Reviews and feature article
A translational perspective on epigenetics in allergic diseases

https://doi.org/10.1016/j.jaci.2018.07.009Get rights and content

Information for Category 1 CME Credit

Credit can now be obtained, free for a limited time, by reading the review articles in this issue. Please note the following instructions.

Method of Physician Participation in Learning Process: The core material for these activities can be read in this issue of the Journal or online at the JACI Web site: www.jacionline.org. The accompanying tests may only be submitted online at www.jacionline.org. Fax or other copies will not be accepted.

Date of Original Release: September 2018. Credit may be obtained for these courses until August 31, 2019.

Copyright Statement: Copyright © 2018-2019. All rights reserved.

Overall Purpose/Goal: To provide excellent reviews on key aspects of allergic disease to those who research, treat, or manage allergic disease.

Target Audience: Physicians and researchers within the field of allergic disease.

Accreditation/Provider Statements and Credit Designation: The American Academy of Allergy, Asthma & Immunology (AAAAI) is accredited by the Accreditation Council for Continuing Medical Education (ACCME) to provide continuing medical education for physicians. The AAAAI designates this journal-based CME activity for a maximum of 1.00 AMA PRA Category 1 Credit™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

List of Design Committee Members: Jörg Tost, PhD (author); Zuhair K. Ballas, MD (editor)

Disclosure of Significant Relationships with Relevant Commercial Companies/Organizations: Work on food allergy in the laboratory of J. Tost is partially funded by DBV Technologies, and J. Tost had conference and travel fees covered by DBV Technologies. The work in the laboratory of J. Tost is further supported by the following grants unrelated to the presented work: ANR (ANR-13-EPIG-0003-05 and ANR-13-CESA-0011-05), Aviesan/INSERM (EPIG2014-01 and EPlG2014-18), INCa (PRT-K14-049), a Sirius research award (UCB Pharma S.A.), a Passerelle research award (Pfizer), iCARE (MSD Avenir), and the institutional budget of the CNRGH. Z. K. Ballas (editor) disclosed no relevant financial relationships.

Activity Objectives:

  • 1.

    To understand the concept of epigenetics and the role of epigenetic modifications in shaping the incidence and phenotype of allergic diseases.

  • 2.

    To know the cellular and molecular processes involved in modification of the epigenome.

  • 3.

    To understand the ways by which the environment, nutrients, and the microbiome can affect the development and course of allergic diseases through alterations in the epigenetic code.

Recognition of Commercial Support: This CME activity has not received external commercial support.

List of CME Exam Authors: Christin Deal, MD, Lisa Kohn, MD, Mona Liu, MD, Monica Tsai, MD, and Maria Garcia-Lloret, MD

Disclosure of Significant Relationships with Relevant Commercial

Companies/Organizations: The exam authors disclosed no relevant financial relationships.

The analysis of epigenetic modifications in allergic diseases has recently attracted substantial interest because epigenetic modifications can mediate the effects of the environment on the development of or protection from allergic diseases. Furthermore, recent research has provided evidence for an altered epigenomic landscape in disease-relevant cell populations. Although still in the early phase, epigenetic modifications, particularly DNA methylation and microRNAs, might have potential for assisting in the stratification of patients for treatment and complement or replace in the future biochemical or clinical tests. The first epigenetic biomarkers correlating with the successful outcome of immunotherapy have been reported, and with personalized treatment options being rolled out, epigenetic modifications might well play a role in monitoring or even predicting the response to tailored therapy. However, further studies in larger cohorts with well-defined phenotypes in specific cell populations need to be performed before their implementation. Furthermore, the epigenome provides an interesting target for therapeutic intervention, with microRNA mimics, inhibitors, and antisense oligonucleotides being evaluated in clinical trials in patients with other diseases. Selection or engineering of populations of extracellular vesicles and epigenetic editing represent novel tools for modulation of the cellular phenotype and responses, although further technological improvements are required. Moreover, interactions between the host epigenome and the microbiome are increasingly recognized, and interventions of the microbiome could contribute to modulation of the epigenome with a potential effect on the overall goal of prevention of allergic diseases.

Section snippets

DNA methylation

A number of epigenome-wide association studies (EWASs) have been performed and have identified, at least in asthmatic patients, common themes, such as the importance of eosinophils and regulatory T (Treg) cells and probably an altered epigenetic state of these cells.9, 10, 11 Large-scale EWASs have also shown that disease-predisposing environmental factors, such as prenatal maternal smoking or prenatal or postnatal air pollution, lead to DNA methylation changes at genes with relevance for

miRNAs and extracellular vesicles

miRNAs constitute an additional level of epigenetic regulation influencing gene expression without altering the genomic sequence. miRNAs are small noncoding RNA molecules (22-25 nucleotides) that posttranscriptionally regulate gene expression by blocking mRNA translation and/or altering the stability of the mRNA by binding perfectly or with mismatches to the 3′ untranslated region of the mRNA (Fig 2).87 Each miRNA can potentially directly or indirectly regulate many target genes, and each gene

Conclusions

Investigating epigenetics will further our understanding of the disease mechanisms, and some epigenetic marks might be useful as biomarkers to guide clinical decisions for personalized management of allergic diseases, including the detection, management, and ideally prevention of these diseases. Because most published studies are limited to small exploratory cohorts and therefore had low statistical power, systematic inclusion of the analysis of epigenetic modifications, in particular DNA

References (126)

  • E.R. Bleecker et al.

    Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting beta2-agonists (SIROCCO): a randomised, multicentre, placebo-controlled phase 3 trial

    Lancet

    (2016)
  • J.M. FitzGerald et al.

    Benralizumab, an anti-interleukin-5 receptor alpha monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): a randomised, double-blind, placebo-controlled phase 3 trial

    Lancet

    (2016)
  • C. Xiao et al.

    Vanin-1 expression and methylation discriminate pediatric asthma corticosteroid treatment response

    J Allergy Clin Immunol

    (2015)
  • K.J. Baines et al.

    Sputum gene expression signature of 6 biomarkers discriminates asthma inflammatory phenotypes

    J Allergy Clin Immunol

    (2014)
  • T. Kouzarides

    Chromatin modifications and their function

    Cell

    (2007)
  • T. Colley et al.

    Defective sirtuin-1 increases IL-4 expression through acetylation of GATA-3 in patients with severe asthma

    J Allergy Clin Immunol

    (2016)
  • H.Y. Jang et al.

    Overexpression of sirtuin 6 suppresses allergic airway inflammation through deacetylation of GATA3

    J Allergy Clin Immunol

    (2016)
  • S. Brand et al.

    DNA methylation of TH1/TH2 cytokine genes affects sensitization and progress of experimental asthma

    J Allergy Clin Immunol

    (2012)
  • K.S. Oh et al.

    Anti-inflammatory chromatinscape suggests alternative mechanisms of glucocorticoid receptor action

    Immunity

    (2017)
  • P. Bhavsar et al.

    The role of histone deacetylases in asthma and allergic diseases

    J Allergy Clin Immunol

    (2008)
  • Y. Kobayashi et al.

    Passive smoking impairs histone deacetylase-2 in children with severe asthma

    Chest

    (2014)
  • X.S. Liu et al.

    Editing DNA methylation in the mammalian genome

    Cell

    (2016)
  • V. Woo et al.

    Host-microbiota interactions: epigenomic regulation

    Curr Opin Immunol

    (2017)
  • Y.J. Huang et al.

    The microbiome in allergic disease: current understanding and future opportunities-2017 PRACTALL document of the American Academy of Allergy, Asthma & Immunology and the European Academy of Allergy and Clinical Immunology

    J Allergy Clin Immunol

    (2017)
  • D.R. Wesemann et al.

    The microbiome, timing, and barrier function in the context of allergic disease

    Immunity

    (2016)
  • J. Park et al.

    Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway

    Mucosal Immunol

    (2015)
  • S. Bunyavanich et al.

    Early-life gut microbiome composition and milk allergy resolution

    J Allergy Clin Immunol

    (2016)
  • G. Du Toit et al.

    Food allergy: update on prevention and tolerance

    J Allergy Clin Immunol

    (2018)
  • T. Brick et al.

    Omega-3 fatty acids contribute to the asthma-protective effect of unprocessed cow's milk

    J Allergy Clin Immunol

    (2016)
  • T.X. Lu et al.

    MicroRNA signature in patients with eosinophilic esophagitis, reversibility with glucocorticoids, and assessment as disease biomarkers

    J Allergy Clin Immunol

    (2012)
  • T. Maes et al.

    Asthma inflammatory phenotypes show differential microRNA expression in sputum

    J Allergy Clin Immunol

    (2016)
  • D.W. Greening et al.

    Exosomes and their roles in immune regulation and cancer

    Semin Cell Dev Biol

    (2015)
  • H. Renz et al.

    Food allergy

    Nat Rev Dis Primers

    (2018)
  • N.J. Osborne et al.

    Prevalence of challenge-proven IgE-mediated food allergy using population-based sampling and predetermined challenge criteria in infants

    J Allergy Clin Immunol

    (2011)
  • R. Gupta et al.

    The economic impact of childhood food allergy in the United States

    JAMA Pediatr

    (2013)
  • A. Bird

    Perceptions of epigenetics

    Nature

    (2007)
  • D.P. Potaczek et al.

    Epigenetics and allergy: from basic mechanisms to clinical applications

    Epigenomics

    (2017)
  • R. Arathimos et al.

    Epigenome-wide association study of asthma and wheeze in childhood and adolescence

    Clin Epigenet

    (2017)
  • K.J. Brunst et al.

    Forkhead box protein 3 (FOXP3) hypermethylation is associated with diesel exhaust exposure and risk for childhood asthma

    J Allergy Clin Immunol

    (2013)
  • O. Gruzieva et al.

    Epigenome-wide meta-analysis of methylation in children related to prenatal NO2 air pollution exposure

    Environ Health Perspect

    (2017)
  • J. Nicodemus-Johnson et al.

    DNA methylation in lung cells is associated with asthma endotypes and genetic risk

    JCI Insight

    (2016)
  • I.V. Yang et al.

    The nasal methylome: a key to understanding allergic asthma

    Am J Respir Crit Care Med

    (2017)
  • L. Mondoulet et al.

    Gata3 hypermethylation and Foxp3 hypomethylation are associated with sustained protection and bystander effect following epicutaneous immunotherapy in peanut-sensitized mice

    Allergy

    (2018)
  • L. Liang et al.

    An epigenome-wide association study of total serum immunoglobulin E concentration

    Nature

    (2015)
  • N.C. Petrus et al.

    Cow's milk allergy in Dutch children: an epigenetic pilot survey

    Clin Transl Allergy

    (2016)
  • D. Martino et al.

    Epigenome-wide association study reveals longitudinally stable DNA methylation differences in CD4+ T cells from children with IgE-mediated food allergy

    Epigenetics

    (2014)
  • C.E. Nestor et al.

    DNA methylation changes separate allergic patients from healthy controls and may reflect altered CD4+ T-cell population structure

    PLoS Genet

    (2014)
  • D. Martino et al.

    Blood DNA methylation biomarkers predict clinical reactivity in food-sensitized infants

    J Allergy Clin Immunol

    (2015)
  • J.A. Dantzer et al.

    The use of omalizumab in allergen immunotherapy

    Clin Exp Allergy

    (2018)
  • M.L. Fajt et al.

    Asthma phenotypes and the use of biologic medications in asthma and allergic disease: the next steps toward personalized care

    J Allergy Clin Immunol

    (2015)
  • Cited by (46)

    • Impact of climate change on immune responses and barrier defense

      2024, Journal of Allergy and Clinical Immunology
    • Developments and emerging technologies in allergic and immunologic disease management

      2022, Allergic and Immunologic Diseases: A Practical Guide to the Evaluation, Diagnosis and Management of Allergic and Immunologic Diseases
    • CircRNA expression profiles and circRNA-miRNA-mRNA crosstalk in allergic rhinitis

      2021, World Allergy Organization Journal
      Citation Excerpt :

      Indeed, circRNAs can function as ceRNAs to sponge miRNAs14 and thereby manipulate transcription and alternative splicing,20 cooperate with RNA-binding proteins,21 and affect protein translation.22 CircRNAs can also control the cellular levels of specific miRNAs,23 the majority of which harbor highly conserved sequences and act in multiple cellular processes via post-transcriptional regulation. Thus, disturbed miRNA activity can be partly explained by circRNA involvement.24

    • Shared DNA methylation signatures in childhood allergy: The MeDALL study

      2021, Journal of Allergy and Clinical Immunology
    View all citing articles on Scopus
    View full text