Skip to main content

Advertisement

Log in

The Genetics of Food Allergy

  • Food Allergy (T Green, Section Editor)
  • Published:
Current Allergy and Asthma Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Food allergy likely arises from a complex interplay between environmental triggers and genetic susceptibility. Here, we review recent studies that have investigated the genetic pathways and mechanisms that may contribute to the pathogenesis of food allergy.

Recent Findings

A heritability component of food allergy has been observed in multiple studies. A number of monogenic diseases characterized by food allergy have elucidated pathways that may be important in pathogenesis. Several population-based genetic variants associated with food allergy have also been identified.

Summary

The genetic mechanisms that play a role in the development of food allergy are heterogeneous and complex. Advances in our understanding of the genetics of food allergy, and how this predisposition interacts with environmental exposures to lead to disease, will improve our understanding of the key pathways leading to food allergy and inform more effective prevention and treatment strategies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Gupta RS, Springston EE, Warrier MR, Smith B, Kumar R, Pongracic J, et al. The prevalence, severity, and distribution of childhood food allergy in the United States. Pediatrics. 2011;128(1):e9–17. https://doi.org/10.1542/peds.2011-0204.

    Article  PubMed  Google Scholar 

  2. Campbell DE, Boyle RJ, Thornton CA, Prescott SL. Mechanisms of allergic disease - environmental and genetic determinants for the development of allergy. Clin Exp Allergy. 2015;45(5):844–58. https://doi.org/10.1111/cea.12531.

    Article  CAS  PubMed  Google Scholar 

  3. Hong X, Tsai HJ, Wang X. Genetics of food allergy. Curr Opin Pediatr. 2009;21(6):770–6. https://doi.org/10.1097/MOP.0b013e32833252dc.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Hong X, Tsai HJ, Liu X, Arguelles L, Kumar R, Wang G, et al. Does genetic regulation of IgE begin in utero? Evidence from T(H)1/T(H)2 gene polymorphisms and cord blood total IgE. J Allergy Clin Immunol. 2010;126(5):1059–67, 67 e1. https://doi.org/10.1016/j.jaci.2010.08.029.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Tan TH, Ellis JA, Saffery R, Allen KJ. The role of genetics and environment in the rise of childhood food allergy. Clin Exp Allergy. 2012;42(1):20–9. https://doi.org/10.1111/j.1365-2222.2011.03823.x.

    Article  CAS  PubMed  Google Scholar 

  6. Neeland MR, Martino DJ, Allen KJ. The role of gene-environment interactions in the development of food allergy. Expert Rev Gastroenterol Hepatol. 2015;9(11):1371–8. https://doi.org/10.1586/17474124.2015.1084873.

    Article  PubMed  Google Scholar 

  7. Sicherer SH, Furlong TJ, Maes HH, Desnick RJ, Sampson HA, Gelb BD. Genetics of peanut allergy: a twin study. J Allergy Clin Immunol. 2000;106(1):53–6. https://doi.org/10.1067/mai.2000.108105.

    Article  CAS  PubMed  Google Scholar 

  8. Liu X, Zhang S, Tsai HJ, Hong X, Wang B, Fang Y, et al. Genetic and environmental contributions to allergen sensitization in a Chinese twin study. Clin Exp Allergy. 2009;39(7):991–8. https://doi.org/10.1111/j.1365-2222.2009.03228.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Koplin JJ, Allen KJ, Gurrin LC, Peters RL, Lowe AJ, Tang ML, et al. The impact of family history of allergy on risk of food allergy: a population-based study of infants. Int J Environ Res Public Health. 2013;10(11):5364–77. https://doi.org/10.3390/ijerph10115364.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Tsai HJ, Kumar R, Pongracic J, Liu X, Story R, Yu Y, et al. Familial aggregation of food allergy and sensitization to food allergens: a family-based study. Clin Exp Allergy. 2009;39(1):101–9. https://doi.org/10.1111/j.1365-2222.2008.03111.x.

    Article  CAS  PubMed  Google Scholar 

  11. •• Gupta RS, Walkner MM, Greenhawt M, Lau CH, Caruso D, Wang X, et al. Food allergy sensitization and presentation in siblings of food allergic children. J Allergy Clin Immunol Pract. 2016;4(5):956–62. https://doi.org/10.1016/j.jaip.2016.04.009. Demonstrated that only a small proportion of siblings of food-allergic patients have clinical reactivity to foods, despite sensititivity on testing, suggesting that such patients should not be evaluated differently than the general population.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Tuano KS, Orange JS, Sullivan K, Cunningham-Rundles C, Bonilla FA, Davis CM. Food allergy in patients with primary immunodeficiency diseases: prevalence within the US Immunodeficiency Network (USIDNET). J Allergy Clin Immunol. 2015;135(1):273–5. https://doi.org/10.1016/j.jaci.2014.09.024.

    Article  PubMed  Google Scholar 

  13. •• Aydin SE, Kilic SS, Aytekin C, Kumar A, Porras O, Kainulainen L, et al. DOCK8 deficiency: clinical and immunological phenotype and treatment options—a review of 136 patients. J Clin Immunol. 2015;35(2):189–98. https://doi.org/10.1007/s10875-014-0126-0. International retrospective survey of the clinical phenotype of DOCK8 deficiency in a large cohort, describing clinical severity of disease, therapeutic measures, and poor prognosis without intervention.

    Article  CAS  PubMed  Google Scholar 

  14. Happel CS, Stone KD, Freeman AF, Shah NN, Wang A, Lyons JJ, et al. Food allergies can persist after myeloablative hematopoietic stem cell transplantation in dedicator of cytokinesis 8-deficient patients. J Allergy Clin Immunol. 2016;137(6):1895–8 e5. https://doi.org/10.1016/j.jaci.2015.11.017.

    Article  PubMed  PubMed Central  Google Scholar 

  15. • Boos AC, Hagl B, Schlesinger A, Halm BE, Ballenberger N, Pinarci M, et al. Atopic dermatitis, STAT3- and DOCK8-hyper-IgE syndromes differ in IgE-based sensitization pattern. Allergy. 2014;69(7):943–53. https://doi.org/10.1111/all.12416. Revealed difference in serum IgE sensitization patterns and Th-cell subset data between hyper-IgE syndromes and atopic dermatitis phenotypes, implicating role of DOCK8 in development of food allergy.

    Article  CAS  PubMed  Google Scholar 

  16. • Tangye SG, Pillay B, Randall KL, Avery DT, Phan TG, Gray P, et al. Dedicator of cytokinesis 8-deficient CD4+ T cells are biased to a TH2 effector fate at the expense of TH1 and TH17 cells. J Allergy Clin Immunol. 2017;139(3):933–49. https://doi.org/10.1016/j.jaci.2016.07.016. DOCK8-deficient memory CD4+ T cells were biased toward a Th2 type, at the expense of Th1 and Th17 cells. Therefore, the TH2 bias is likely to contribute to severe food allergy noted in DOCK8-deficient patients.

    Article  CAS  PubMed  Google Scholar 

  17. • Frischmeyer-Guerrerio PA, Guerrerio AL, Oswald G, Chichester K, Myers L, Halushka MK, et al. TGFbeta receptor mutations impose a strong predisposition for human allergic disease. Sci Transl Med. 2013;5(195):195ra94. https://doi.org/10.1126/scitranslmed.3006448. Demonstrated that patients with mutations in the receptor for TGFβ are more prone to developing a wide spectrum of allergic disease, including food allergy.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Minegishi Y, Saito M, Tsuchiya S, Tsuge I, Takada H, Hara T, et al. Dominant-negative mutations in the DNA-binding domain of STAT3 cause hyper-IgE syndrome. Nature. 2007;448(7157):1058–62. https://doi.org/10.1038/nature06096.

    Article  CAS  PubMed  Google Scholar 

  19. • Siegel AM, Stone KD, Cruse G, Lawrence MG, Olivera A, Jung MY, et al. Diminished allergic disease in patients with STAT3 mutations reveals a role for STAT3 signaling in mast cell degranulation. J Allergy Clin Immunol. 2013;132(6):1388–96. https://doi.org/10.1016/j.jaci.2013.08.045. Inhibition of STAT3 signaling in mast cells was noted to cause impaired FcεRI-mediated proximal and distal signaling, as well as reduced degranulation, suggesting that STAT3 signaling may play a role in mast cell degranulation.

    Article  CAS  PubMed  Google Scholar 

  20. Hox V, O’Connell MP, Lyons JJ, Sackstein P, Dimaggio T, Jones N, et al. Diminution of signal transducer and activator of transcription 3 signaling inhibits vascular permeability and anaphylaxis. J Allergy Clin Immunol. 2016;138(1):187–99. https://doi.org/10.1016/j.jaci.2015.11.024.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Avery DT, Deenick EK, Ma CS, Suryani S, Simpson N, Chew GY, et al. B cell-intrinsic signaling through IL-21 receptor and STAT3 is required for establishing long-lived antibody responses in humans. J Exp Med. 2010;207(1):155–71. https://doi.org/10.1084/jem.20091706.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ma CS, Avery DT, Chan A, Batten M, Bustamante J, Boisson-Dupuis S, et al. Functional STAT3 deficiency compromises the generation of human T follicular helper cells. Blood. 2012;119(17):3997–4008. https://doi.org/10.1182/blood-2011-11-392985.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. •• Zhang Y, Yu X, Ichikawa M, Lyons JJ, Datta S, Lamborn IT, et al. Autosomal recessive phosphoglucomutase 3 (PGM3) mutations link glycosylation defects to atopy, immune deficiency, autoimmunity, and neurocognitive impairment. J Allergy Clin Immunol. 2014;133(5):1400-9–9 e1-5. https://doi.org/10.1016/j.jaci.2014.02.013. PGM3-deficient patients were demonstrated to have an increased percentage of Th17 and Th2 cells while maintaining normal IFN-gamma production, suggesting that altered glycosylation might be important in the pathophysiology of allergic diseases in the general population.

    Article  Google Scholar 

  24. Sassi A, Lazaroski S, Wu G, Haslam SM, Fliegauf M, Mellouli F, et al. Hypomorphic homozygous mutations in phosphoglucomutase 3 (PGM3) impair immunity and increase serum IgE levels. J Allergy Clin Immunol. 2014;133(5):1410-9–9 e1-13. https://doi.org/10.1016/j.jaci.2014.02.025.

    Article  Google Scholar 

  25. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27(1):20–1. https://doi.org/10.1038/83713.

    Article  CAS  PubMed  Google Scholar 

  26. Torgerson TR, Linane A, Moes N, Anover S, Mateo V, Rieux-Laucat F, et al. Severe food allergy as a variant of IPEX syndrome caused by a deletion in a noncoding region of the FOXP3 gene. Gastroenterology. 2007;132(5):1705–17. https://doi.org/10.1053/j.gastro.2007.02.044.

    Article  CAS  PubMed  Google Scholar 

  27. • Lexmond WS, Goettel JA, Lyons JJ, Jacobse J, Deken MM, Lawrence MG, et al. FOXP3+ Tregs require WASP to restrain Th2-mediated food allergy. J Clin Invest. 2016;126(10):4030–44. https://doi.org/10.1172/JCI85129. Study of WASP-deficient mice suggested Treg-specific role for WASP that is required for prevention of Th2 effector cell differentiation and allergic sensitization to dietary allergens.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Lexmond WS, Goettel JA, Sallis BF, McCann K, Rings E, Jensen-Jarolim E, et al. Spontaneous food allergy in Was-/- mice occurs independent of FcepsilonRI-mediated mast cell activation. Allergy. 2017; https://doi.org/10.1111/all.13219.

  29. • Samuelov L, Sarig O, Harmon RM, Rapaport D, Ishida-Yamamoto A, Isakov O, et al. Desmoglein 1 deficiency results in severe dermatitis, multiple allergies and metabolic wasting. Nat Genet. 2013;45(10):1244–8. https://doi.org/10.1038/ng.2739. Desmoglein 1 deficiency was associated with increased expression of a number of genes encoding allergy-promoting cytokines.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chavanas S, Bodemer C, Rochat A, Hamel-Teillac D, Ali M, Irvine AD, et al. Mutations in SPINK5, encoding a serine protease inhibitor, cause Netherton syndrome. Nat Genet. 2000;25(2):141–2. https://doi.org/10.1038/75977.

    Article  CAS  PubMed  Google Scholar 

  31. Lack G. Epidemiologic risks for food allergy. J Allergy Clin Immunol. 2008;121(6):1331–6. https://doi.org/10.1016/j.jaci.2008.04.032.

    Article  CAS  PubMed  Google Scholar 

  32. •• Venkataraman D, Soto-Ramirez N, Kurukulaaratchy RJ, Holloway JW, Karmaus W, Ewart SL, et al. Filaggrin loss-of-function mutations are associated with food allergy in childhood and adolescence. J Allergy Clin Immunol. 2014;134(4):876–82 e4. https://doi.org/10.1016/j.jaci.2014.07.033. First study to associate FLG-LOF with all causes of FA rather than with a specific food allergen. Demonstrated association between FA and FLG-LOF mutations in older children and young adults (10 and 18 years old) but not during earlier years.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. van Ginkel CD, Flokstra-de Blok BM, Kollen BJ, Kukler J, Koppelman GH, Dubois AE. Loss-of-function variants of the filaggrin gene are associated with clinical reactivity to foods. Allergy. 2015;70(4):461–4. https://doi.org/10.1111/all.12569.

    Article  PubMed  Google Scholar 

  34. Asai Y, Greenwood C, Hull PR, Alizadehfar R, Ben-Shoshan M, Brown SJ, et al. Filaggrin gene mutation associations with peanut allergy persist despite variations in peanut allergy diagnostic criteria or asthma status. J Allergy Clin Immunol. 2013;132(1):239–42. https://doi.org/10.1016/j.jaci.2013.03.043.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Brown SJ, Asai Y, Cordell HJ, Campbell LE, Zhao Y, Liao H, et al. Loss-of-function variants in the filaggrin gene are a significant risk factor for peanut allergy. J Allergy Clin Immunol. 2011;127(3):661–7. https://doi.org/10.1016/j.jaci.2011.01.031.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. •• Brough HA, Simpson A, Makinson K, Hankinson J, Brown S, Douiri A, et al. Peanut allergy: effect of environmental peanut exposure in children with filaggrin loss-of-function mutations. J Allergy Clin Immunol. 2014;134(4):867–75 e1. https://doi.org/10.1016/j.jaci.2014.08.011. Reported association between early-life environmental peanut exposure with an increased risk of peanut sensitization and allergy in children with a FLG mutation, supporting hypothesis that peanut allergy may develop following transcutaneous sensitization.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Johansson EK, Bergstrom A, Kull I, Lind T, Soderhall C, van Hage M, et al. IgE sensitization in relation to preschool eczema and filaggrin mutation. J Allergy Clin Immunol. 2017;140(6):1572–1579.e5. https://doi.org/10.1016/j.jaci.2017.04.008.

    Article  CAS  PubMed  Google Scholar 

  38. Tan HT, Ellis JA, Koplin JJ, Matheson MC, Gurrin LC, Lowe AJ, et al. Filaggrin loss-of-function mutations do not predict food allergy over and above the risk of food sensitization among infants. J Allergy Clin Immunol. 2012;130(5):1211–3 e3. https://doi.org/10.1016/j.jaci.2012.07.022.

    Article  CAS  PubMed  Google Scholar 

  39. • Flohr C, Perkin M, Logan K, Marrs T, Radulovic S, Campbell LE, et al. Atopic dermatitis and disease severity are the main risk factors for food sensitization in exclusively breastfed infants. J Invest Dermatol. 2014;134(2):345–50. https://doi.org/10.1038/jid.2013.298. Large prospective study examining breastfed infants found that those with severe atopic dermatitis are significantly more likely to be sensitized to food, independent of FLG mutation carriage. Clinical reactivity not confirmed.

    Article  CAS  PubMed  Google Scholar 

  40. Thyssen JP, Tang L, Husemoen LL, Stender S, Szecsi PB, Menne T, et al. Filaggrin gene mutations are not associated with food and aeroallergen sensitization without concomitant atopic dermatitis in adults. J Allergy Clin Immunol. 2015;135(5):1375–8 e1. https://doi.org/10.1016/j.jaci.2015.01.001.

    Article  CAS  PubMed  Google Scholar 

  41. Savilahti EM, Ilonen J, Kiviniemi M, Saarinen KM, Vaarala O, Savilahti E. Human leukocyte antigen (DR1)-DQB1*0501 and (DR15)-DQB1*0602 haplotypes are associated with humoral responses to early food allergens in children. Int Arch Allergy Immunol. 2010;152(2):169–77. https://doi.org/10.1159/000265538.

    Article  CAS  PubMed  Google Scholar 

  42. Martino DJ, Ashley S, Koplin J, Ellis J, Saffery R, Dharmage SC, et al. Genomewide association study of peanut allergy reproduces association with amino acid polymorphisms in HLA-DRB1. Clin Exp Allergy. 2017;47(2):217–23. https://doi.org/10.1111/cea.12863.

    Article  CAS  PubMed  Google Scholar 

  43. Kusunoki T, Okafuji I, Yoshioka T, Saito M, Nishikomori R, Heike T, et al. SPINK5 polymorphism is associated with disease severity and food allergy in children with atopic dermatitis. J Allergy Clin Immunol. 2005;115(3):636–8. https://doi.org/10.1016/j.jaci.2004.12.1114.

    Article  CAS  PubMed  Google Scholar 

  44. Hemler JA, Phillips EJ, Mallal SA, Kendall PL. The evolving story of human leukocyte antigen and the immunogenetics of peanut allergy. Ann Allergy Asthma Immunol. 2015;115(6):471–6. https://doi.org/10.1016/j.anai.2015.10.008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Li J, Maggadottir SM, Hakonarson H. Are genetic tests informative in predicting food allergy? Curr Opin Allergy Clin Immunol. 2016;16(3):257–64. https://doi.org/10.1097/ACI.0000000000000268.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. •• Hirota T, Nakayama T, Sato S, Yanagida N, Matsui T, Sugiura S, et al. Association study of childhood food allergy with genome-wide association studies-discovered loci of atopic dermatitis and eosinophilic esophagitis. J Allergy Clin Immunol. 2017;140(6):1713–6. https://doi.org/10.1016/j.jaci.2017.05.034. Identified significant associations between childhood food allergy and 14 GWAS-discovered loci of AD and EoE. FLG null variants also appear to influence the susceptibility to childhood food allergy.

    Article  PubMed  Google Scholar 

  47. •• Hong X, Hao K, Ladd-Acosta C, Hansen KD, Tsai HJ, Liu X, et al. Genome-wide association study identifies peanut allergy-specific loci and evidence of epigenetic mediation in US children. Nat Commun. 2015;6:6304. https://doi.org/10.1038/ncomms7304. First genome-wide association study of well-defined food allergy in a large cohort. Peanut allergy-specific loci identified in the HLA-DR and -DQ regions, which are associated with differential DNA methylation patterns

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Li J, Fung I, Glessner JT, Pandey R, Wei Z, Bakay M, et al. Copy number variations in CTNNA3 and RBFOX1 associate with pediatric food allergy. J Immunol. 2015;195(4):1599–607. https://doi.org/10.4049/jimmunol.1402310.

    Article  CAS  PubMed  Google Scholar 

  49. Watson CT, Cohain AT, Griffin RS, Chun Y, Grishin A, Hacyznska H, et al. Integrative transcriptomic analysis reveals key drives of acute peanut allergic reactions. Nat Commun. 2017;8(1):1–13. https://doi.org/10.1038/s41467-017-02188-7.

    Article  Google Scholar 

Download references

Acknowledgements

This research was supported by the Intramural Research Program of the National Institute of Allergy and Infectious Diseases, NIH.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Pamela A. Frischmeyer-Guerrerio.

Ethics declarations

Conflicts of Interest

The authors declare no conflicts of interest relevant to this manuscript.

Human and Animal Rights

All reported studies/experiments with human or animal subjects performed by the authors have been previously published and complied with all applicable ethical standards (including the Helsinki declaration and its amendments, institutional/national research committee standards, and international/national/institutional guidelines).

Additional information

This article is part of the Topical Collection on Food Allergy

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Carter, C.A., Frischmeyer-Guerrerio, P.A. The Genetics of Food Allergy. Curr Allergy Asthma Rep 18, 2 (2018). https://doi.org/10.1007/s11882-018-0756-z

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11882-018-0756-z

Keywords

Navigation