2019 [cited 2019 Jun 24]. Available from: http://www.nature.com/articles/s41591-019-0468-5
  • Liang L, Willis-Owen SAG, Laprise C, et al. An epigenome-wide association study of total serum immunoglobulin E concentration. Nature. 2015;520:670–674.
  • Breeze CE, Paul DS, van Dongen J, et al. eFORGE: A tool for identifying cell type-specific signal in epigenomic data. Cell Rep. 2016;17:2137–2150.
  • Zheng SC, Breeze CE, Beck S, et al. Identification of differentially methylated cell types in epigenome-wide association studies. Nat Methods. 2018;15:1059–1066.
  • Chen X, Miragaia RJ, Natarajan KN, et al. A rapid and robust method for single cell chromatin accessibility profiling. Nat Commun. 2018;9:5345.
  • Smallwood SA, Lee HJ, Angermueller C, et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat Methods. 2014;11:817–820.
  • Guo H, Zhu P, Guo F, et al. Profiling DNA methylome landscapes of mammalian cells with single-cell reduced-representation bisulfite sequencing. Nat Protoc. 2015;10:645–659.