Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-26T03:26:29.169Z Has data issue: false hasContentIssue false

Allergic disease: understanding how in utero events set the scene

Published online by Cambridge University Press:  29 June 2010

Susan L. Prescott*
Affiliation:
School of Paediatrics and Child Health Research, University of Western Australia, PO Box D184, Princess Margaret Hospital, Perth, WA6001, Australia
*
Corresponding author: Professor Susan L. Prescott, fax +61 8 9388 2097, email sprescott@meddent.uwa.edu.au
Rights & Permissions [Opens in a new window]

Abstract

Events and exposures in pregnancy can have critical effects on fetal development with lasting implications for subsequent health and disease susceptibility. There is growing interest in how modern environmental changes influence fetal immune development and contribute to the recent epidemic of allergy and other immune disorders. Rising rates of allergic disease in early infancy, together with pre-symptomatic differences in immune function at birth, suggest that antenatal events play a predisposing role in the development of disease. A number of environmental exposures in pregnancy can modify neonatal immune function including diet, microbial exposure and maternal smoking, and there is emerging evidence from animal models that these factors may have epigenetic effects on immune gene expression and disease susceptibility. Furthermore, functional genetic polymorphisms also alter individual vulnerability to the effects of these environmental exposures, highlighting the complexity of gene–environmental interactions in this period. All these observations underscore the need for ongoing research to understand the pathogenesis and rising incidence of disease in the hope of better strategies to reverse this.

Type
3rd International Immunonutrition Workshop
Copyright
Copyright © The Author 2010

Abbreviations:
Th1

T-helper cell type 1

Th2

T-helper cell type 2

Treg

regulatory T-cells

While the spotlight is on the early postnatal period for understanding the events that lead to oral tolerance, it is becoming clearer that the scene is set much earlier in development. Now extensive data from both epidemiological and experimental studies indicate that gene–environmental interactions during pregnancy can induce permanent changes in physiological processes and disease susceptibility(Reference Barker1) by altering gene expression and disease predisposition through epigenetic mechanisms(Reference Waterland and Michels2). This has been the foundation of the newly established field of Developmental Origins of Health and Disease(Reference Waterland and Michels2). Although this has been best studied in the context of cardiovascular and metabolic disease, the epidemic rise in both allergic and autoimmune diseases also highlights the susceptibility of immune pathways to modern environmental changes. Moreover, the rising rate of disease in early infancy is further evidence that early events must have a critical role. A recent animal model has provided the first evidence that alterations in the maternal diet in pregnancy can alter the risk of allergic airways disease in the offspring through epigenetic changes in gene expression(Reference Hollingsworth, Maruoka and Boon3). The mother provides the first environment for the developing fetus, and this review explores the range of maternal factors that may influence fetal immune development including both exogenous environmental exposures and endogenous factors.

Immune development and regulation in pregnancy

Human lymphocytes derived from the yolk sac appear in the liver within several weeks of conception. By 10–12 weeks of gestation, they are evident in the thymus(Reference Stites and Pavia4) and show responsiveness to mitogen stimulation(Reference Pegrum5) and allogeneic graft v. host reactivity(Reference Asantila, Sorvani and Hirvonen6). Thymocytes appear to be capable of binding antigens from 20 to 22 weeks gestation, and allergen-specific responses have been also recorded as early as 22 weeks gestation(Reference Jones, Miles and Warner7). However, while many groups have demonstrated that cord blood mononuclear cells can respond to environmental allergens, there has been ongoing debate about whether these reflect conventional memory responses as they do not correlate well with either maternal exposure or subsequent development of allergic disease (reviewed in Holt(Reference Holt8)). There is some evidence that these responses reflect a default response by recent thymic emigrants to first antigen encounter, which also leads to the activation of regulatory T-cells (Treg)(Reference Thornton, Upham and Wikstrom9). At birth, cytokine production is dominated by T-helper cell type 2 (Th2) cytokines(Reference Prescott, Macaubas and Holt10) and many aspects of neonatal immune function are still immature including antigen presenting cells, T-helper cell type 1 (Th1) and pro-inflammatory Th17 effector T-cells and Treg function(Reference Schaub, Liu and Schleich11). It has been proposed that postnatal microbial exposure provides an essential source of immune stimulation for all of these pathways and protection from allergic diseases(Reference Schaub, Liu and Schleich11). There is therefore intense interest in factors which influence the patterns of perinatal immune function and their subsequent regulation.

Immune development is under epigenetic regulation

Epigenetic regulation is fundamental to cellular differentiation and all aspects of normal development. Specifically, changes in the methylation of DNA and histones, and histone acetylation regulate gene expression by altering the DNA compaction and accessibility for gene transcription(Reference Song, Mahmood and Ghosh12). There is clear evidence that T-cell differentiation is under epigenetic control(Reference Janson, Winerdal and Winqvist13), including Th1 and Th2 differentiation(Reference Fields, Kim and Flavell14Reference Santangelo, Cousins and Winkelmann18), FoxP3 expression and Treg differentiation(Reference Janson, Winerdal and Marits19, Reference Polansky, Kretschmer and Freyer20) and Th17 differentiation(Reference Koenen, Smeets and Vink21).

The main epigenetic mechanism controlling Th1 expression is methylation of the interferon-γ gene promoter. This is hypermethylated (i.e. underexpressed) in neonatal CD4+ T-cells and shows progressive demethylation by adulthood(Reference White, Watt and Holt22). Changes in methylation (demethylation) are also prerequisite for FOXP3 expression and Treg differentiation(Reference Janson, Winerdal and Marits19, Reference Polansky, Kretschmer and Freyer20). Another major mechanism of epigenetic regulation is histone acetylation. Removal of acetyl groups by histone deacetylase generally leads to gene silencing, whereas acetylation by histone acetyl transferase opens chromatin structure for enhanced gene transcription(Reference Bhavsar, Ahmad and Adcock23). Exposures that inhibit histone deacetylase such as oxidative stress up-regulate Th2 cytokine (IL-13 and IL-5) and GATA3-mediated T-cell responses(Reference Bhavsar, Ahmad and Adcock23, Reference Su, Becker and Kozyrskyj24). The Th17 lineage also appears to be regulated through similar epigenetic mechanisms(Reference Janson, Winerdal and Winqvist13, Reference Koenen, Smeets and Vink21).

These insights have logically led to interest in factors which may promote allergic propensity by increased histone acetylation (Th2 promotion) and/or increased gene methylation (Th1 and Treg silencing)(Reference Bousquet, Jacot and Yssel25, Reference Miller and Ho26). As discussed further later, the first evidence of this comes from an animal model in which maternal folate supplementation (a dietary methyl donor) resulted in hypermethylation (suppression) of regulatory genes and the development of allergic disease in the offspring(Reference Hollingsworth, Maruoka and Boon3). At this stage, the implications in human subjects are not clear, but this provides a platform for investigating epigenetic pathways as a mechanism for gene–environmental interactions in allergic disease.

Altered patterns of immune response begin to emerge in fetal life

There have been numerous studies showing pre-symptomatic differences in the immune responses of newborns who later develop allergic disease (reviewed in Prescott and Clifton(Reference Prescott and Clifton27) and Prescott(Reference Prescott28)). This was initially thought to largely reflect inherited genetic risk. However, the epidemic rise in allergic disease has raised the alternative hypothesis that this may be due to more complex alteration in immune gene expression conferred by gene–environmental interactions in utero. Thus, at least some of the environmental effects driving the rise in allergic disease may begin in utero, and the differences in neonatal immune function may be the first signs of this increasing allergic predisposition.

A relative immaturity of neonatal Th1 immune function has been one of the clearest and most replicated neonatal associations with allergic disease(Reference Rinas, Horneff and Wahn29Reference Warner, Miles and Jones31). Although Th1 responses are generally suppressed at birth under the Th2-dominant influence of pregnancy, this appears to be more marked in neonates with allergic predisposition or subsequent allergic disease(Reference Rinas, Horneff and Wahn29Reference Warner, Miles and Jones31). Other aspects of neonatal effector T-cell function may be impaired in this population(Reference Prescott, Macaubas and Holt10). More recently, there has been emerging evidence that allergic disease is also associated with attenuated neonatal Treg function(Reference Smith, Tourigney and Noakes32, Reference Schaub, Liu and Hoppler33) and differences in innate immunity(Reference Prescott, Noakes and Chow34Reference Amoudruz, Holmlund and Malmstrom37). A number of other neonatal markers have been identified in relation to allergic disease (reviewed in Prescott and Clifton(Reference Prescott and Clifton27)), though none of these has so far been shown to be of accurate predictive value. Further research is needed to understand the functional significance and the possible contribution to the disease pathogenesis. It is possible that that impaired Th1 and Treg function may contribute to a reduced capacity to suppress Th2 responses in the early postnatal period; however, this is likely to be oversimplistic. While there has been long-standing speculation that dysfunction of antigen presenting cells and innate immunity may contribute to the apparent immaturity of Th1 activity, there is still only indirect evidence to support this(Reference Upham, Holt and Taylor38, Reference Gabrielsson, Soderlund and Nilsson39). Furthermore, while some groups have shown that markers of innate activity (such as Toll-like receptor function or expression) are lower in neonates at risk of allergic disease(Reference Schaub, Campo and He35, Reference Amoudruz, Holmlund and Malmstrom37), we have shown the opposite(Reference Prescott, Noakes and Chow34). While this needs to be examined further, collectively these observations do suggest that differences in neonatal immune function confer increased susceptibility to subsequent postnatal environmental influences and contribute to an evolving allergic phenotype.

Evidence that maternal environmental factors can modify fetal immune development

While there is a hereditary component of allergy, only environmental change can account for the rapid rise in disease. There is growing evidence that maternal environmental exposures including dietary factors(Reference Dunstan, Mori and Barden40, Reference Devereux, Barker and Seaton41), cigarette smoke(Reference Noakes, Holt and Prescott42, Reference Noakes, Hale and Thomas43) and microbial exposure(Reference Ege, Bieli and Frei36, Reference Matsuoka, Matsubara and Katayama44) can modify neonatal immune responses.

Maternal dietary influences on immune development

Maternal nutrition is critically important for most aspects of fetal development, including the immune system. Complex dietary changes with progressive industrialisation have been implicated with the rise of allergic disease. As with other exposures, nutritional changes are likely to have more profound effects on pregnancy when the organ systems and physiological responses are developing. Many dietary nutrients have recognised immunomodulatory properties and plausible biological mechanisms of influence(Reference Shaheen45). This includes PUFA(Reference Calder46), antioxidants and other vitamins(Reference Devereux47). Of these, PUFA are among the most extensively studied in this context. A declining intake of anti-inflammatory n-3 PUFA (found in oily fish) has been implicated in the rise in allergic disease(Reference Black and Sharpe48), and a series of studies have shown a protective relationship between maternal n-3 PUFA consumption in pregnancy and subsequent infant allergic disease(Reference Salam, Li and Langholz49Reference Sausenthaler, Koletzko and Schaaf54), though not all were significant after allowing multiple comparisons(Reference Newson, Shaheen and Henderson55). Several intervention studies using fish-oil supplementation in pregnancy have also suggested protective effects against allergic disease in early childhood(Reference Dunstan, Mori and Barden40, Reference Furuhjelm, Warstedt and Larsson56) and a long-term (16-year) follow-up study showed a reduction in subsequent asthma(Reference Olsen, Osterdal and Salvig57). A number of other studies are currently in progress to hopefully assess this more definitively.

In the last 12 months, dietary folate has become one of the most topical dietary nutrients in this area. As a dietary methyl donor, there has been established interest in the broader context of epigenetics; however, the recent pro-allergic effects demonstrated in an animal model have extended this interest into the field of allergic disease, and for the first time provided an epigenetic model for these immune disorders. As indicated earlier, supplementation with folate in pregnancy induced hypermethylation (silencing) of regulatory genes in lung tissue, and was associated with the development of allergic airway disease and systemic allergic responses(Reference Hollingsworth, Maruoka and Boon3). This effect was also transmitted epigenetically to subsequent generations. This has been followed by reports in human subjects(Reference Haberg, London and Stigum58, Reference Whitrow, Moore and Rumbold59) linking folic acid supplementation during pregnancy with increased risk of asthma and respiratory disease in infants. There have been links between folate status in the postnatal period and allergic disease, but if anything, folate was protective(Reference Matsui and Matsui60). At this stage, the significance of these findings is not yet clear. At this stage, it is premature to change current practice based on animal studies and preliminary epidemiological reports, but there is now an urgent call for further studies to address this now pressing question(Reference Miller61).

Most other nutrients linked with allergic disease in epidemiological studies have not been investigated in intervention studies. Maternal antioxidant vitamins have been associated with differences in neonatal immune function(Reference Devereux, Barker and Seaton41), as well as reduced risk of possible allergic outcomes including recurrent wheezing (vitamin C(Reference Farchi, Forastiere and Agabiti62, Reference Forastiere, Pistelli and Sestini63), vitamin D(Reference Devereux, Litonjua and Turner64), vitamin E(Reference Devereux, Turner and Craig65, Reference Litonjua, Rifas-Shiman and Ly66) and Zn(Reference Litonjua, Rifas-Shiman and Ly66)), and eczema (vitamin E(Reference Martindale, McNeill and Devereux67)). However, these correlative findings are not consistent between studies and concerns have been raised about the potential for paradoxical or adverse effects of antioxidants on allergic disease(Reference Murr, Schroecksnadel and Winkler68). This is based on speculation that antioxidant supplementation could promote Th2 differentiation by inhibiting oxidative stress(Reference Murr, Schroecksnadel and Winkler68). At this stage, there is no place for specific dietary supplementation in pregnancy for allergy prevention, although this may be included in future strategies once these relationships are more fully understood.

Maternal microbial exposure

The decline in the level and diversity of microbial exposure is a leading candidate in the allergy epidemic. While focus has been on the role of postnatal microbial exposure, animal studies clearly demonstrate that in utero (maternal) exposure to both pathogenic(Reference Blumer, Herz and Wegmann69) and non-pathogenic microbial products(Reference Blumer, Sel and Virna70) can inhibit the development of allergic phenomena in the offspring. In human subjects, maternal exposure to high microbial burden in German farming environments has been associated with altered expression of innate immune genes and reduced risk of allergic disease in the children(Reference Ege, Bieli and Frei36). Similar protective effects of farming environments have also been observed in New Zealand(Reference Douwes, Cheng and Travier71). This effect was independent of postnatal exposure in both studies(Reference Ege, Bieli and Frei36, Reference Douwes, Cheng and Travier71). In what may be another example of epigenetic regulation, there is preliminary evidence that an apathogenic microbial strain isolated from the German farming environment can mediate allergy-protective effects by epigenetic changes(Reference Teich, Conrad and Ferstl72). Intranasal administration of this strain (Acinetobacter lwoffii) to pregnant mice was associated with significant effects on the ontogeny of splenic CD4+ Th1 interferon-γ production in the offspring of exposed mothers. These differences were directly related to epigenetic changes in the interferon-γ promoter(Reference Teich, Conrad and Ferstl72). This supports notions that microbial exposure may modify foetal gene expression and provides a potential epigenetic mechanism. Intervention studies using microbial products in human pregnancy are mainly limited to probiotics(Reference Johannsen and Prescott73). Although there is some evidence that these products may reduce the risk of eczema(Reference Osborn and Sinn74), there is wide heterogeneity in study protocols and findings between studies. This appears to be species-dependent(Reference Wickens, Black and Stanley75) and although there has been speculation that antenatal supplementation may explain the beneficial effects in some studies(Reference Abrahamsson, Jakobsson and Bottcher76), this is also not consistent(Reference Johannsen and Prescott73). Furthermore, while one study suggested that probiotic bacteria during the final weeks of pregnancy was associated with an increase in cytokine (interferon-γ) detection in cord blood(Reference Prescott, Wickens and Westcott77), another more comprehensive investigation found no effects on any aspect of neonatal immune function(Reference Boyle, Mah and Chen78). At this stage, the role of probiotics in the prevention of allergic disease is still unclear and no specific recommendation can be made.

Maternal allergen exposure

Although early allergy prevention strategies focused on allergen avoidance, there is little clear evidence that changes in food or inhalant allergen exposure in pregnancy are responsible for the rise in allergic disease. Moreover, there is no clear evidence that restrictive dietary recommendations actually prevent allergic disease. In contrast, there are a growing number of reports that an attempt to avoid or delay allergen exposure may actually increase the risk of allergic sensitisation(Reference Woodcock, Lowe and Murray79, Reference Snijders, Thijs and van Ree80). Many international expert bodies have independently concluded that there is insufficient evidence to justify the continued use of these allergen-restrictive diets in either pregnancy or early infancy(Reference Greer, Sicherer and Burks81Reference Agostoni, Decsi and Fewtrell83).

Maternal smoking

Maternal cigarette smoking in pregnancy has many adverse effects on the fetus, including effects on lung function and asthma risk. While there are documented effects on neonatal immune function(Reference Devereux, Barker and Seaton41Reference Noakes, Hale and Thomas43), the relationship with other allergic sensitisation has been less clear. Regardless of this, the avoidance of cigarette smoke is an unequivocal recommendation in view of the many toxic effects on the fetus.

Other maternal exposures

A range of other maternal exposures could potentially influence fetal immune development. Firstly, the use of a number of medications in pregnancy has been associated with an increased risk of childhood asthma. The most consistent relationship has been seen with paracetamol, with a series of independent studies(Reference Rebordosa, Kogevinas and Sorensen84Reference Persky, Piorkowski and Hernandez87) supporting the initial reports(Reference Shaheen, Newson and Sherriff88). Documented depletion of antioxidant glutathione has been proposed as a mechanism of effect on immune function and lung development. Another notable relationship has been a recent large-scale study showing that acid-suppressive medications in pregnancy are associated with an increased risk of developing childhood allergy(Reference Dehlink, Yen and Leichtner89). Although the mechanisms are unclear, it does follow animal studies showing similar effects.

Secondly, modern environmental changes have been associated with the appearance of many new chemicals and persistent organic pollutants. A number of these products of industry and agriculture have been linked with immune disorders(Reference Hertz-Picciotto, Park and Dostal90) because of oestrogenic (proTh2) properties, which have earned them the title of ‘hormone imposters’. Although environmental measures have helped reduce the level of exposure, we have detected pesticides in 94% of maternal abdominal fat samples (collected at caesarean section) and 62% breast milk samples, albeit at very low levels(Reference Noakes, Taylor and Wilkinson91). The effects of these and other modern environmental exposure on immune development is still unclear and difficult to investigate, but should not be ignored as potential contributors to the rise in modern diseases.

Endogenous influences in pregnancy

In addition to the external environment, endogenous factors may also influence development during pregnancy. Our recent studies suggest that maternal allergic status is associated with a relative reduction of Th1 responses to both environmental allergens(Reference Breckler, Hale and Jung92) and fetal alloantigens(Reference Breckler, Hale and Taylor93), and modified expression of cytokine genes in the placenta(Reference Scott, Hodyl and Murphy94). We propose that this could influence immune development by modifying the cytokine milieu experienced by the fetus. Direct maternal effects are also in keeping with the observation that maternal is a greater risk factor than paternal allergy.

The placental immune system is also partially regulated by glucocorticoids, and there is evidence that activation of the hypothalamic–pituitary–adrenal axis is also associated with up-regulation of placental Th1 cytokines and poor fetal outcomes. Animal studies show that other early stressors (exposure to endotoxin) have long-lived effects on both hypothalamic–pituitary–adrenal function and immune function in the offspring(Reference Shanks, Windle and Perks95). It is certain that the effects of physical and psychological stress in pregnancy on immune development need to be investigated further.

Variations in genetic predisposition add a further dimension of complexity

All of these interactions need to be viewed in the context of genetic predisposition. Functional polymorphisms confer variations in susceptibility to both disease and the effects of environmental exposures. For example, in the context of high bacterial exposure, polymorphisms in microbial recognition pathways (Toll-like receptor 2) confer protection from allergic disease, but this relationship is not seen in a low microbial burden environment(Reference Eder, Klimecki and Yu96). Thus, the effects of genetic polymorphisms may only be relevant in certain environments. These complex interactions could obscure potentially important causal pathways and could account for the many inconsistencies between studies. There are now recognised functional genetic polymorphisms in many other pathways, which could modify the biological effects of other environmental exposures including PUFA(Reference Rzehak, Heinrich and Klopp97), folate(Reference Husemoen, Toft and Fenger98) and cigarette smoke(Reference Kabesch, Hoefler and Carr99). This has highlighted the need for new research approaches to further explore these complexities.

Summary and conclusions

With the advent of the Developmental Origins of Health and Disease hypothesis(Reference Barker1), pregnancy is now widely recognised as a critical time for developmental programming, when the scene is set for future patterns of health and disease. New technologies and the discovery of epigenetic regulation has provided mechanisms for how environmental exposures can alter gene expression and influence the evolving phenotype(Reference Waterland and Michels2). Extensive environmental changes have been implicated in the epidemic rise of allergy and other immune disorders, and there is now emerging evidence of how environmental factors may modify fetal immune development. A deeper understanding of these pathways will hopefully reveal both the pathogenesis of these diseases and the reasons for the rise in prevalence. This in turn may lead to more effective strategies for disease prevention. Complex multi-factorial genetic and environmental interactions may ultimately translate to individualised early interventions tailored and targeted according to genetic predisposition. Although future developments are difficult to predict in this rapidly evolving field, events in pregnancy should remain a research priority.

Acknowledgements

S. L. P. has been a speaker at meetings sponsored by SHS/Nutricia and Nestlé. She has been a member of the independent Scientific Advisory Board of Nestlé Nutrition Institute Oceania, an expert panel on Cows Milk Allergy for Nutricia Australia and expert panels for Mead Johnson and Fonterra. She has received travel assistance and speaker fees from these companies to present at or attend scientific meetings.

References

1.Barker, DJ (1998) In utero programming of chronic disease. Clin Sci (Colch) 95, 115128.CrossRefGoogle ScholarPubMed
2.Waterland, RA & Michels, KB (2007) Epigenetic epidemiology of the developmental origins hypothesis. Annu Rev Nutr 27, 363388.Google Scholar
3.Hollingsworth, JW, Maruoka, S, Boon, K et al. (2008) In utero supplementation with methyl donors enhances allergic airway disease in mice. J Clin Invest 118, 34623469.Google Scholar
4.Stites, D & Pavia, C (1979) Ontogeny of human T cells. Pediatrics 64, Suppl., 795802.CrossRefGoogle ScholarPubMed
5.Pegrum, G (1971) Mixed cultures of human fetal and adult cells. Immunology 2, 159167.Google Scholar
6.Asantila, T, Sorvani, T, Hirvonen, T et al. (1973) Xenogeneic reactivity of human fetal lymphocytes. J Immunol 111, 984987.CrossRefGoogle ScholarPubMed
7.Jones, A, Miles, E, Warner, J et al. (1996) Fetal peripheral blood mononuclear cell proliferative responses to mitogenic and allergenic stimuli during gestation. Pediatric Allergy Immunol 7, 109116.CrossRefGoogle ScholarPubMed
8.Holt, PG (2008) Prenatal versus postnatal priming of allergen specific immunologic memory: the debate continues. J Allergy Clin Immunol 122, 717718.CrossRefGoogle ScholarPubMed
9.Thornton, CA, Upham, JW, Wikstrom, ME et al. (2004) Functional maturation of CD4+CD25+CTLA4+CD45RA+ T regulatory cells in human neonatal T cell responses to environmental antigens/allergens. J Immunol 173, 30843092.CrossRefGoogle ScholarPubMed
10.Prescott, S, Macaubas, C, Holt, B et al. (1998) Transplacental priming of the human immune system to environmental allergens: universal skewing of initial T-cell responses towards Th-2 cytokine profile. J Immunol 160, 47304737.CrossRefGoogle Scholar
11.Schaub, B, Liu, J, Schleich, I et al. (2008) Impairment of T helper and T regulatory cell responses at birth. Allergy 63, 14381447.CrossRefGoogle Scholar
12.Song, F, Mahmood, S, Ghosh, S et al. (2009) Tissue specific differentially methylated regions (TDMR): changes in DNA methylation during development. Genomics 93, 130139.CrossRefGoogle ScholarPubMed
13.Janson, PC, Winerdal, ME & Winqvist, O (2009) At the crossroads of T helper lineage commitment – epigenetics points the way. Biochim Biophys Acta 1790(9), 906919.CrossRefGoogle ScholarPubMed
14.Fields, PE, Kim, ST & Flavell, RA (2002) Cutting edge: changes in histone acetylation at the IL-4 and IFN-gamma loci accompany Th1/Th2 differentiation. J Immunol 169, 647650.CrossRefGoogle ScholarPubMed
15.Lee, GR, Kim, ST, Spilianakis, CG et al. (2006) T helper cell differentiation: regulation by cis elements and epigenetics. Immunity 24, 369379.CrossRefGoogle ScholarPubMed
16.Lee, DU, Agarwal, S & Rao, A (2002) Th2 lineage commitment and efficient IL-4 production involves extended demethylation of the IL-4 gene. Immunity 16, 649660.Google Scholar
17.Shin, HJ, Park, HY, Jeong, SJ et al. (2005) STAT4 expression in human T cells is regulated by DNA methylation but not by promoter polymorphism. J Immunol 175, 71437150.CrossRefGoogle Scholar
18.Santangelo, S, Cousins, DJ, Winkelmann, NE et al. (2002) DNA methylation changes at human Th2 cytokine genes coincide with DNase I hypersensitive site formation during CD4(+) T cell differentiation. J Immunol 169, 18931903.Google Scholar
19.Janson, PC, Winerdal, ME, Marits, P et al. (2008) FOXP3 promoter demethylation reveals the committed Treg population in humans. PLoS ONE 3, e1612.CrossRefGoogle ScholarPubMed
20.Polansky, JK, Kretschmer, K, Freyer, J et al. (2008) DNA methylation controls Foxp3 gene expression. Eur J Immunol 38, 16541663.CrossRefGoogle ScholarPubMed
21.Koenen, HJ, Smeets, RL, Vink, PM et al. (2008) Human CD25highFoxp3pos regulatory T cells differentiate into IL-17-producing cells. Blood 112, 23402352.Google Scholar
22.White, GP, Watt, PM, Holt, BJ et al. (2002) Differential patterns of methylation of the IFN-gamma promoter at CpG and non-CpG sites underlie differences in IFN-gamma gene expression between human neonatal and adult CD45RO- T cells. J Immunol 168, 28202827.CrossRefGoogle ScholarPubMed
23.Bhavsar, P, Ahmad, T & Adcock, IM (2008) The role of histone deacetylases in asthma and allergic diseases. J Allergy Clin Immunol 121, 580584.CrossRefGoogle ScholarPubMed
24.Su, R, Becker, A, Kozyrskyj, A et al. (2008) Epigenetic regulation of established human type 1 versus type 2 cytokine responses. J Allergy Clin Immunol 121, 5763.Google Scholar
25.Bousquet, J, Jacot, W, Yssel, H et al. (2004) Epigenetic inheritance of fetal genes in allergic asthma. Allergy 59, 138147.Google Scholar
26.Miller, RL & Ho, SM (2008) Environmental epigenetics and asthma: current concepts and call for studies. Am J Respir Crit Care Med 177, 567573.Google Scholar
27.Prescott, SL & Clifton, VL (2009) Asthma and pregnancy: emerging evidence of epigenetic interactions in utero. Curr Opin Allergy Clin Immunol 9, 417426.CrossRefGoogle ScholarPubMed
28.Prescott, SL (2003) Early origins of allergic disease: a review of processes and influences during early immune development. Curr Opin Allergy Clin Immunol 3, 125132.CrossRefGoogle ScholarPubMed
29.Rinas, U, Horneff, G & Wahn, V (1993) Interferon gamma production by cord blood mononuclear cells is reduced in newborns with a family history of atopic disease and is independant from cord blood IgE levels. Pediatr Allergy Immunol 4, 6064.CrossRefGoogle Scholar
30.Tang, MLK, Kemp, AS, Thorburn, J et al. (1994) Reduced interferon gamma secretion in neonates and subsequent atopy. Lancet 344, 983985.Google Scholar
31.Warner, JA, Miles, EA, Jones, AC et al. (1994) Is deficiency of interferon gamma production by allergen triggered cord blood cells a predictor of atopic eczema? Clin Exp Allergy 24, 423430.CrossRefGoogle Scholar
32.Smith, M, Tourigney, MR, Noakes, P et al. (2008) Egg allergic children have evidence of reduced neonatal CD4+CD25+CD127 lo/− T regulatory cell function. J Allergy Clin Immunol 121, 14601466.Google Scholar
33.Schaub, B, Liu, J, Hoppler, S et al. (2008) Impairment of T-regulatory cells in cord blood of atopic mothers. J Allergy Clin Immunol 121, 14911499, 9 e1–13.CrossRefGoogle ScholarPubMed
34.Prescott, SL, Noakes, P, Chow, B et al. (2008) Presymptomatic differences in toll-like receptor function in infants who develop allergy. J Allergy Clin Immunol 122, 391399.Google Scholar
35.Schaub, B, Campo, M, He, H et al. (2006) Neonatal immune responses to TLR2 stimulation: influence of maternal atopy on Foxp3 and IL-10 expression. Respir Res 7, 40.CrossRefGoogle ScholarPubMed
36.Ege, MJ, Bieli, C, Frei, R et al. (2006) Prenatal farm exposure is related to the expression of receptors of the innate immunity and to atopic sensitization in school-age children. J Allergy Clin Immunol 117, 817823.CrossRefGoogle Scholar
37.Amoudruz, P, Holmlund, U, Malmstrom, V et al. (2005) Neonatal immune responses to microbial stimuli: is there an influence of maternal allergy? J Allergy Clin Immunol 115, 13041310.CrossRefGoogle ScholarPubMed
38.Upham, JW, Holt, PG, Taylor, A et al. (2004) HLA-DR expression on neonatal monocytes is associated with allergen-specific immune responses. J Allergy Clin Immunol 114, 12021208.CrossRefGoogle ScholarPubMed
39.Gabrielsson, S, Soderlund, A, Nilsson, C et al. (2001) Influence of atopic heredity on IL-4-, IL-12- and IFN-gamma-producing cells in in vitro activated cord blood mononuclear cells. Clin Exp Immunol 126, 390396.CrossRefGoogle ScholarPubMed
40.Dunstan, J, Mori, TA, Barden, A et al. (2003) Fish oil supplementation in pregnancy modifies neonatal allergen-specific immune responses and clinical outcomes in infants at high risk of atopy: a randomised controlled trial. J Allergy Clin Immunol 112, 11781184.CrossRefGoogle Scholar
41.Devereux, G, Barker, RN & Seaton, A (2002) Antenatal determinants of neonatal immune responses to allergens. Clin Exp Allergy 32, 4350.CrossRefGoogle ScholarPubMed
42.Noakes, PS, Holt, PG & Prescott, SL (2003) Maternal smoking in pregnancy alters neonatal cytokine responses. Allergy 58, 10531058.CrossRefGoogle ScholarPubMed
43.Noakes, PS, Hale, J, Thomas, R et al. (2006) Maternal smoking is associated with impaired neonatal toll-like-receptor-mediated immune responses. Eur Respir J 28, 721729.CrossRefGoogle ScholarPubMed
44.Matsuoka, T, Matsubara, T, Katayama, K et al. (2001) Increase of cord blood cytokine-producing T cells in intrauterine infection. Pediatr Int 43, 453457.CrossRefGoogle ScholarPubMed
45.Shaheen, SO (2008) Prenatal nutrition and asthma: hope or hype? Thorax 63, 483485.Google Scholar
46.Calder, PC (2008) The relationship between the fatty acid composition of immune cells and their function. Prostaglandins Leukot Essent Fatty Acids 79, 101108.Google Scholar
47.Devereux, G (2006) The increase in the prevalence of asthma and allergy: food for thought. Nat Rev Immunol 6, 869874.CrossRefGoogle ScholarPubMed
48.Black, P & Sharpe, S (1997) Dietary fat and asthma: is there a connection? Eur Respir J 10, 6–12.CrossRefGoogle ScholarPubMed
49.Salam, MT, Li, YF, Langholz, B et al. (2005) Maternal fish consumption during pregnancy and risk of early childhood asthma. J Asthma 42, 513518.Google Scholar
50.Romieu, I, Torrent, M, Garcia-Esteban, R et al. (2007) Maternal fish intake during pregnancy and atopy and asthma in infancy. Clin Exp Allergy 37, 518525.CrossRefGoogle ScholarPubMed
51.Calvani, M, Alessandri, C, Sopo, SM et al. (2006) Consumption of fish, butter and margarine during pregnancy and development of allergic sensitizations in the offspring: role of maternal atopy. Pediatr Allergy Immunol 17, 94–102.CrossRefGoogle ScholarPubMed
52.Willers, SM, Devereux, G, Craig, LC et al. (2007) Maternal food consumption during pregnancy and asthma, respiratory and atopic symptoms in 5-year-old children. Thorax 62, 773779.CrossRefGoogle Scholar
53.Fitzsimon, N, Fallon, U, O'Mahony, D et al. (2007) Mothers' dietary patterns during pregnancy and risk of asthma symptoms in children at 3 years. Ir Med J 100, Suppl., 2732.Google ScholarPubMed
54.Sausenthaler, S, Koletzko, S, Schaaf, B et al. (2007) Maternal diet during pregnancy in relation to eczema and allergic sensitization in the offspring at 2 y of age. Am J Clin Nutr 85, 530.Google Scholar
55.Newson, RB, Shaheen, SO, Henderson, AJ et al. (2004) Umbilical cord and maternal blood red cell fatty acids and early childhood wheezing and eczema. J Allergy Clin Immunol 114, 531537.CrossRefGoogle ScholarPubMed
56.Furuhjelm, C, Warstedt, K, Larsson, J et al. (2009) Fish oil supplementation in pregnancy and lactation may decrease the risk of infant allergy. Acta Paediatr 98, 14611467.Google Scholar
57.Olsen, SF, Osterdal, ML, Salvig, JD et al. (2008) Fish oil intake compared with olive oil intake in late pregnancy and asthma in the offspring: 16 y of registry-based follow-up from a randomized controlled trial. Am J Clin Nutr 88, 167175.Google Scholar
58.Haberg, SE, London, SJ, Stigum, H et al. (2009) Folic acid supplements in pregnancy and early childhood respiratory health. Arch Dis Child 94, 180184.Google Scholar
59.Whitrow, MJ, Moore, VM, Rumbold, AR et al. (2009) Effect of supplemental folic acid in pregnancy on childhood asthma: a prospective birth cohort study. Am J Epidemiol 170, 14861493.Google Scholar
60.Matsui, EC & Matsui, W (2009) Higher serum folate levels are associated with a lower risk of atopy and wheeze. J Allergy Clin Immunol 123, 12531259 e2.CrossRefGoogle ScholarPubMed
61.Miller, RL (2008) Prenatal maternal diet affects asthma risk in offspring. J Clin Invest 118, 32653268.Google Scholar
62.Farchi, S, Forastiere, F, Agabiti, N et al. (2003) Dietary factors associated with wheezing and allergic rhinitis in children. [see comment]. Eur Respir J 22, 772780.CrossRefGoogle ScholarPubMed
63.Forastiere, F, Pistelli, R, Sestini, P et al. (2000) Consumption of fresh fruit rich in vitamin C and wheezing symptoms in children. SIDRIA Collaborative Group, Italy (Italian Studies on Respiratory Disorders in Children and the Environment). Thorax 55, 283288.Google Scholar
64.Devereux, G, Litonjua, AA, Turner, SW et al. (2007) Maternal vitamin D intake during pregnancy and early childhood wheezing. Am J Clin Nutr 85, 853859.Google Scholar
65.Devereux, G, Turner, SW, Craig, LCA et al. (2006) Low maternal vitamin E intake during pregnancy is associated with asthma in 5-year-old children. Am J Respir Crit Care Med 174, 499507.Google Scholar
66.Litonjua, AA, Rifas-Shiman, SL, Ly, NP et al. (2006) Maternal antioxidant intake in pregnancy and wheezing illnesses in children at 2 y of age. Am J Clin Nutr 84, 903911.CrossRefGoogle ScholarPubMed
67.Martindale, S, McNeill, G, Devereux, G et al. (2005) Antioxidant intake in pregnancy in relation to wheeze and eczema in the first two years of life. Am J Respir Crit Care Med 171, 121128.CrossRefGoogle ScholarPubMed
68.Murr, C, Schroecksnadel, K, Winkler, C et al. (2005) Antioxidants may increase the probability of developing allergic disease and asthma. Med Hypotheses 64, 973977.Google Scholar
69.Blumer, N, Herz, U, Wegmann, M et al. (2005) Prenatal lipopolysaccharide-exposure prevents allergic sensitisation and airway inflammation, but not airway responsiveness in a murine model of experimental asthma. Clin Exp Allergy 35, 397402.Google Scholar
70.Blumer, N, Sel, S, Virna, S et al. (2007) Perinatal maternal application of Lactobacillus rhamnosus GG suppresses allergic airway inflammation in mouse offspring. Clin Exp Allergy 37, 348357.Google Scholar
71.Douwes, J, Cheng, S, Travier, N et al. (2008) Farm exposure in utero may protect against asthma, hay fever and eczema. Eur Respir J 32, 603611.Google Scholar
72.Teich, R, Conrad, M, Ferstl, R et al. Epigenetic regulation of asthma protection early in life by the nonpathogenic microbe Acinetobacter lwoffii F78. (In the Press.)Google Scholar
73.Johannsen, H & Prescott, SL (2009) Practical prebiotics, probiotics and synbiotics for allergists: how useful are they? Clin Exp Allergy 39, 18011814.Google Scholar
74.Osborn, D & Sinn, J (2007) Probiotics in infants for prevention of allergic disease and food hypersensitivity. Cochrane Database Syst Rev 4, CD006475.Google Scholar
75.Wickens, K, Black, PN, Stanley, TV et al. (2008) A differential effect of 2 probiotics in the prevention of eczema and atopy: a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol 122, 788794.Google Scholar
76.Abrahamsson, TR, Jakobsson, T, Bottcher, MF et al. (2007) Probiotics in prevention of IgE-associated eczema: a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol 119, 11741180.CrossRefGoogle ScholarPubMed
77.Prescott, SL, Wickens, K, Westcott, L et al. (2008) Supplementation with Lactobacillus rhamnosus or Bifidobacterium lactis probiotics in pregnancy increases cord blood interferon-gamma and breast milk transforming growth factor-beta and immunoglobin A detection. Clin Exp Allergy 38, 16061614.Google Scholar
78.Boyle, RJ, Mah, LJ, Chen, A, et al. (2008) Effects of Lactobacillus GG treatment during pregnancy on the development of fetal antigen-specific immune responses. Clin Exp Allergy 38, 18821890.Google Scholar
79.Woodcock, A, Lowe, LA, Murray, CS et al. (2004) Early life environmental control: effect on symptoms, sensitization, and lung function at age 3 years. Am J Respir Crit Care Med 170, 433439.CrossRefGoogle Scholar
80.Snijders, BE, Thijs, C, van Ree, R et al. (2008) Age at first introduction of cow milk products and other food products in relation to infant atopic manifestations in the first 2 years of life: the KOALA Birth Cohort Study. Pediatrics 122, e115e122.Google Scholar
81.Greer, FR, Sicherer, SH & Burks, AW (2008) Effects of early nutritional interventions on the development of atopic disease in infants and children: the role of maternal dietary restriction, breastfeeding, timing of introduction of complementary foods, and hydrolyzed formulas. Pediatrics 121, 183191.Google Scholar
82.Prescott, SL, Smith, P, Tang, MLK et al. (2008) The importance of early complementary feeding in the development of oral tolerance: concerns and controversies. Pediatr Allergy Immunol 19(5), 375380.Google Scholar
83.Agostoni, C, Decsi, T, Fewtrell, M et al. (2008) Complementary feeding: a commentary by the ESPGHAN Committee on Nutrition. J Pediatr Gastroenterol Nutr 46, 99–110.Google Scholar
84.Rebordosa, C, Kogevinas, M, Sorensen, HT et al. (2008) Pre-natal exposure to paracetamol and risk of wheezing and asthma in children: a birth cohort study. Int J Epidemiol 37, 583590.Google Scholar
85.Bisgaard, H, Loland, L, Holst, KK et al. (2009) Prenatal determinants of neonatal lung function in high-risk newborns. J Allergy Clin Immunol 123, 651657; 7 e1–4.Google Scholar
86.Garcia-Marcos, L, Sanchez-Solis, M, Perez-Fernandez, V et al. (2008) Is the effect of prenatal paracetamol exposure on wheezing in preschool children modified by asthma in the mother? Int Arch Allergy Immunol 149, 3337.CrossRefGoogle ScholarPubMed
87.Persky, V, Piorkowski, J, Hernandez, E, et al. (2008) Prenatal exposure to acetaminophen and respiratory symptoms in the first year of life. Ann Allergy Asthma Immunol 101, 271278.CrossRefGoogle ScholarPubMed
88.Shaheen, SO, Newson, RB, Sherriff, A et al. (2002) Paracetamol use in pregnancy and wheezing in early childhood. Thorax 57, 958963.Google Scholar
89.Dehlink, E, Yen, E, Leichtner, AM et al. (2009) First evidence of a possible association between gastric acid suppression during pregnancy and childhood asthma: a population-based register study. Clin Exp Allergy 39, 246253.Google Scholar
90.Hertz-Picciotto, I, Park, HY, Dostal, M et al. (2008) Prenatal exposures to persistent and non-persistent organic compounds and effects on immune system development. Basic Clin Pharmacol Toxicol 102, 146154.CrossRefGoogle ScholarPubMed
91.Noakes, PS, Taylor, P, Wilkinson, S et al. (2006) The relationship between persistent organic pollutants in maternal and neonatal tissues and immune responses to allergens: a novel exploratory study. Chemosphere 63, 13041311.CrossRefGoogle ScholarPubMed
92.Breckler, L, Hale, J, Jung, W et al. (2010) Modulation of in vivo and in vitro cytokine production over the course of pregnancy in allergic and non-allergic mothers. Pediatr Allergy Immunol 21, 1421.CrossRefGoogle ScholarPubMed
93.Breckler, LA, Hale, J, Taylor, A et al. (2008) Pregnancy IFN-gamma responses to foetal alloantigens are altered by maternal allergy and gravidity status. Allergy 63, 14731480.Google Scholar
94.Scott, NM, Hodyl, NA, Murphy, VE et al. (2009) Placental cytokine expression co-varies with maternal asthma severity and fetal sex. J Immunol 182(3), 14111412.CrossRefGoogle Scholar
95.Shanks, N, Windle, RJ, Perks, PA, et al. (2000) Early-life exposure to endotoxin alters hypothalamic-pituitary-adrenal function and predisposition to inflammation. Proc Natl Acad Sci USA 97, 56455650.CrossRefGoogle ScholarPubMed
96.Eder, W, Klimecki, W, Yu, L et al. (2004) Toll-like receptor 2 as a major gene for asthma in children of European farmers. J Allergy Clin Immunol 113, 482488.CrossRefGoogle Scholar
97.Rzehak, P, Heinrich, J, Klopp, N et al. (2009) Evidence for an association between genetic variants of the fatty acid desaturase 1 fatty acid desaturase 2 (FADS1 FADS2) gene cluster and the fatty acid composition of erythrocyte membranes. Br J Nutr 101, 2026.Google Scholar
98.Husemoen, LL, Toft, U, Fenger, M et al. (2006) The association between atopy and factors influencing folate metabolism: is low folate status causally related to the development of atopy? Int J Epidemiol 35, 954961.CrossRefGoogle Scholar
99.Kabesch, M, Hoefler, C, Carr, D et al. (2004) Glutathione S transferase deficiency and passive smoking increase childhood asthma. Thorax 59, 569573.Google Scholar