Hostname: page-component-8448b6f56d-t5pn6 Total loading time: 0 Render date: 2024-04-23T12:47:28.467Z Has data issue: false hasContentIssue false

Immunology of pregnancy

Published online by Cambridge University Press:  25 June 2010

Catherine A. Thornton*
Affiliation:
Institute of Life Science, School of Medicine, Swansea University, Swansea SA2 8PP, UK
*
Corresponding author: Dr Catherine A. Thornton, email c.a.thornton@swansea.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

The conceptual framework for reproductive immunology was put in place over 50 years ago when the survival of the fetal semi-allograft within an immunocompetent mother was first considered. During this time, a number of paradigms have emerged and the mechanisms receiving current attention are those related to immune tolerance, such as regulatory T-cells and indoleamine 2,3,-dioxygenase, and innate immunity, such as natural killer cells, trophoblast debris and inflammation. A key consideration is the temporal and spatial variation in any of these pathways (e.g. implantation v. parturition). As fetally derived trophoblasts are the semi-allogeneic cells with which the maternal immune system comes into contact, understanding the immune response to these cells is critical. There is much interest in the immunological pathways that support a healthy pregnancy and how they might be perturbed in adverse pregnancy outcomes. Additionally, there is increasing awareness that antenatal determinants of the immune function of pregnant women and their offspring have consequences for health and disease in childhood and beyond. Changes in maternal diet over recent decades coincide with the increasing prevalence of allergic and other immune-mediated diseases, and the modification of maternal diet has emerged as a strategy for disease prevention. Approaches undergoing trial at numerous sites around the world include dietary supplementation with fish oil and/or probiotics. Understanding the underlying mechanisms of any positive effect on disease outcomes should reveal further novel strategies for disease prevention.

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

Abbreviations:
CTLA-4

cytotoxic T lymphocyte antigen-4

HLA

human leukocyte antigen

IDO

indoleamine 2,3,-dioxygenase

NK

natural killer

TGF

transforming growth factor

Th1

T helper 1

Th2

T helper 2

Over 50 years ago, three strategies were suggested to explain the immunological survival of the fetus within the self/non-self paradigm: (1) anatomic separation of maternal and fetal tissues; (2) antigenic immaturity of the foetus; (3) lack of response by the maternal immune system (reviewed in Bilington(Reference Billington1)). Along with the possibility of the pregnant uterus as an immunoprivileged site, investigators in reproductive immunology have sought to identify the mechanisms that explain the ability of the immunocompetent mother to tolerate the semi-allogeneic foetus within this conceptual framework.

One of the key things to remember while studying the immunology of pregnancy is the variety of discrete compartments that must be considered: the embryonically derived placenta, the embryonically derived amnion and chorion that form the placental/fetal membranes, the maternally derived decidua (lining of uterus/endometrium of pregnancy), amniotic fluid, the foetus itself and the systemic maternal immune function. Mammary gland development and breast milk could also come under this umbrella. It is also worth noting that temporal differences need to be considered, e.g. implantation v. parturition. Finally, the fetal cells that actually come into direct contact with maternal cells are the trophoblast of the placenta and fetal membranes giving two main sites of interaction to consider, that between the maternal decidua and trophoblast especially in early pregnancy, and that between syncytiotrophoblast on the surface of placental villi and cells in the maternal circulation(Reference Billington1, Reference Sargent, Borzychowski and Redman2).

There are a few examples of impaired responses to infectious agents in pregnancy: pregnant women are more susceptible to respiratory viruses, especially influenza(Reference Longman and Johnson3); pregnant women are more likely than non-pregnant women to develop malaria and to have severe infection with adverse pregnancy outcomes including fetal and maternal death(Reference Lagerberg4). Despite this, the concept of immune suppression during pregnancy has been superseded by the current paradigm of active immunological tolerance of fetal cells by the pregnant woman. Consequently, an inadequate or perturbed tolerance response has been hypothesised to underlie adverse pregnancy outcomes such as preeclampsia(Reference Saito, Sakai and Sasaki5). A number of different immunological mechanisms have been postulated and the more prevalent of these are considered briefly herein.

Non-classical MHC class I

Semi-allogeneic cells of embryonic origin (trophoblast) invade the maternal decidua to open up maternal uterine arteries and facilitate blood supply to the foetus. The interaction with decidual natural killer (NK) cells has a key role in this. One of the roles of NK cells is to destroy human leucocyte antigen (HLA) null cells and the expression of non-classical HLA by trophoblast is postulated to be a mechanism of evading NK cell killing. The non-classical MHC class I molecules HLA-E, -F and -G are less polymorphic than the classical HLA class I molecules and have different expression patterns: all three (as well as HLA-C) are expressed on the extravillous trophoblast that invades the maternal decidua, whereas the villous cytotrophoblast and syncytiotrophoblast found within the placenta express surface HLA-E and secrete a soluble form of HLA-G.

HLA-G is the most extensively studied non-classical MHC class I molecule and the role of HLA-G in modulating the maternal immune response to semi-allogeneic cells of fetal origin is well established(Reference Hunt, Langat and McIntire6, Reference Hunt, Petroff and McIntire7). Soluble and membrane forms of HLA-G are derived by the alternative splicing of HLA-G transcripts. HLA-G interacts with inhibitory receptors on leucocytes and mediates numerous effects: it modulates NK cell activity(Reference Hunt, Langat and McIntire6, Reference Hunt, Petroff and McIntire7); stimulates high levels of anti-inflammatory cytokines, such as transforming growth factor (TGF)-β1, from mononuclear phagocytes(Reference Le Friec, LaupΦze and Fardel8); induces Fas/Fas ligand-mediated apoptosis of CD8+T and CD8+NK cells and inhibits antigen-specific cytotoxic activity of CD8+T-cells(Reference Contini, Ghio and Poggi9). Since the immunomodulatory role of HLA-G in pregnancy was identified, there has been much interest in its potential as either a therapeutic or diagnostic tool in transplant immunology, autoimmunity and cancer(Reference Carosella, Favier and Rouas-Freiss10).

The study of HLA-E and HLA-F was hampered initially by the observation that the expression of these appeared to be restricted to inside the cell. Surface expression of HLA-E has now been found to be dependent on the presence of peptides from other HLA class I molecules. This complex of HLA-E with HLA class I molecule-derived peptides, especially from HLA-G and HLA-C, interacts with either activating or inhibiting CD94/NKG2 on NK cells to modulate NK cell activity. There is little knowledge about HLA-F and the peptides that it binds have not been identified(Reference Ishitani, Sageshima and Hatake11).

Natural killer and natural killer T cells

NK cells have a role in the control of implantation and transformation of the uterine vasculature by trophoblasts, on which blood supply to the foetoplacental unit depends(Reference King and Loke12). There is an accumulation of NK cells in the maternal decidua in the first and second trimester with a reduction in the third trimester(Reference Bulmer, Morrison and Longfellow13, Reference Williams, Searle and Robson14). About 30% of cells in first trimester decidual tissue are leucocytes and up to 70% of these are CD56brightCD16−NK cells. This contrasts with the peripheral blood where CD56dimCD16+NK cells dominate(Reference Bulmer, Morrison and Longfellow13). The CD56bright population of NK cells accumulates at the implantation site and when activated these cells secrete an array of cytokines including macrophage inflammatory protein 1α, granulocyte/macrophage-colony stimulating factor, TNFα, interferon-γ, TGF-β and leukaemia inhibitory factor, with minimal impact on cytotoxicity. CD56bright NK cells express high levels of CD94 and their accumulation at the implantation site has been postulated to be driven by HLA-E/G expression on extravillous cytotrophoblast that acts as a homing receptor. Alternatively, a cascade of cytokine production at the implantation site has been postulated to cause NK cell differentiation.

Interest in natural killer T (NKT) cells at the materno-fetal interface stems from their immunoregulatory properties. As their name suggests, NKT cells have the features of NK and T-cells: like T-cells they express a T-cell receptor, but this is invariant(Reference Godfrey, MacDonald and Kronenberg15); unlike T-cells they are not restricted by MHC class I or class II molecules, but recognise glycolipids in the context of the MHC-like molecule CD1d. Trophoblasts within first trimester decidua express CD1d and 0·48% of CD3+T-cells in the decidua have an NKT phenotype; this represents a 10-fold higher concentration compared to blood. Decidual CD4+NKT cells have a T helper 1 (Th1) bias and produce granulocyte/macrophage-colony stimulating factor, whereas those in the peripheral blood are T helper 2 (Th2)-biased(Reference Boyson, Rybalov and Koopman16).

T helper cytokine profiles

Since the observation that Th2 cytokines are normally produced at the materno-fetal interface, the hypothesis that established pregnancy (i.e. excluding implantation and labour) is associated with a Th2-biased immune response has dominated the reproductive immunology literature(Reference Lin, Mosmann and Guilbert17Reference Piccinni, Scaletti and Maggi19). This is despite the lack of any really convincing data from human subjects and the observation that mice lacking expression of the main Th2 cytokines IL-4, IL-5, IL-9 and IL-13 (i.e. quadruple knockouts) do not have fertility problems (no differences in litter sizes and all offspring healthy) indicating that these cytokines are not essential for fetal survival even during allogeneic pregnancy(Reference Fallon, Jolin and Smith20). It must be noted that progesterone-induced blocking factor induces a Th2 cytokine profile(Reference Szekeres-Bartho and Wegmann21, Reference Szereday, Varga and Szekeres-Bartho22). Progesterone-induced blocking factor is a feature of normal human pregnancy and can be measured in urine from pregnant women; the lack of progesterone-induced blocking factor predicts spontaneous pregnancy termination and neutralising endogenous progesterone-induced blocking factor leads to pregnancy termination in mice(Reference Polgar, Kispal and Lachmann23, Reference Polgar, Nagy and Miko24).

At the outset of the study of Th1/Th2 balance in pregnancy the focus was T-cells themselves but the materno-fetal interface itself is relatively devoid of T-cells. Thus, the paradigm has evolved to encompass type 1 and type 2 cytokines irrespective of their cell source which can include NK cells(Reference Borzychowski, Croy and Chan25Reference Croy, Chantakru and Esadeg27) and the gestation-associated tissues themselves(Reference Jones, Finlay-Jones and Hart28). An appreciation of the temporal and spatial variation in the dominance of either type 1 or type 2 cytokines has also emerged. For example, NK cells and interferon-γ are pivotal to remodelling the uterine vasculature during implantation(Reference Ashkar, Di Santo and Croy26, Reference Croy, Chantakru and Esadeg27). Conversely, in a study using surface markers of Th1 (IL-18 receptor) and Th2 (ST2L) in maternal peripheral blood, NK cells rather than CD4+ and CD8+T cells were found to have a bias to a type 2 phenotype, a bias lost with preeclampsia(Reference Borzychowski, Croy and Chan25).

Overall, there is now an appreciation of the need for balanced type 1/type 2 responses depending on the tissue site and the stage of pregnancy. More recently the role of the innate immune response and inflammation has become of interest. Given that IL-4, IL-10 and IL-13 are anti-inflammatory cytokines(Reference Hart, Bonder and Balogh29), they might serve to prevent an overactive inflammatory response; the type 1/type 2 paradigm should be re-evaluated from this perspective.

Innate immunity

Innate immunity/inflammation is now considered to have a physiological role in implantation, the maintenance of pregnancy and parturition(Reference Ashkar, Di Santo and Croy26, Reference Croy, Chantakru and Esadeg27, Reference Patni, Flynn and Wynen30). The innate immune response also contributes to adverse outcomes in fertility and pregnancy, including tubal infertility associated with Chlamydia trachomatis, infection-associated preterm delivery and preeclampsia(Reference Germain, Sacks and Soorana31, Reference Horne, Stock and King32). There is extensive evidence of a heightened systemic inflammatory response in pregnant women(Reference Sacks, Sargent and Redman33), for example, increased leucocyte count and increased levels of C-reactive protein and other acute phase proteins(Reference Miller34). This might be balanced by increased circulating levels of anti-inflammatory molecules such as TGF-β1(Reference Power, Popplewell and Holloway35) and IL-4(Reference Zhao, Lei and Liu36).

The enhanced systemic inflammatory response during pregnancy has been postulated to be mediated by particulate debris or soluble products derived from the placenta, which enter the maternal circulation. Cell-derived microparticles occur commonly in the circulation, but the levels of these are increased with normal pregnancy being derived from numerous cell types, especially the syncytiotrophoblast. These placenta-derived microparticles are postulated to drive normal systemic inflammation in pregnant women and to be further increased in preeclampsia(Reference Sargent, Borzychowski and Redman2, Reference Redman and Sargent37). These microparticles might also have a role in down-modulated T-cell function during pregnancy. Syncytiotrophoblast microparticles can be found attached to circulating monocytes and unbound in the circulation from the second and increasing into the third trimester. When isolated in vitro from the placenta, they can induce an inflammatory response in non-pregnant women(Reference Germain, Sacks and Soorana31). From animal studies, it has been shown that the presentation of alloantigens coincides with the detectable release of placental debris from around mid-gestation(Reference Erlebacher, Vencato and Price38).

There is much interest in identifying molecules and signalling pathways that might link the so-called type 2 bias of pregnancy with the innate immune function. Recently, it was shown that the immunoregulatory molecule T-cell Ig and mucin domain (Tim)-3 which has been suggested to modulate the Th1/Th2 balance and have a role in the innate immune response is up-regulated on monocytes but not T- and B-cells in the peripheral blood of pregnant women(Reference Zhao, Lei and Liu36). Tim-3 was found to enhance innate immunity and Th1-associated anti-pathogen responses in pregnancy: blocking Tim-3 impaired phagocytosis, signalling via lipopolysaccharide, T-cell proliferation and interferon-γ∴ production. The IL-4 signalling pathway has a role in the up-regulation of Tim-3 expression on monocytes(Reference Zhao, Lei and Liu36). Abnormalities in Tim-3 expression were also observed in women suffering miscarriages at 12–20 weeks of pregnancy.

Regulatory T-cells

Recent years have seen resurged interest in T-cell populations with immunoregulatory or suppressive properties. Initially identified by their ability to suppress autoimmune disease, regulatory T-cells also have been shown to modify a broad range of immunological activities associated with inflammatory and infectious diseases, cancer and transplantation(Reference Read and Powrie39). Abnormalities in the activity of these cells have been implicated in susceptibility to many conditions with underlying immune aetiology. Several types of regulatory T-cells have been described and these can be subdivided broadly into natural and inducible regulatory T-cells. To date, only natural CD4+ regulatory T-cells that can be identified by the expression of CD25 and low expression of CD127 (IL-7 receptor α chain)(Reference Seddiki, Santner-Nanan and Martinson40) have been studied in pregnancy. CD4+/CD25+/CD127lo regulatory T-cells express FoxP3 which has been described as the master regulator of development and function of this population(Reference Fontenot and Rudensky41), and their functional properties have been attributed variously to CTLA-4 (cytotoxic T lymphocyte antigen-4), TGF-β and IL-10, but this remains an area of ongoing investigation and controversy.

In mice and human subjects, CD4+CD25+T-cell numbers increase in early pregnancy and decline from mid-gestation to reach levels equivalent to the non-pregnant state by term(Reference Somerset, Zheng and Kilby42Reference Heikkinen, Mottonen and Alanen44). As CD25 is also a marker of T-cell activation, it is imperative that other phenotypic or functional data be available to clarify that the CD4+CD25+T-cell population being studied is regulatory. CD4+CD25+T-cells able to suppress autoimmune and allogeneic responses are expanded in the spleen, blood and various lymph nodes of pregnant mice. The absence of these cells leads to immunological rejection of the foetus in allogeneic but not syngeneic pregnancies(Reference Aluvihare, Kallikourdis and Betz43). In human subjects, an increase in CD4+CD25+T-cells in peripheral blood in the first and second trimester was described, but without additional phenotypic/functional data it was unclear whether this represented increased numbers of regulatory or activated T-cells(Reference Heikkinen, Mottonen and Alanen44). In a recent study, where a more definitive phenotyping strategy was used, it was found that CD4+CD25+CD127loFoxP3+T-cells were reduced in the second trimester and that expression of FoxP3 itself was lower with pregnancy. The function of this population in pregnant women was comparable to that in non-pregnant women(Reference Mjosberg, Svensson and Johansson45).

CD4+CD25+T-cells are relatively abundant in first trimester decidua (about 20% of decidual CD4+T-cells v. about 8% of CD4+T-cells in peripheral blood of pregnant women) and they respond poorly to T-cell stimulation and have suppressive activity that is cell-contact dependent(Reference Sasaki, Sakai and Miyazaki46). The numbers of CD4+CD25+T-cells in both decidua and peripheral blood in early pregnancy are decreased in spontaneous abortion(Reference Sasaki, Sakai and Miyazaki46). As in peripheral blood, there is a decrease in the relative abundance of these cells by term to about 14%. As many of these express CTLA-4, OX40 and glucocorticoid-induced TNF receptor-related protein, they are likely to have regulatory function(Reference Heikkinen, Mottonen and Alanen44).

The expansion of CD4+CD25+T-regulatory cells occurs very early in pregnancy and declines in both the decidua and blood by term. Therefore, it has been suggested that the expansion in these cells is not driven by exposure to fetal alloantigen. This has led to interest in the impact of pregnancy-related hormones on these cells and both estradiol and progesterone have been implicated(Reference Mjosberg, Svensson and Johansson45, Reference Tai, Wang and Jin47). However, in another animal study, CD4+CD25+T-regulatory cell numbers were greater in allogeneic v. syngeneic pregnancies with the number of male offspring in the litter having a positive impact on numbers of these cells suggesting a role for fetal alloantigens(Reference Lin, Liang and Chen48).

Indoleamine 2,3,-dioxygenase

Indoleamine 2,3,-dioxygenase (IDO) is the first and rate-limiting enzyme in tryptophan degradation. IDO degrades the indole moiety of tryptophan, serotonin and melatonin, and initiates the production of neuroactive and immunoregulatory metabolites known as kynurenines. This leads to local depletion of tryptophan and increased kynurenines both of which affect T-cell proliferation and survival: tryptophan starvation inhibits T-cell activation, and the products of tryptophan catabolism regulate T-cell proliferation and survival(Reference Curti, Trabanelli and Salvestrini49Reference von Bubnoff, Hanau and Wenzel51).

IDO has been implicated in the induction of immune tolerance during many immunologically mediated events, including pregnancy. The treatment of pregnant mice with a pharmacologic inhibitor of IDO-induced maternal T-cell-mediated rejection of allogeneic but not syngeneic foetuses(Reference Munn, Zhou and Attwood52). Inhibition of IDO is also associated with T-cell-dependent, antibody-independent complement deposition and haemorrhagic necrosis at the materno-fetal interface in allogeneic but not syngeneic pregnancies(Reference Mellor, Sivakumar and Chandler53). The timing of fetal loss coincided with the up-regulated expression of IDO(Reference Munn, Zhou and Attwood52). IDO can be detected in day 6 human blastocysts and then throughout pregnancy in syncytiotrophoblast, extravillous cytotrophoblast, placental macrophages and the fetal membranes. Down-regulated phytohaemagglutinin-stimulated proliferation of peripheral blood mononuclear cells by conditioned medium from first trimester placental and decidual cultures is IDO-dependent(Reference Kudo, Boyd and Spyropoulou54). Much of the interest in IDO and pregnancy has focused on the capacity of placenta-/decidua-derived IDO to suppress maternal T-cell reactivity directed at paternal alloantigens. An alternative hypothesis has been put forward, focusing on the potential effects of the products of tryptophan degradation (i.e. kynurenines) that include the removal of oxygen radicals, opening of blood vessels and increased production of NAD(Reference Bonney and Matzinger55).

There is a link between IDO and regulatory T-cells(Reference Curti, Trabanelli and Salvestrini49). CD4+CD25+T-cells induce tryptophan catabolism by dendritic cells and a tolerogenic phenotype in a CTLA-4-dependent manner(Reference Fallarino, Grohmann and Hwang56). Similarly, expression of IDO by dendritic cells and macrophages in maternal decidua is up-regulated via CTLA-4, and this has been suggested to indicate that CTLA-4 expressing CD4+CD25+ regulatory T-cells that infiltrate the decidua in early pregnancy induce IDO expression by decidual dendritic cells/macrophages expressing the appropriate counterligand (B7 family members CD80 and CD86)(Reference Miwa, Hayakawa and Miyazaki57).

Complement

Activation of complement is a strategy for destruction and elimination of pathogens, but can also damage host cells and tissues. The maternal circulation and the placenta contribute to a fully active complement system at the materno-fetal interface. The regulation of complement during pregnancy is critical in preventing maternal-mediated damage of the placenta and foetus. Deficiency in a negative regulator of complement activation (Crry) resulted in embryonic lethality, due to a failure to control complement. This was associated with complement deposition and inflammation, including an extensive polymorphonuclear granulocyte infiltrate in the foetoplacental unit(Reference Xu, Mao and Holers58). There is no direct human counterpart of Crry, but other regulators of complement activation such as decay accelerating factor (CD55), CD59 and membrane co-factor protein (CD46), are expressed extensively in the human placenta. Syncytiotrophoblasts that form the outermost layer of the chorionic villous tree of the placenta and are in direct contact with maternal blood express all three regulatory proteins and these are known to be functional(Reference Girardi, Bulla and Salmon59). Currently, there is particular interest in additional roles for CD46 in reproduction because of its expression on the inner acrosomal membrane of spermatozoa and the observation that CD3/CD46 cross-linking on T-cells induces an IL-10 producing regulatory T-cell population(Reference Liszewski, Kemper and Price60, Reference Kemper, Chan and Green61).

Fetal origins of adult disease

Since Barker first hypothesised on the fetal origins of adult disease there has been a wealth of scientific literature published that relates to this. The essence of this hypothesis is that during fetal development, the organs and systems of the body go through periods when they are plastic and sensitive to the environment. This leads to physiological or morphological changes associated with health and disease in adulthood(Reference Barker62). The original model was proposed for the group of diseases including CHD, stroke, high-blood pressure and type-2 diabetes. The general concept has now been extended to encompass the early life origins of other diseases, such as allergy, and now includes the development of disease in childhood.

Antenatal exposure to dietary components, cigarette smoke, microbial products and many others have been implicated to have a role in the development of the fetal immune system with knock-on effects for health in childhood and beyond. How these exposures impact on maternal systemic immune function during pregnancy, immune function at the materno-fetal interface and fetal immune development are relatively understudied. Recent years have seen an interest in the effect of these exposures on epigenetic modification of chromatin structure enabling heritable gene regulation without changing the nucleotide sequence. This occurs particularly via the modification of histone tails that alter the interaction between histones and DNA. These modifications include acetylation, methylation, phosphorylation, poly-ADP ribosylation and ubiquitination(Reference Burdge, Hanson and Slater-Jefferies63). For example, chronic maternal consumption of a high-fat diet in primates is associated with increased acetylation of candidate genes and altered gene-specific expression in fetal hepatic tissue(Reference Aagaard-Tillery, Grove and Bishop64).

Diet and the early life origins of allergic disease

The concurrent decrease in the consumption of saturated fat and increase in that of polyunsaturated fat parallels changes in allergic disease prevalence. Similarly, the prevalence of obesity in western populations has increased concurrently with asthma and other allergic diseases. These observations have piqued interest in a link between dietary factors, obesity and allergic disease. Of particular interest is the maternal intake of various dietary components during pregnancy. Maternal Zn intake during pregnancy has been inversely associated with wheezing to 2 years of age(Reference Litonjua, Rifas-Shiman and Ly65), asthma ever and current asthma at 5 years of age(Reference Devereux, Turner and Craig66). Low maternal vitamin E intake during pregnancy has been associated with increased proliferative responses by cord blood mononuclear cells(Reference Devereux, Barker and Seaton67), increased likelihood of wheezing to 2 years of age(Reference Litonjua, Rifas-Shiman and Ly65, Reference Martindale, McNeill and Devereux68) and an increased likelihood of asthma/wheezing symptoms during the first 5 years of life(Reference Devereux, Turner and Craig66). The child's own nutrient intake was not associated with any of the outcomes measured.

Folate supplementation before and during early pregnancy has been advised since the demonstration that folic acid supplementation during this time dramatically reduces the risk of neural-tube defect in newborns. More recently, folic acid supplementation during early pregnancy has been suggested to increase the risk of asthma, wheezing and respiratory disease(Reference Ownby69). The physiological effects observed in an animal model were associated with epigenetic effects, and post-natal supplementation of the pups’ diets did not induce the same physiological effects(Reference Hollingsworth, Maruoka and Boon70). An epidemiologic study in over 32 000 children found a small but significant increase in the relative risk for wheezing, lower respiratory tract infections and hospitalisation associated with such infections during the first 18 months of life with maternal intake of folic acid and cod liver oil(Reference Haberg, London and Stigum71). In contrast, serum folate levels in children aged 2 years and older were inversely associated with high total IgE levels, atopy and wheeze(Reference Matsui and Matsui72). This study did not address the issue of maternal supplementation and the observation is in keeping with that from the mouse study(Reference Hollingsworth, Maruoka and Boon70) that it is folate exposure during pregnancy and the associated epigenetic effects that might have a negative impact on allergic disease.

Nutritional approaches to modulating immune function in pregnancy and the peri-natal period

Nutritional supplementation during pregnancy offers the most acceptable way of potentially modulating the immune function of the mother and her offspring and thereby preventing diseases with origins in fetal life. Studies of maternal supplementation with vitamin D, fish oil or probiotics dominate this field.

Vitamin D

Many women are deficient in vitamin D both before and during pregnancy. This has raised concerns for development of the child (e.g. optimal skeletal development) and has been linked to increasing prevalence of allergic diseases such as asthma(Reference Litonjua and Weiss73) and type-1 diabetes(Reference Mathieu and Badenhoop74). The provision of adequate vitamin D to pregnant and breast-feeding women is a leading public health issue in many countries and randomised controlled trials of vitamin D supplementation are underway at numerous sites. This raises concerns that excessive vitamin D might have its own health consequences. Elevated maternal vitamin D in late pregnancy was not associated with the child's body size, intelligence, psychological or cardiovascular health at 9 months and 9 years of age(Reference Gale, Robinson and Harvey75). Despite the epidemiological evidence linking reduced vitamin D with increasing prevalence of allergy, elevated maternal vitamin D in late pregnancy was associated with increased risk of eczema at 9 months and asthma at 9 years of age(Reference Gale, Robinson and Harvey75). Thus, high-maternal exposures have little impact on the parameters of general health, but high levels might have a negative impact on allergic diseases. This observation is supported by studies in mice(Reference Matheu, Back and Mondoc76) and other studies of supplementation in infancy and increased risk of atopic disease in adulthood(Reference Hypponen, Sovio and Wjst77). Larger studies are required as are studies to clarify the impact on immune function at birth of low and high levels of maternal vitamin D. A key consideration is the source of vitamin D in the pregnant woman's diet as maternal vitamin D intake from food is negatively related to the risk of asthma and allergic rhinitis at 5 years of age, but supplements alone were not associated with any outcome(Reference Erkkola, Kaila and Nwaru78).

Fish oil

Decreased n-3:n-6 long chain PUFA in the diet have been suggested to contribute to increased atopic disease in westernised countries(Reference Kompauer, Demmelmair and Koletzko79). There are many published trials of n-3 fatty acid supplementation in children and adults with mixed results. More recently, there has been a shift to supplementation during pregnancy with anticipated immunological effects evident at birth and a positive effect on the development of allergic disease in early infancy and beyond. Fish oil supplements during pregnancy have the desired effects on the n-3:n-6 fatty acid profile of the newborn(Reference Dunstan, Mori and Barden80), but the perceived health benefits of fish oil to the pregnant women themselves remain uncertain(Reference Barden, Dunstan and Beilin81). Maternal fish oil supplements do have functional consequences for the mother and the newborn(Reference Dunstan, Mori and Barden82, Reference Krauss-Etschmann, Hartl and Rzehak83); however, in the newborn this might be a generalised down-regulation of immune function(Reference Krauss-Etschmann, Hartl and Rzehak83, Reference Dunstan, Mori and Barden84). Supplementation during pregnancy also alters the immunomodulatory profile of breast milk with postulated benefits for the development of the infant's mucosal immune system(Reference Dunstan, Roper and Mitoulas85). Maternal supplementation with fish oil tends to have a beneficial effect on the development of allergic disease by the child. However, the size of any effect might relate to the duration of supplementation during pregnancy, the dose of n-3 fatty acids used with higher doses having the most dramatic effects, and the atopic status of the mother(Reference Dunstan, Mori and Barden84, Reference Furuhjelm, Warstedt and Larsson86, Reference Olsen, Osterdal and Salvig87).

Probiotics

Studies on peri-natal supplementation with probiotics stem from observations of altered gastrointestinal microflora associated with and preceding the development of allergic disease in infancy(Reference Bjorksten88). These disease prevention studies typically take the form of maternal supplementation in the last few weeks of pregnancy and then supplementation of the infant for a variable period of time once it is born, though studies with only maternal or infant supplementation have been reported. There are many groups around the world undertaking such studies and many of these are summarised in a recent review of this field(Reference Prescott and Bjorksten89). The outcomes of these studies are variable and the longer term impact of peri-natal supplementation as an allergic disease prevention strategy remains to be determined.

While any effects of probiotics relate to the species used and the population studied, one intriguing aspect of these studies is that supplementation to the pregnant woman for the last few weeks of pregnancy might be the critical component(Reference Tang90). This has led to studies investigating the impact of maternal supplementation with probiotics on immune parameters and gastrointestinal microbiota at birth and in early infancy. Maternal supplementation with Lactobacillus rhamnosus strain GG does not lead to colonisation of the infant with the probiotic strain, despite mothers being colonised at the delivery of the baby, but affects intestinal colonisation of the infants: infants of L. rhamnosus strain GG-exposed mothers were more colonised with species belonging to Bifidobacterium longum group strains, indicating early beneficial effects on the infant's gastrointestinal microflora(Reference Tang90). Maternal supplementation with L. rhamnosus or Bifidobacterium lactis was associated with changes in some immunological mediators in the umbilical cord plasma (e.g. increased interferon-γ) and breast milk (increased TGF-β1, but not IL-6, IL-10 or TNFα levels in breast milk collected 3–7 d postpartum)(Reference Prescott, Wickens and Westcott91). The effects of maternal supplementation with probiotics on TGF-β content of breast milk are variable: lower TGF-β2, but no difference in TGF-β1 in colostrum of women given Lactobacillus reuteri (Reference Bottcher, Abrahamsson and Fredriksson92); higher TGF-β2, but no significant difference in TGF-β1 in breast milk collected 3 months postpartum from women taking L. rhamnosus (Reference Rautava, Kalliomaki and Isolauri93). Again, strain-specific effects that might differ with the length of exposure to the probiotic of interest or time since the exposure was stopped need to be taken into consideration.

The mechanisms of any effect of maternal supplementation with probiotics on immune function and allergic disease risk in the newborn await elucidation. Of interest, supplementation of adults with L. rhamnosus strain GG affects the antigen-specific immune responses of those adults, but maternal supplementation does not influence the same antigen-specific immune responses by her newborn(Reference Boyle, Mah and Chen94). Therefore, understanding the underlying mechanisms requires studies of pregnant women and their newborns rather than extrapolation from studies in healthy non-pregnant adults.

Summary

A number of immunomodulatory pathways contribute to the success of pregnancy with attention currently focused on regulatory T-cells, IDO, NK cells and the innate immune system. The impact of antenatal variation in the immune function on the development of the fetal immune system and thereby health and disease in childhood and beyond is of increasing interest. Changes in the maternal diet over recent decades coincide with the increasing prevalence of allergic and other immune-mediated diseases(Reference Shaheen95, Reference Devereux96). The modification of maternal diet has therefore emerged as a strategy for disease prevention.

Acknowledgements

The author declares no conflict of interest.

References

1.Billington, WD (2003) The immunological problem of pregnancy: 50 years with the hope of progress. A tribute to Peter Medawar. J Reprod Immunol 60, 111.CrossRefGoogle ScholarPubMed
2.Sargent, IL, Borzychowski, AM & Redman, CWG (2006) NK cells and human pregnancy – an inflammatory view. Trends Immunol 27, 399404.CrossRefGoogle ScholarPubMed
3.Longman, RE & Johnson, TR (2007) Viral respiratory disease in pregnancy. Curr Opin Obstet Gynecol 19, 120125.CrossRefGoogle ScholarPubMed
4.Lagerberg, RE (2008) Malaria in pregnancy: a literature review. J Midwifery Womens Health 53, 209215.CrossRefGoogle ScholarPubMed
5.Saito, S, Sakai, M, Sasaki, Y et al. (2007) Inadequate tolerance induction may induce pre-eclampsia. J Reprod Immunol 76, 3039.Google Scholar
6.Hunt, JS, Langat, DK, McIntire, RH et al. (2006) The role of HLA-G in human pregnancy. Reprod Biol Endocrinol 4, Suppl. 1, S10.CrossRefGoogle ScholarPubMed
7.Hunt, JS, Petroff, MG, McIntire, RH et al. (2005) HLA-G and immune tolerance in pregnancy. FASEB J 19, 681693.CrossRefGoogle ScholarPubMed
8.Le Friec, G, LaupΦze, B, Fardel, O et al. (2003) Soluble HLA-G inhibits human dendritic cell-triggered allogeneic T-cell proliferation without altering dendritic differentiation and maturation processes. Hum Immunol 64, 752761.CrossRefGoogle ScholarPubMed
9.Contini, P, Ghio, M, Poggi, A et al. (2003) Soluble HLA-A,-B,-C and -G molecules induce apoptosis in T and NK CD8+ cells and inhibit cytotoxic T cell activity through CD8 ligation. Eur J Immunol 33, 125134.Google Scholar
10.Carosella, ED, Favier, B, Rouas-Freiss, N et al. (2008) Beyond the increasing complexity of the immunomodulatory HLA-G molecule. Blood 111, 48624870.CrossRefGoogle ScholarPubMed
11.Ishitani, A, Sageshima, N, Hatake, K (2006) The involvement of HLA-E and -F in pregnancy. J Reprod Immunol 69, 101113.CrossRefGoogle Scholar
12.King, A & Loke, YW (1991) On the nature and function of human uterine granular lymphocytes. Immunol Today 12, 432435.CrossRefGoogle ScholarPubMed
13.Bulmer, JN, Morrison, L, Longfellow, M et al. (1991) Granulated lymphocytes in human endometrium: histochemical and immunohistochemical studies. Hum Reprod 6, 791798.CrossRefGoogle ScholarPubMed
14.Williams, PJ, Searle, RF, Robson, SC et al. (2009) Decidual leucocyte populations in early to late gestation normal human pregnancy. J Reprod Immunol 82, 2431.CrossRefGoogle ScholarPubMed
15.Godfrey, DI, MacDonald, HR, Kronenberg, M et al. (2004) NKT cells: what's in a name? Nat Rev Immunol 4, 231237.CrossRefGoogle Scholar
16.Boyson, JE, Rybalov, B, Koopman, LA et al. (2002) CD1d and invariant NKT cells at the human maternal–fetal interface. Proc Natl Acad Sci USA 99, 1374113746.CrossRefGoogle ScholarPubMed
17.Lin, H, Mosmann, TR, Guilbert, L et al. (1993) Synthesis of T helper 2-type cytokines at the maternal–fetal interface. J Immunol 151, 45624573.CrossRefGoogle ScholarPubMed
18.Wegmann, TG, Lin, H, Guilbert, L et al. (1993) Bidirectional cytokine interactions in the maternal–fetal relationship: is successful pregnancy a TH2 phenomenon? Immunol Today 14, 353356.CrossRefGoogle ScholarPubMed
19.Piccinni, MP, Scaletti, C, Maggi, E et al. (2000) Role of hormone-controlled Th1- and Th2-type cytokines in successful pregnancy. J Neuroimmunol 109, 3033.Google Scholar
20.Fallon, PG, Jolin, HE, Smith, P et al. (2002) IL-4 induces characteristic Th2 responses even in the combined absence of IL-5, IL-9, and IL-13. Immunity 17, 7–17.CrossRefGoogle ScholarPubMed
21.Szekeres-Bartho, J & Wegmann, TG (1996) A progesterone-dependent immunomodulatory protein alters the Th1/Th2 balance. J Reprod Immunol 31, 8195.Google Scholar
22.Szereday, L, Varga, P & Szekeres-Bartho, J (1997) Cytokine production by lymphocytes in pregnancy. Am J Reprod Immunol 38, 418422.Google Scholar
23.Polgar, B, Kispal, G, Lachmann, M et al. (2003) Molecular cloning and immunologic characterization of a novel cDNA coding for progesterone-induced blocking factor. J Immunol 171, 59565963.CrossRefGoogle ScholarPubMed
24.Polgar, B, Nagy, E, Miko, E et al. (2004) Urinary progesterone-induced blocking factor concentration is related to pregnancy outcome. Biol Reprod 71, 16991705.CrossRefGoogle ScholarPubMed
25.Borzychowski, AM, Croy, BA, Chan, WL et al. (2005) Changes in systemic type 1 and type 2 immunity in normal pregnancy and pre-eclampsia may be mediated by natural killer cells. Eur J Immunol 35, 30543063.Google Scholar
26.Ashkar, AA, Di Santo, JP & Croy, BA (2000) Interferon gamma contributes to initiation of uterine vascular modification, decidual integrity, and uterine natural killer cell maturation during normal murine pregnancy. J Exp Med 192, 259270.CrossRefGoogle ScholarPubMed
27.Croy, BA, Chantakru, S, Esadeg, S et al. (2002) Decidual natural killer cells: key regulators of placental development (a review). J Reprod Immunol 57, 151168.CrossRefGoogle ScholarPubMed
28.Jones, CA, Finlay-Jones, JJ & Hart, PH (1997) Type-1 and type-2 cytokines in human late-gestation decidual tissue. Biol Reprod 57, 303311.CrossRefGoogle ScholarPubMed
29.Hart, PH, Bonder, CS, Balogh, J et al. (1999) Differential responses of human monocytes and macrophages to IL-4 and IL-13. J Leukoc Biol 66, 575578.CrossRefGoogle ScholarPubMed
30.Patni, S, Flynn, P, Wynen, LP et al. (2007) An introduction to Toll-like receptors and their possible role in the initiation of labour. BJOG 114, 13261334.Google Scholar
31.Germain, SJ, Sacks, GP, Soorana, SR et al. (2007) Systemic inflammatory priming in normal pregnancy and preeclampsia: the role of circulating syncytiotrophoblast microparticles. J Immunol 178, 59495956.Google Scholar
32.Horne, AW, Stock, SJ & King, AE (2008) Innate immunity and disorders of the female reproductive tract. Reproduction 135, 739749.Google Scholar
33.Sacks, G, Sargent, I & Redman, C (1999) An innate view of human pregnancy. Immunol Today 20, 114118.CrossRefGoogle ScholarPubMed
34.Miller, EM (2009) Changes in serum immunity during pregnancy. Am J Hum Biol 21, 401403.Google Scholar
35.Power, LL, Popplewell, EJ, Holloway, JA et al. (2002) Immunoregulatory molecules during pregnancy and at birth. J Reprod Immunol 56, 1928.Google Scholar
36.Zhao, J, Lei, Z, Liu, Y et al. (2009) Human pregnancy up-regulates Tim-3 in innate immune cells for systemic immunity. J Immunol 182, 66186624.Google Scholar
37.Redman, CWG & Sargent, IL (2008) Circulating microparticles in normal pregnancy and pre-eclampsia. Placenta 29, Suppl. 1, 7377.Google Scholar
38.Erlebacher, A, Vencato, D, Price, KA et al. (2007) Constraints in antigen presentation severely restrict T cell recognition of the allogeneic fetus. J Clin Invest 117, 13991411.Google Scholar
39.Read, S & Powrie, F (2001) CD4+ regulatory T cells. Curr Opin Immunol 13, 644649.CrossRefGoogle ScholarPubMed
40.Seddiki, N, Santner-Nanan, B, Martinson, J et al. (2006) Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med 203, 16931700.CrossRefGoogle ScholarPubMed
41.Fontenot, JD & Rudensky, AY (2005) A well adapted regulatory contrivance: regulatory T cell development and the forkhead family transcription factor Foxp3. Nat Immunol 6, 331337.Google Scholar
42.Somerset, DA, Zheng, Y, Kilby, MD et al. (2004) Normal human pregnancy is associated with an elevation in the immune suppressive CD25+ CD4+ regulatory T-cell subset. Immunology 112, 3843.Google Scholar
43.Aluvihare, VR, Kallikourdis, M & Betz, AG (2004) Regulatory T cells mediate maternal tolerance to the fetus. Nat Immunol 5, 266271.CrossRefGoogle ScholarPubMed
44.Heikkinen, J, Mottonen, M, Alanen, A et al. (2004) Phenotypic characterization of regulatory T cells in the human decidua. Clin Exp Immunol 136, 373378.Google Scholar
45.Mjosberg, J, Svensson, J, Johansson, E et al. (2009) Systemic reduction of functionally suppressive CD4dimCD25highFoxp3+ Tregs in human second trimester pregnancy is induced by progesterone and 17{beta}-estradiol. J Immunol 183, 759769.Google Scholar
46.Sasaki, Y, Sakai, M, Miyazaki, S et al. (2004) Decidual and peripheral blood CD4+CD25+ regulatory T cells in early pregnancy subjects and spontaneous abortion cases. Mol Hum Reprod 10, 347353.CrossRefGoogle ScholarPubMed
47.Tai, P, Wang, J, Jin, H et al. (2008) Induction of regulatory T cells by physiological level estrogen. J Cell Physiol 214, 456464.CrossRefGoogle ScholarPubMed
48.Lin, Y, Liang, Z, Chen, Y et al. (2006) TLR3-involved modulation of pregnancy tolerance in double-stranded RNA-stimulated NOD/SCID mice. J Immunol 176, 41474154.Google Scholar
49.Curti, A, Trabanelli, S, Salvestrini, V et al. (2009) The role of indoleamine 2,3-dioxygenase in the induction of immune tolerance: focus on hematology. Blood 113, 23942401.Google Scholar
50.Terness, P, Kallikourdis, M, Betz, AG et al. (2007) Tolerance signalling molecules and pregnancy: IDO, galectins, and the renaissance of regulatory T cells. Am J Reprod Immunol 58, 238254.CrossRefGoogle ScholarPubMed
51.von Bubnoff, D, Hanau, D, Wenzel, J et al. (2003) Indoleamine 2,3-dioxygenase-expressing antigen-presenting cells and peripheral T-cell tolerance: another piece to the atopic puzzle? J Allergy Clin Immunol 112, 854860.CrossRefGoogle Scholar
52.Munn, DH, Zhou, M, Attwood, JT et al. (1998) Prevention of allogeneic fetal rejection by tryptophan catabolism. Science 281, 11911193.CrossRefGoogle ScholarPubMed
53.Mellor, AL, Sivakumar, J, Chandler, P et al. (2001) Prevention of T cell-driven complement activation and inflammation by tryptophan catabolism during pregnancy. Nat Immunol 2, 6468.Google Scholar
54.Kudo, Y, Boyd, CAR, Spyropoulou, I et al. (2004) Indoleamine 2,3-dioxygenase: distribution and function in the developing human placenta. J Reprod Immunol 61, 8798.CrossRefGoogle ScholarPubMed
55.Bonney, EA & Matzinger, P (1998) Much IDO about pregnancy. Nat Med 4, 11281129.Google Scholar
56.Fallarino, F, Grohmann, U, Hwang, KW et al. (2003) Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol 4, 12061212.CrossRefGoogle ScholarPubMed
57.Miwa, N, Hayakawa, S, Miyazaki, S et al. (2005) IDO expression on decidual and peripheral blood dendritic cells and monocytes/macrophages after treatment with CTLA-4 or interferon-{gamma} increase in normal pregnancy but decrease in spontaneous abortion. Mol Hum Reprod 11, 865870.CrossRefGoogle ScholarPubMed
58.Xu, C, Mao, D, Holers, VM et al. (2000) A critical role for murine complement regulator crry in fetomaternal tolerance. Science 287, 498501.Google Scholar
59.Girardi, G, Bulla, R, Salmon, JE et al. (2006) The complement system in the pathophysiology of pregnancy. Mol Immunol 43, 6877.CrossRefGoogle ScholarPubMed
60.Liszewski, M, Kemper, C, Price, J et al. (2005) Emerging roles and new functions of CD46. Springer Semin Immunopath 27, 345358.Google Scholar
61.Kemper, C, Chan, AC, Green, JM et al. (2003) Activation of human CD4+ cells with CD3 and CD46 induces a T-regulatory cell 1 phenotype. Nature 421, 388392.CrossRefGoogle ScholarPubMed
62.Barker, DJ (2004) The developmental origins of well-being. Philos Trans R Soc Lond B Biol Sci 359, 13591366.CrossRefGoogle ScholarPubMed
63.Burdge, GC, Hanson, MA, Slater-Jefferies, JL et al. (2007) Epigenetic regulation of transcription: a mechanism for inducing variations in phenotype (fetal programming) by differences in nutrition during early life? Br J Nutr 97, 10361046.CrossRefGoogle ScholarPubMed
64.Aagaard-Tillery, KM, Grove, K, Bishop, J et al. (2008) Developmental origins of disease and determinants of chromatin structure: maternal diet modifies the primate fetal epigenome. J Mol Endocrinol 41, 91–102.Google Scholar
65.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.Google Scholar
66.Devereux, G, Turner, SW, Craig, LC 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
67.Devereux, G, Barker, RN & Seaton, A (2002) Antenatal determinants of neonatal immune responses to allergens. Clin Exp Allergy 32, 4350.Google Scholar
68.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.Google Scholar
69.Ownby, DR (2009) Has mandatory folic acid supplementation of foods increased the risk of asthma and allergic disease? J Allergy Clin Immunol 123, 12601261.Google Scholar
70.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
71.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
72.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.Google Scholar
73.Litonjua, AA & Weiss, ST (2007) Is vitamin D deficiency to blame for the asthma epidemic? J Allergy Clin Immunol 120, 10311035.Google Scholar
74.Mathieu, C & Badenhoop, K (2005) Vitamin D and type 1 diabetes mellitus: state of the art. Trends Endocrinol Metab 16, 261266.CrossRefGoogle ScholarPubMed
75.Gale, CR, Robinson, SM, Harvey, NC et al. (2008) Maternal vitamin D status during pregnancy and child outcomes. Eur J Clin Nutr 62, 6877.CrossRefGoogle ScholarPubMed
76.Matheu, V, Back, O, Mondoc, E et al. (2003) Dual effects of vitamin D-induced alteration of TH1/TH2 cytokine expression: enhancing IgE production and decreasing airway eosinophilia in murine allergic airway disease. J Allergy Clin Immunol 112, 585592.CrossRefGoogle ScholarPubMed
77.Hypponen, E, Sovio, U, Wjst, M et al. (2004) Infant vitamin D supplementation and allergic conditions in adulthood: northern Finland birth cohort 1966. Ann NY Acad Sci 1037, 8495.Google Scholar
78.Erkkola, M, Kaila, M, Nwaru, BI et al. (2009) Maternal vitamin D intake during pregnancy is inversely associated with asthma and allergic rhinitis in 5-year-old children. Clin Exp Allergy 39, 875882.Google Scholar
79.Kompauer, I, Demmelmair, H, Koletzko, B et al. (2004) n6/n3 hypothesis and allergies: biologically plausible, but not confirmed. Eur J Med Res 9, 378382.Google Scholar
80.Dunstan, JA, Mori, TA, Barden, A et al. (2004) Effects of n-3 polyunsaturated fatty acid supplementation in pregnancy on maternal and fetal erythrocyte fatty acid composition. Eur J Clin Nutr 58, 429437.Google Scholar
81.Barden, AE, Dunstan, JA, Beilin, LJ et al. (2006) n-3 fatty acid supplementation during pregnancy in women with allergic disease: effects on blood pressure, and maternal and fetal lipids. Clin Sci (Lond) 111, 289294.Google Scholar
82.Dunstan, JA, 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 randomized, controlled trial. J Allergy Clin Immunol 112, 11781184.Google Scholar
83.Krauss-Etschmann, S, Hartl, D, Rzehak, P et al. (2008) Decreased cord blood IL-4, IL-13, and CCR4 and increased TGF-[beta] levels after fish oil supplementation of pregnant women. J Allergy Clin Immunol 121, 464470.Google Scholar
84.Dunstan, JA, 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 randomized, controlled trial. J Allergy Clin Immunol 112, 11781184.Google Scholar
85.Dunstan, JA, Roper, J & Mitoulas, L (2004) The effect of supplementation with fish oil during pregnancy on breast milk immunoglobulin A, soluble CD14, cytokine levels and fatty acid composition. Clin Exp Allergy 34, 12371242.Google Scholar
86.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.CrossRefGoogle ScholarPubMed
87.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
88.Bjorksten, B (2004) Effects of intestinal microflora and the environment on the development of asthma and allergy. Springer Semin Immunopathol 25, 257270.Google Scholar
89.Prescott, SL & Bjorksten, B (2007) Probiotics for the prevention or treatment of allergic diseases. J Allergy Clin Immunol 120, 255262.CrossRefGoogle ScholarPubMed
90.Tang, ML (2009) Probiotics and prebiotics: immunological and clinical effects in allergic disease. Nestle Nutr Workshop Ser Pediatr Program 64, 219238.Google Scholar
91.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
92.Bottcher, MF, Abrahamsson, TR & Fredriksson, M (2008) Low breast milk TGF-beta2 is induced by Lactobacillus reuteri supplementation and associates with reduced risk of sensitization during infancy. Pediatr Allergy Immunol 19, 497504.Google Scholar
93.Rautava, S, Kalliomaki, M & Isolauri, E (2002) Probiotics during pregnancy and breast-feeding might confer immunomodulatory protection against atopic disease in the infant. J Allergy Clin Immunol 109, 119121.Google Scholar
94.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.CrossRefGoogle ScholarPubMed
95.Shaheen, SO (2008) Prenatal nutrition and asthma: hope or hype? Thorax 63, 483485.Google Scholar
96.Devereux, G (2007) Early life events in asthma – diet. Pediatr Pulmonol 42, 663673.Google Scholar