Search
2025 Volume 18
Article Contents
MINI REVIEW   Open Access    

Epigenetic changes in childhood asthma: a path to precision medicine

More Information
  • The 2023 Global Initiative for Asthma (GINA) report highlights a significant rise in asthma rates particularly in low- and middle-income countries, with urbanization and exposure to environmental pollutants being key contributing factors. Children exposed to environmental influences throughout their critical developmental periods may experience epigenetic changes that could predispose them to asthma in the future. However, there are still issues implementing these findings in clinical settings, necessitating more research and technological advancements. The knowledge of epigenetics in childhood asthma might bridge the gap in knowledge of disease etiology and help the creation of precision medicine approaches. By integrating epigenetic indicators into diagnostic and therapy methods, clinicians can move closer to customized care, benefiting children with asthma. This review emphasizes the significance of epigenetic mechanisms in the development of pediatric asthma and the important associated genes like ORAI1, IL4, ADAM33, GSTM1, FLG, GATA3, STAT6, TNF, CD14, IKZF3, MUC5AC, and ORMDL3. We will also review the role of important epigenetic changes including non-coding RNAs, DNA methylation, and histone modifications in the pathophysiology of asthma. Furthermore, the potential of epigenetics as biomarkers for the diagnosis and prognosis of asthma and the therapeutic uses of epigenetic-based therapies such as DNA methyltransferase inhibitors and histone deacetylase (HDAC) inhibitors have been explored.
  • 加载中
  • [1] Conrad LA, Cabana MD, Rastogi D. 2021. Defining pediatric asthma: phenotypes to endotypes and beyond. Pediatric Research 90(1):45−51 doi: 10.1038/s41390-020-01231-6

    CrossRef   Google Scholar

    [2] Venkatesan P. 2023. GINA report for asthma. The Lancet Respiratory Medicine 11(7):589 doi: 10.1016/S2213-2600(23)00230-8

    CrossRef   Google Scholar

    [3] Zhang T, Huang H, Liang L, Lu H, Liang D. 2024. Long non-coding RNA (LncRNA) non-coding RNA activated by DNA damage (NORAD) knockdown alleviates airway remodeling in asthma via regulating miR-410-3p/RCC2 and inhibiting Wnt/β-catenin pathway. Heliyon 10(1):e23860 doi: 10.1016/j.heliyon.2023.e23860

    CrossRef   Google Scholar

    [4] Chogtu B, Bhattacharjee D, Magazine R. 2016. Epigenetics: the new frontier in the landscape of asthma. Scientifica (Cairo) 2016:4638949 doi: 10.1155/2016/4638949

    CrossRef   Google Scholar

    [5] Cheng Q, Shang Y, Huang W, Zhang Q, Li X, et al. 2019. p300 mediates the histone acetylation of ORMDL3 to affect airway inflammation and remodeling in asthma. International Immunopharmacology 76:105885 doi: 10.1016/j.intimp.2019.105885

    CrossRef   Google Scholar

    [6] Zhang Y. 2023. From gene identifications to therapeutic targets for asthma: focus on great potentials of TSLP, ORMDL3, and GSDMB. Chinese Medical Journal Pulmonary and Critical Care Medicine 1(3):139−47 doi: 10.1016/j.pccm.2023.08.001

    CrossRef   Google Scholar

    [7] Mijač S, Banić I, Genc AM, Lipej M, Turkalj M. 2024. The Effects of Environmental Exposure on Epigenetic Modifications in Allergic Diseases. Medicina (Lithuania) 60(1):110 doi: 10.3390/medicina60010110

    CrossRef   Google Scholar

    [8] Prabhakaran R, Thamarai R, Sivasamy S, Dhandayuthapani S, Batra J, et al. 2024. Epigenetic frontiers: miRNAs, long non-coding RNAs and nanomaterials are pioneering to cancer therapy. Epigenetics and Chromatin 17:31 doi: 10.1186/s13072-024-00554-6

    CrossRef   Google Scholar

    [9] Ranjbar M, Whetstone CE, Omer H, Power L, Cusack RP, et al. 2022. The genetic factors of the airway epithelium associated with the pathology of asthma. Genes 13(10):1870 doi: 10.3390/genes13101870

    CrossRef   Google Scholar

    [10] Hizawa N. 2023. The understanding of asthma pathogenesis in the era of precision medicine. Allergology International 72(1):3−10 doi: 10.1016/j.alit.2022.09.001

    CrossRef   Google Scholar

    [11] Wrennall JA, Ahmad S, Worthington EN, Wu T, Goriounova AS, et al. 2022. A SPLUNC1 peptidomimetic inhibits orai1 and reduces inflammation in a murine allergic asthma model. American Journal of Respiratory Cell and Molecular Biology 66(3):271−82 doi: 10.1165/rcmb.2020-0452OC

    CrossRef   Google Scholar

    [12] Steinke JW, Borish L. 2001. Th2 cytokines and asthma Interleukin-4: its role in the pathogenesis of asthma, and targeting it for asthma treatment with interleukin-4 receptor antagonists. Respiratory Research 2:66−70 doi: 10.1186/rr40

    CrossRef   Google Scholar

    [13] Elsaid A, Shoaib RMS, Badr SS, Wahba Y, Ayyad SEN. 2021. Polymorphisms of interleukin 4 and interleukin 4 receptor genes and bronchial asthma risk among Egyptian children. Clinical Biochemistry 93:66−72 doi: 10.1016/j.clinbiochem.2021.04.006

    CrossRef   Google Scholar

    [14] Li HF, Yan LP, Wang K, Li XT, Liu HX, et al. 2019. Association between ADAM33 polymorphisms and asthma risk: a systematic review and meta-analysis. Respiratory Research 20:38 doi: 10.1186/s12931-019-1006-1

    CrossRef   Google Scholar

    [15] Romieu I, Ramirez-Aguilar M, Sienra-Monge JJ, Moreno-Macías H, del Rio-Navarro BE, et al. 2006. GSTM1 and GSTP1 and respiratory health in asthmatic children exposed to ozone. European Respiratory Journal 28(5):953−59 doi: 10.1183/09031936.06.00114905

    CrossRef   Google Scholar

    [16] Castro M, Ferreira J, Sarmento D, Carvalho C, Matos A, et al. 2018. The role of GSTT1 and GSTM1 gene polymorphisms in bronchial asthma. European Respiratory Journal 52:PA4223 doi: 10.1183/13993003.congress-2018.PA4223

    CrossRef   Google Scholar

    [17] Su X, Ren Y, Li M, Kong L, Kang J. 2020. Association of glutathione S -transferase M1 and T1 genotypes with asthma: a meta-analysis. Medicine (United States) 99(34):E21732 doi: 10.1097/MD.0000000000021732

    CrossRef   Google Scholar

    [18] Osawa R, Akiyama M, Shimizu H. 2011. Filaggrin gene defects and the risk of developing allergic disorders. Allergology International 60:1−9 doi: 10.2332/allergolint.10-RAI-0270

    CrossRef   Google Scholar

    [19] Cubero JL, Isidoro-García M, Segura N, Benito Pescador D, Sanz C, et al. 2016. Filaggrin gene mutations and new SNPs in asthmatic patients: a cross-sectional study in a Spanish population. Allergy, Asthma and Clinical Immunology 12(1):31 doi: 10.1186/s13223-016-0137-x

    CrossRef   Google Scholar

    [20] Kandil R, Baldassi D, Böhlen S, Müller JT, Jürgens DC, et al. 2023. Targeted GATA3 knockdown in activated T cells via pulmonary siRNA delivery as novel therapy for allergic asthma. Journal of Controlled Release 354:305−15 doi: 10.1016/j.jconrel.2023.01.014

    CrossRef   Google Scholar

    [21] Gavitt TD, Hartmann AK, Sawant SS, Mara AB, Szczepanek SM, et al. 2021. A GATA3 targeting nucleic acid nanocapsule for in vivo gene regulation in asthma. ACS Nano 15(7):11192−201 doi: 10.1021/acsnano.0c07781

    CrossRef   Google Scholar

    [22] Kristjansdottir K, Norddahl GL, Ivarsdottir EV, Jonsdottir I, Olafsdottir TA, et al. 2024. A partial loss-of-function variant in STAT6 protects against T2 asthma. The Journal of Allergy and Clinical Immunology 155:228−35 doi: 10.1016/j.jaci.2024.10.002

    CrossRef   Google Scholar

    [23] Niessen NM, Gibson PG, Simpson JL, Scott HA, Baines KJ, et al. 2021. Airway monocyte modulation relates to tumour necrosis factor dysregulation in neutrophilic asthma. ERJ Open Research 7(3):00131-2021 doi: 10.1183/23120541.00131-2021

    CrossRef   Google Scholar

    [24] Martinez FD. 2007. CD14, endotoxin, and asthma risk: actions and interactions. Proceedings of the American Thoracic Society 4(3):221−25 doi: 10.1513/pats.200702-035AW

    CrossRef   Google Scholar

    [25] Şahin F, Yildiz P, Kuskucu A, Kuskucu MA, Karaca N, et al. 2014. The effect of CD14 and TLR4 gene polimorphisms on asthma phenotypes in adult Turkish asthma patients: a genetic study. BMC Pulmonary Medicine 14:20 doi: 10.1186/1471-2466-14-20

    CrossRef   Google Scholar

    [26] Forsström V, Toivonen L, Homil K, Waris M, Pedersen CT, et al. 2023. Association of asthma risk alleles with acute respiratory tract infections and wheezing illnesses in young children. Journal of Infectious Diseases 228(8):990−98 doi: 10.1093/infdis/jiad075

    CrossRef   Google Scholar

    [27] Cai X, Qiao Y, Diao C, Xu X, Chen Y, et al. 2014. Association between polymorphisms of the IKZF3 gene and systemic lupus erythematosus in a Chinese Han Population. PLoS One 9(10):0108661 doi: 10.1371/journal.pone.0108661

    CrossRef   Google Scholar

    [28] Khorasani AM, Mohammadi B, Saghafi MR, Mohammadi S, Ghaffari S, et al. 2023. The association between MUC5AC and MUC5B genes expression and remodeling progression in severe neutrophilic asthma: a direct relationship. Respiratory Medicine 213:107260 doi: 10.1016/J.RMED.2023.107260

    CrossRef   Google Scholar

    [29] Nowakowska J, Olechnowicz A, Langwiński W, Koteluk O, Lemańska Ż, et al. 2023. Increased expression of ORMDL3 in allergic asthma: a case control and in vitro study. Journal of Asthma 60(3):458−67 doi: 10.1080/02770903.2022.2056896

    CrossRef   Google Scholar

    [30] Qi C, Jiang Y, Yang IV, Forno E, Wang T, et al. 2020. Nasal DNA methylation profiling of asthma and rhinitis. Journal of Allergy and Clinical Immunology 145(6):1655−63 doi: 10.1016/j.jaci.2019.12.911

    CrossRef   Google Scholar

    [31] Kumar S, Chinnusamy V, Mohapatra T. 2018. Epigenetics of modified DNA bases: 5-methylcytosine and beyond. Frontiers in Genetics 9:640 doi: 10.3389/fgene.2018.00640

    CrossRef   Google Scholar

    [32] Jang HS, Shin WJ, Lee JE, Do JT. 2017. CpG and non-CpG methylation in epigenetic gene regulation and brain function. Genes 8(6):2−20 doi: 10.3390/genes8060148

    CrossRef   Google Scholar

    [33] Wang J, Hu J, Qin D, Han D, Hu J. 2024. A multi-omics Mendelian randomization identifies putatively causal genes and DNA methylation sites for asthma. World Allergy Organization Journal 17(12):101008 doi: 10.1016/j.waojou.2024.101008

    CrossRef   Google Scholar

    [34] Yang IV, Pedersen BS, Liu A, O'Connor GT, Teach SJ, et al. 2015. DNA methylation and childhood asthma in the inner city. Journal of Allergy and Clinical Immunology 136(1):69−80 doi: 10.1016/j.jaci.2015.01.025

    CrossRef   Google Scholar

    [35] Bhat S, Rotti H, Prasad K, Kabekkodu SP, Saadi AV, et al. 2023. Genome-wide DNA methylation profiling after Ayurveda intervention to bronchial asthmatics identifies differential methylation in several transcription factors with immune process related function. Journal of Ayurveda and Integrative Medicine 14(2):100692 doi: 10.1016/j.jaim.2023.100692

    CrossRef   Google Scholar

    [36] Liang W, Chen Z, Li C, Liu J, Tao J, et al. 2021. Accurate diagnosis of pulmonary nodules using a noninvasive DNA methylation test. Journal of Clinical Investigation 131(10):e145973 doi: 10.1172/JCI145973

    CrossRef   Google Scholar

    [37] Milazzo G, Mercatelli D, Di Muzio G, Triboli L, De Rosa P, et al. 2020. Histone deacetylases (HDACs): evolution, specificity, role in transcriptional complexes, and pharmacological actionability. Genes 11(5):556 doi: 10.3390/genes11050556

    CrossRef   Google Scholar

    [38] Farsetti A, Illi B, Gaetano C. 2023. How epigenetics impacts on human diseases. European Journal of Internal Medicine 114:15−22 doi: 10.1016/j.ejim.2023.05.036

    CrossRef   Google Scholar

    [39] Alaskhar Alhamwe B, Khalaila R, Wolf J, von Bülow V, Harb H, et al. 2018. Histone modifications and their role in epigenetics of atopy and allergic diseases. Allergy, Asthma and Clinical Immunology 14:39 doi: 10.1186/s13223-018-0259-4

    CrossRef   Google Scholar

    [40] Pelaia C, Heffler E, Crimi C, Maglio A, Vatrella A, et al. 2022. Interleukins 4 and 13 in asthma: key pathophysiologic cytokines and druggable molecular targets. Frontiers in Pharmacology 13:851940 doi: 10.3389/fphar.2022.851940

    CrossRef   Google Scholar

    [41] Berthon BS, McLoughlin RF, Jensen ME, Hosseini B, Williams EJ, et al. 2021. The effects of increasing fruit and vegetable intake in children with asthma: a randomized controlled trial. Clinical and Experimental Allergy 51(9):1144−56 doi: 10.1111/cea.13979

    CrossRef   Google Scholar

    [42] Seumois G, Chavez L, Gerasimova A, Lienhard M, Omran N, et al. 2014. Epigenomic analysis of primary human T cells reveals enhancers associated with TH2 memory cell differentiation and asthma susceptibility. Nature Immunology 15(8):777−88 doi: 10.1038/ni.2937

    CrossRef   Google Scholar

    [43] Ratti M, Lampis A, Ghidini M, Salati M, Mirchev MB, et al. 2020. MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) as new tools for cancer therapy: first steps from bench to bedside. Targeted Oncology 15(3):261−78 doi: 10.1007/s11523-020-00717-x

    CrossRef   Google Scholar

    [44] Taka S, Tzani-Tzanopoulou P, Wanstall H, Papadopoulos NG. 2020. MicroRNAs in asthma and respiratory infections: Identifying common pathways. Allergy, Asthma & Immunology Research 12(1):4−23 doi: 10.4168/aair.2020.12.1.4

    CrossRef   Google Scholar

    [45] Sharma R, Tiwari A, McGeachie MJ. 2022. Recent miRNA Research in Asthma. Current Allergy and Asthma Reports 22(12):231−58 doi: 10.1007/s11882-022-01050-1

    CrossRef   Google Scholar

    [46] Zheng R, Gao F, Xiao Y, Liang J, Mao Z, et al. 2024. PM2.5-derived exosomal long noncoding RNA PAET participates in childhood asthma by enhancing DNA damage via m6A-dependent OXPHOS regulation. Environment International 183:108386 doi: 10.1016/j.envint.2023.108386

    CrossRef   Google Scholar

    [47] Wang AL, Gruzieva O, Qiu W, Merid SK, Celedón JC, et al. 2019. DNA methylation is associated with inhaled corticosteroid response in persistent childhood asthmatics. Clinical & Experimental Allergy 49(9):1225−34 doi: 10.1111/cea.13447

    CrossRef   Google Scholar

    [48] Islam R, Dash D, Singh R. 2023. An antioxidant ameliorates allergic airway inflammation by inhibiting HDAC 1 via HIF-1α/VEGF axis suppression in mice. Scientific Reports 13:9637 doi: 10.1038/s41598-023-36678-0

    CrossRef   Google Scholar

    [49] Li ML, Su XM, Ren Y, Zhao X, Kong LF, et al. 2020. HDAC8 inhibitor attenuates airway responses to antigen stimulus through synchronously suppressing galectin-3 expression and reducing macrophage-2 polarization. Respiratory Research 21:62 doi: 10.1186/s12931-020-1322-5

    CrossRef   Google Scholar

    [50] Zhang Z, Wang G, Li Y, Lei D, Xiang J, et al. 2022. Recent progress in DNA methyltransferase inhibitors as anticancer agents. Frontiers in Pharmacology 13:1072651 doi: 10.3389/fphar.2022.1072651

    CrossRef   Google Scholar

    [51] Xu CJ, Söderhäll C, Bustamante M, Baïz N, Gruzieva O, et al. 2018. DNA methylation in childhood asthma: an epigenome-wide meta-analysis. The Lancet Respiratory Medicine 6(5):379−88 doi: 10.1016/S2213-2600(18)30052-3

    CrossRef   Google Scholar

    [52] Sun J, Wang J, Zheng D, Hu X. 2020. Advances in therapeutic application of CRISPR-Cas9. Briefings in Functional Genomics 19(3):164−74 doi: 10.1093/bfgp/elz031

    CrossRef   Google Scholar

  • Cite this article

    Soni P, Edwin D'Souza I. 2025. Epigenetic changes in childhood asthma: a path to precision medicine. Epigenetics Insights 18: e007 doi: 10.48130/epi-0025-0005
    Soni P, Edwin D'Souza I. 2025. Epigenetic changes in childhood asthma: a path to precision medicine. Epigenetics Insights 18: e007 doi: 10.48130/epi-0025-0005

Tables(3)

Article Metrics

Article views(2600) PDF downloads(584)

Other Articles By Authors

MINI REVIEW   Open Access    

Epigenetic changes in childhood asthma: a path to precision medicine

Epigenetics Insights  18 Article number: e007  (2025)  |  Cite this article

Abstract: The 2023 Global Initiative for Asthma (GINA) report highlights a significant rise in asthma rates particularly in low- and middle-income countries, with urbanization and exposure to environmental pollutants being key contributing factors. Children exposed to environmental influences throughout their critical developmental periods may experience epigenetic changes that could predispose them to asthma in the future. However, there are still issues implementing these findings in clinical settings, necessitating more research and technological advancements. The knowledge of epigenetics in childhood asthma might bridge the gap in knowledge of disease etiology and help the creation of precision medicine approaches. By integrating epigenetic indicators into diagnostic and therapy methods, clinicians can move closer to customized care, benefiting children with asthma. This review emphasizes the significance of epigenetic mechanisms in the development of pediatric asthma and the important associated genes like ORAI1, IL4, ADAM33, GSTM1, FLG, GATA3, STAT6, TNF, CD14, IKZF3, MUC5AC, and ORMDL3. We will also review the role of important epigenetic changes including non-coding RNAs, DNA methylation, and histone modifications in the pathophysiology of asthma. Furthermore, the potential of epigenetics as biomarkers for the diagnosis and prognosis of asthma and the therapeutic uses of epigenetic-based therapies such as DNA methyltransferase inhibitors and histone deacetylase (HDAC) inhibitors have been explored.

    • Childhood asthma is a serious respiratory condition predominantly affecting the airways, causing airway constriction and chronic inflammation interfering with normal respiratory function presenting clinically with chest tightness, coughing, wheezing, and dyspnoea[1]. According to the Global Initiative for Asthma's (GINA) 2023 report, urbanization, and environmental pollutants are recognized as the major causes of notable prevalence in many low- and middle-income nations[2]. It is worrisome to note that childhood asthma is one of the leading causes of school absences, emergency department admissions, and hospitalizations with the incidence of asthma increasing in developing countries[2,3].

      Recent scientific explorations into the pathogenesis of asthma have revealed it to possess a very complex and multitiered foundation, influenced by genetic and environmental factors causing chronic airway inflammation, airway hyperreactivity (AHR), and irreversible airway remodeling, mucus hypersecretion, subepithelial fibrosis, and airway remodeling[35]. Asthma susceptibility has been linked to many critical genes, with important loci found on chromosomes 2, 6, 9, 15, 17, and 22. ORMDL3/GSDMB is the most prevalent gene found in these genomic areas, and it plays a crucial role in the etiology of asthma. This chromosome 17 gene has been closely linked to childhood asthma, suggesting that it plays a crucial role in the initiation and development of asthma in young people[6].

      Epigenetics is defined as the study of the interaction of the environment and the functioning of the genome without changing the genome sequence[7]. The gene-environment interaction hypothesis holds that the functions of many known genes are determined by environmental factors and that epigenetic modification is an important mechanism underlying the interplay between environmental and genetic factors[5]. The three major types of epigenetic regulation include histone modifications, DNA methylation, and non-coding RNAs (ncRNAs), such as microRNAs[7,8]. Research reveals that epigenetic approaches targeting specific genes can identify potentially treat life-threatening illnesses like cardiovascular disease, cancer, neurodevelopmental, and neurodegenerative disorders. The astounding increase observed in the incidence, prevalence, and severity of asthma in the past few decades strongly substantiates the claim that environmental exposure plays a titular role in the pathogenesis of asthma, especially via their interactions with the genetic variants[4]. Despite ongoing research and an improved understanding, the prevalence of asthma has been rising in recent years with the absence of current epigenetic-based treatments.

      This review explores the potential of epigenetic-based treatments for the condition. It gives a thorough summary of the state of research on the relationship between epigenetics and childhood asthma focusing on the processes of non-coding RNAs, histone modifications, and DNA methylation. This article also discusses the therapeutic potential of epigenetics, including the application of medicines that target enzymes like DNMT inhibitors and HDACs.

    • Many genes have been found to impact airway inflammation, remodeling, and immunological responses, and along with environmental stressors like pollution, tobacco smoke, and allergens, these genetic variants increase the incidence of asthma[9,10]. By understanding the epigenetic processes that control these genetic variations, they can be targeted for proper diagnosis, treatment, and prevention of childhood asthma. Table 1 lists the main genes linked to asthma and their roles in the underlying pathophysiology.

      Table 1.  Genes associated with asthma pathogenesis and their genetic variations.

      Gene symbol Gene function Association with asthma pathogenesis Etiology of genetic variations
      ORAI1[11] Calcium ion regulation, T-cell activation, cytokine production. Variants linked to increased asthma susceptibility; especially, related to the immune response to allergens. Genetic mutations and allergens can influence its expression and immune response.
      IL4[12,13] Promotes differentiation of T-helper cells, leading to Th2-mediated inflammation IL4 drives IgE production and allergic responses, contributing to asthma; especially, allergic asthma. Genetic mutations and environmental allergens like pollen and dust mites increase IL4 expression.
      ADAM33[14] Involved in airway remodeling and smooth muscle regulation Contribute to airway hyperresponsiveness and remodeling features of asthma. Genetic changes; tobacco smoke and air pollution can trigger ADAM33 activation.
      GSTM1[1517] Detoxifies harmful substances like environmental pollutants GSTM1 gene deletion is associated with asthma risk; especially, in children exposed to pollution. Genetic deletion of GSTM1 and environmental pollutants (e.g. tobacco smoke).
      FLG (Filaggrin)[18,19] Maintains skin integrity and prevents allergens entering to the body FLG mutations are linked to asthma; especially, in children with eczema, due to a weakened skin barrier. Genetic mutations; allergens or irritants like harsh chemicals and pollution.
      GATA3[20,21] Transcription factor regulating Th2 differentiation. GATA3 variants are associated with allergic asthma and Th2-mediated inflammation. Genetic variants; and environmental allergens contribute to GATA3 overexpression.
      STAT6[22] Transcription factor helps in Th2 differentiation and cytokine production in allergic responses. Variants increase susceptibility to asthma, especially in children with allergic tendencies. Genetic variants; and environmental allergens like pollen can promote STAT6 activation.
      TNF[23] Pro-inflammatory cytokine, immune activation & airway inflammation. Variants linked to asthma, particularly through increased airway inflammation and tissue remodeling. Genetic mutations; air pollution and allergens exacerbate TNF expression.
      CD14[24,25] Receptor activates immune responses to environmental triggers, such as bacteria and allergens. Variants in CD14 are associated with asthma, particularly in children exposed to environmental allergens. Genetic changes; and environmental exposures (e.g., dust mites, mold) increase CD14-mediated immune activation.
      IKZF3[26,27] Regulates immune cell differentiation and activation. Contribute to asthma susceptibility, influencing immune responses. Genetic predisposition; environmental allergens.
      MUC5AC[28] Secreted protein involved in mucus production in the airways. Overexpression of MUC5AC is associated with asthma and excessive mucus production. Genetic variation; tobacco smoke and air pollution can upregulate MUC5AC production.
      ORMDL3[29] Regulates endoplasmic reticulum stress and inflammation. It plays a role in maintaining cellular homeostasis and regulating immune response. Variants in ORDLM3 are associated with asthma; particularly, childhood asthma. The gene contributes to airway inflammation and immune response dysregulation by regulating cytokine synthesis and immune cell activation. Genetic variations, incorporating single nucleotide polymorphisms (SNPs); and environmental factors (e.g. allergens) may influence gene expression and immune response.
    • DNA methylation is an epigenetic process the refers to the addition of a methylgroup to DNA, which may regulate gene expression[30]. Around 1.5% of the genome contains 5-methylcytosine (5-mC), a change that occurs when a methyl group is introduced to the cytosine base's 5th carbon. Although more frequently found at CpG sites, 5-mC can exist at non-CpG sites; especially, in embryonic stem cells. Some CpG sites are unmethylated, but most are methylated because methylation inhibits transcriptional enzymes from linking to DNA, suppressing gene expression[31,32]. DNA methylation in adaptive evolution is closely associated with the potential ability to produce novel pathways with DNA methylation for phenotypic diversity.

      Wang et al., pinpointed positive causal methylation sites related to asthma. The report identified three genes—ETS1, ITPKB, and JAK2, which had eight distinct DNA methylation sites. Two methylation sites, cg16265553 and cg13661497 downregulated ITPKB expression and inhibited JAK2 expression respectively[33]. This suggests that DNA methylation influences gene expression, which influences the risk of asthma. Potential drug therapies targeting CEP95, RBM6, ITPKB, ETS1, and JAK2 may be formulated for asthma.

      A case-control study showed 81 regions that were differentially methylated in asthmatic patients. The authors reported 11 IgE-associated differentially methylated regions (DMR). The association of methylation changes of KLF 6 identifies novel genes and networks that are involved in the pathogenesis of asthma[34]. While Wang et al.[33] employed Mendelian Randomization (MR), Yang et al.[34] built on the understanding that the cause of asthma is associated with non-Mendelian inheritance. The potential of DNA methylation for treatment was highlighted in the use of classical Ayurvedic treatment which follows a holistic therapeutic approach rather than targeting any specific type of cell. Bhat et al.[35] reported that the signaling pathways of Neurotrophin TRK receptors, neurotrophin, ERBB, and epidermal growth factor receptors were enriched for differentially methylated genes in bronchial asthmatics. The Pulmo Seek model uses targeted DNA methylation sequencing with a blood-based test which is less invasive and a cost-effective strategy for identifying early-stage lung cancer. A similar, strategy in improving early diagnosis, and tracking the treatment's efficacy to manage asthma can be offered[36].

    • HDACs play a critical role in epigenetic control by modifying gene expression through chemical alterations that do not affect the underlying DNA sequence. These alterations may result in notable phenotypic shifts. Histone acetyl groups are eliminated by HDACs, which alters chromatin shape and gene activity. Deacetylation has a role in inflammation and many cellular processes like differentiation, proliferation, and gene expression[37]. These functions demonstrate the significance of HDACs in preserving cellular homeostasis and their potential as therapeutic targets[38]. A key function of histone acetylation and deacetylation is to control gene accessibility via modification of chromatin structure. A more relaxed chromatin structure caused by histone acetylation makes it possible for transcription factors to attach to genes and increase their expression[39]. Histone acetylation at loci such as IL-4 and IL-13 in asthma exacerbates airway inflammation by promoting Th2-driven inflammation. On the other hand, chromatin condenses due to histone deacetylation, which inhibits gene expression[40]. Targeting histone-modifying enzymes may alter chromatin accessibility, which could help control immunological responses and inflammation in asthma.

      An in-vitro demonstration in asthmatic mice demonstrated that histone hyperacetylation of ORMDL3 is associated with asthma and an increase in ORMDL 3 expression is associated with airway remodeling. A mouse model of asthma demonstrated that inhibiting p300 with C646, ORMDL3 expression, airway hyperactivity (AHR), and remodeling were decreased, indicating that p300 targeting might provide a treatment approach for childhood asthma[5]. A randomized controlled trial of increasing fruit and vegetable intake in asthmatic children did not impact asthma exacerbations over 6 months; however, a high F & V diet could modify histone modifications and related anti-inflammatory pathways. The authors found alterations in the HDAC activity of peripheral blood mononuclear cells (PBMC) and the expression of genes linked to G-protein coupled receptors (GPR41/43)[41].

      A genome-wide mapping of histone modifications revealed that a considerable number of asthma-related single nucleotide polymorphisms (SNPs) were located on Th2 cell enhancers, a region in which the highest rate of H3K4 demethylation was measured in primary human CD4 + T-cell. In addition to the TH 2-associated cytokines IL-4, IL-5, and IL-13, the TH 9-associated cytokines IL-9, IL-10, and IL-21 are increased[42]. These studies suggest chromatin remodeling and histone alterations can provide therapeutic options for asthmatic children by providing a novel way to influence the underlying molecular mechanisms of asthma by altering the epigenetic regulation of gene expression. Normal gene expression patterns linked to inflammation and immunological responses may be restored by modifying histone modifications and the chromatin landscape; thus, alleviating the asthma symptoms and worsening of disease process.

    • Long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) are examples of ncRNAs that do not code for proteins. These non-coding RNAs are important for the regulation of many biological functions. Studies have demonstrated the importance of ncRNAs in asthma as lncRNAs and miRNAs mostly regulate asthma gene expression. miRNAs such as miR-155 and lncRNAs affect immunological responses and increase airway hyperreactivity through transcriptional and post-transcriptional modulation of gene expression[43]. Children with asthma have distinct levels of miRNA and lncRNA expression compared to healthy controls, suggesting that these molecules may be used as diagnostic biomarkers and therapeutic targets[44,45] miRNAs are specifically linked to smooth muscle proliferation and airway inflammation.

      Recent research has demonstrated the role of circRNAs in airway remodeling and smooth muscle cell proliferation; essential for the development of asthma. In vitro and in vivo models showed NORAD silencing decreased the levels of β-catenin and c-Myc.RCC2 upregulation abrogated the influence of NORAD downregulation on their expression levels in TGF-β1-induced BEAS-2B cells. These results suggested that the silence of NORAD inactivated the Wnt/β-catenin pathway by suppressing RCC2 in asthma[3]. A novel mechanism was suggested via which human bronchial epithelial (HBE) cells treated with exosomal lncRNA PAET accelerate DNA damage and contribute to PM2.5-induced pediatric asthma. lncRNA PAET was markedly elevated in exosomes produced from PM2.5-treated HBE cells in children with asthma which interferes with oxidative phosphorylation (OXPHOS) by increasing the m6A modification of COX4I1 mRNA through the regulation of METTL3 stability. Reduced COX4I1 expression and inhibition of OXOPHOS, in turn, raised reactive oxygen species (ROS) levels and caused DNA damage in recipient HBE cells[46].

    • Environmental exposures such as tobacco smoke, air pollution, vehicle exhaust, and pesticides, can significantly alter the epigenome and increase the risk of asthma in children (Table 2)[7]. These pollutants induce oxidative stress, altering miRNA expression, DNA methylation, and histone modifications later in life.

      Table 2.  Implications of environmental variables on childhood asthma and pertinent epigenetic initiatives[7].

      Environmental trigger Percentage of children affected Epigenetic mechanisms involved
      Passive smoking 20%–30% DNA methylation of immune genes (e.g., AHRR, FOXP3), altered miRNA profiles.
      Pollutants from air conditioners ~15% Oxidative stress-driven histone modifications and DNA methylation.
      Pollutants from refrigerators 10%–12% Non-coding RNA dysregulation, DNA methylation of inflammatory pathways.
      Mosquito repellent machines ~10% Disruption of immune gene regulation via non-coding RNAs.
      Vehicle emissions 20%–40% Global hypomethylation, site-specific methylation (e.g., IL-4, IFN-γ genes).
    • The role of epigenetics in disease has become a special focus of research. Epigenetic modifications profoundly affect gene expression and aberrant modifications have been linked to various diseases[7,38]. The rate at which environmental exposure brings changes in DNA sequences can explained by epigenetics[4]. Epigenetic modifications like ncRNAs, histone modifications, and DNA methylation are reversible, functionally relevant changes that do not impact the nucleotide sequence of the DNA[4,7,30].

    • In addition to being identified in several malignancies, aberrant DNA methylation patterns are increasingly becoming recognized as early markers of respiratory conditions including asthma. A DNA methylome analysis of PBMCs from 394 patients with mild-to-moderate asthma, showed that hypomethylation of the precortistatin (CORT) and IL-12 subunit β (IL12b) genes were associated with fewer hospitalizations and less use of oral corticosteroids, respectively, suggesting that these genes could be used as prognostic biomarkers[47]. DNA methylation might be a biomarker for IgE sensitization. The authors indicated IgE stratified analysis of replicated CpGs showed that DNA methylation signals in the nasal epithelium were mainly driven by IgE-positive subjects with asthma and not by IgE negative asthma subjects[30].

    • Histone modification changes are being studied as possible biomarkers for the early diagnosis of malignancies since they can significantly affect gene expression. Genes like ARDID5B, SENS1, and XPA are implicated in cellular senescence, cell cycle regulation, and DNA repair pathways, indicating their potential as biomarkers for Alzheimer's disease (AD)[5]. However, little is known about histone changes or modifications in asthma. The results of the previous study suggest that histone alterations might be promising as asthma biomarkers, but more substantial evidence is required to use it.

    • Non-coding RNAs have been extensively studied as potential biomarkers in the context of different diseases. Recent animal and even human studies have revealed a potentially significant role of miRNAs in asthma pathogenesis[4]. Non-coding RNAs have been established to play an important role in gene expression control, either as transcriptional or post-transcriptional regulators. miRNAs can regulate key genes involved in the development of cancer, thus influencing tumor growth, invasion, and metastasis by increasing the activation of oncogenic pathways and limiting the expression of tumor suppressors[43].

    • Histone deacetylases (HDACs) are evolutionary conserved enzymes which operate by removing acetyl groups from histones and other protein regulatory factors, with functional consequences on chromatin remodeling and gene expression profiles[37]. A recent study demonstrated reduced oxidative stress, airway inflammation, and remodeling in an ovalbumin-induced asthmatic mouse model by HDAC inhibitors, curcumin (CUR), and sodium butyrate (SOB). This may be achieved by modifying the PI3K/Akt and HIF-1α/VEGF signaling pathways[48]. Li et al. also demonstrated reduced allergic airway inflammation and hyperresponsiveness in an allergic asthma model exposed to ovalbumin by HDAC8 inhibitors[49].

    • DNA methyltransferase inhibitors have shown promising results by regulating immunological responses; especially, in treating cancer and respiratory conditions including COPD and asthma. DNMT inhibitors act by inhibiting DNA methyltransferases, enzymes that add methyl groups to DNA; thus, reactivating silenced genes, including those crucial for immune responses[50,51].

    • The future of pediatric asthma treatment lies in leveraging advances in epigenetics for precision medicine. Key directions include the identification of personalized epigenetic biomarkers for early diagnosis and monitoring, allowing tailored treatments for individual patients. Targeting epigenetic modifications, such as DNA methylation, histone modification, and ncRNAs, offers potential therapies to reverse aberrant gene expression linked to asthma. The integration of epigenetic biomarkers into clinical practice could revolutionize asthma management. DNA methylation signatures, such as those identified in the ALOX5 gene, SOCS1, and RUNX3 offer the potential for stratifying patients into phenotypic subgroups and predicting therapeutic responses[51].

      Early identification of such markers in cord blood or peripheral blood can guide the prevention of the same. It has been demonstrated that probiotics and omega-3 fatty acid supplementation during pregnancy lowers the risk by lowering inflammatory indicators and fostering immune balance in the fetus. The immune system is better able to tolerate and reduce epigenetic changes that lead to asthma when vaginal delivery is encouraged, which increases exposure to naturally occurring bacteria while lowering the need for antibiotics throughout infancy. Furthermore, asthma will be impacted by single-cell epigenomic approaches (Table 3), which will allow for more targeted treatments. Lastly, investigating how environmental influences and epigenetic pathways interact will result in the development of better preventative and therapeutic approaches.

      Table 3.  Techniques favoring the study of cellular diversity, enabling the analysis of heterogeneity in tissues, uncovering rare cell populations.

      Technique name Corresponding description
      Single-cell RNA sequencing
      (scRNA-seq)
      Efficient to measure the transcriptome of individual cells further cells are isolated, and RNA is reverse transcribed, and sequenced.
      Single-cell DNA sequencing First in hand to analyze individual cell genomes for mutations, copy number variations, and rearrangements.
      Single-cell epigenomics Studies gene regulation through chromatin accessibility (ATAC-seq), protein-DNA interactions (ChIP-seq), and DNA methylation profiling.
      Spatial transcriptomics Within the tissue architecture, this technique maps gene expression with spatial information.
      Single-cell proteomics Applying advanced mass spectrometry, protein expression, and modifications are measured, as methods like CyTOF.

      Despite providing revolutionary insights, epigenetics still faces obstacles, such as the requirement for long-term research to prove causal links and the standardization of epigenetic analytic techniques. Due to genetic diversity, large-scale research is necessary to identify healthy biomarkers and validate therapies. The fundamental study is completed until clinical applications require overcoming ethical and legal barriers.

      The clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) system are one of the most important strategies that contain palindromic repeats separated by short stretches of DNA called spacers. Spacers from foreign nucleic acids are incorporated to provide immunity against specific diseases[52]. The therapeutic application of CRISPR-Cas9 has raised great hope for curing diseases.

    • By examining the epigenetic alterations in childhood asthma, this review sheds light on the underlying mechanisms of the illness and the newer possibilities for precision therapy. These biomarkers have the potential for early diagnosis, risk assessment, and treatment of asthma; especially, DNA methylation, histone modifications, and ncRNAs. Epigenetic-based therapies can be made possible by advances in epigenomic technologies raising the prospect of individualized and focused treatments to enhance asthma control in children.

      • The authors have not declared a specific grant for this research from any funding agency in the public, commercial, or not-for-profit sectors.

      • The authors confirm contribution to the paper as follows: study conception and design: Soni P; draft manuscript preparation, editing & revision: Soni P, D'Souza E (lead); compiled the predictive models for asthma exacerbations: D'Souza E. Both authors reviewed the results and approved the final version of the manuscript.

      • Data sharing not applicable to this article as no datasets were generated or analyzed during the current study.

      • The authors declare that they have no conflict of interest.

      • Copyright: © 2025 by the author(s). Published by Maximum Academic Press, Fayetteville, GA. This article is an open access article distributed under Creative Commons Attribution License (CC BY 4.0), visit https://creativecommons.org/licenses/by/4.0/.
    Table (3) References (52)
  • About this article
    Cite this article
    Soni P, Edwin D'Souza I. 2025. Epigenetic changes in childhood asthma: a path to precision medicine. Epigenetics Insights 18: e007 doi: 10.48130/epi-0025-0005
    Soni P, Edwin D'Souza I. 2025. Epigenetic changes in childhood asthma: a path to precision medicine. Epigenetics Insights 18: e007 doi: 10.48130/epi-0025-0005

Catalog

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return