Hostname: page-component-8448b6f56d-dnltx Total loading time: 0 Render date: 2024-04-15T06:18:48.632Z Has data issue: false hasContentIssue false

Session 1: Allergic disease Nutrition as a potential determinant of asthma

Symposium on ‘Dietary management of disease’

Published online by Cambridge University Press:  08 December 2009

Graham Devereux*
Affiliation:
Department of Child Health, University of Aberdeen, Royal Aberdeen Children's Hospital, AberdeenAB25 2ZB, UK
*
Corresponding author: Dr Graham Devereux, fax +44 1224 551826, email g.devereux@abdn.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Asthma is characterised by chronic lung airway inflammation, increased airway responsiveness and variable airflow obstruction. In Westernised countries asthma is a public health concern because of its prevalence, associated ill health and high societal and healthcare costs. In recent decades there has been a marked increase in asthma prevalence, particularly in Westernised countries. It has been proposed that changing diet has contributed to the increase in asthma. Several dietary hypotheses exist; the first relates the increase in asthma to declining dietary antioxidant intake, the second to decreased intake of long-chain n-3 PUFA and increasing intake of n-6 PUFA. Vitamin D supplementation and deficiency have also been hypothesised to have contributed to the increase in asthma. Observational studies have reported associations between asthma and dietary antioxidants (vitamin E, vitamin C, carotenoids, Se, flavonoids, fruit), lipids (PUFA, butter, margarine, fish) and vitamin D. However, supplementing the diets of adults with asthma with antioxidants and lipids has minimal, if any, clinical benefit. There is growing interest in the possibility that childhood asthma is influenced by maternal diet during pregnancy, with studies highlighting associations between childhood asthma and maternal intake of some nutrients (vitamin E, vitamin D, Se, PUFA) during pregnancy. It has been suggested that maternal diet during pregnancy influences fetal airway and/or immune development. Further intervention studies are needed to establish whether modification of maternal nutrient intake during pregnancy can be used as a healthy low-cost public health measure to reduce the prevalence of childhood asthma.

Type
Research Article
Copyright
Copyright © The Author 2009

Abbreviations:
CBMC

cord blood mononuclear cell

IFN-γ

interferon-γ

Th

T-helper

Asthma is a chronic inflammatory disorder of the lung airways associated with increased airway responsiveness and variable airflow obstruction. Typical symptoms include periodic wheezing, breathlessness, paroxysmal cough and chest tightness, and severity ranges from occasional minor symptoms to disabling persistent severe symptoms and/or frequent life-threatening exacerbations. Asthma is a public health concern in Westernised countries because of its prevalence, associated ill health and high societal and healthcare costs(1Reference Burney, Barnes, Grunstein, Leff and Woolcock3). In the UK 5·2 million individuals receive treatment for asthma, 1·1 million of whom are children. Asthma is responsible annually in the UK for an estimated 12·7 million lost working days, 4·1 million general practitioner consultations and about 70 000 hospital admissions(1, 2). It has been estimated that asthma costs the UK economy about £2·3×109 annually in lost productivity, benefits and National Health Service costs(1, 2).

Asthma and the allergic diseases of atopic eczema and allergic rhinitis (hayfever) are characterised by inflammatory processes, with T-helper (Th)-cell responses of the Th2 phenotype being considered crucial for the initiation and maintenance of asthmatic and allergic inflammation(Reference Georas, Guo and De Fanis4). Cytokines such as IL-4, IL-5 and IL-13 secreted by Th2 cells are important mediators of asthmatic and allergic inflammation that is characterised by elevated IgE, mast-cell degranulation and eosinophilic inflammation(Reference Georas, Guo and De Fanis4, Reference Abbas, Murphy and Sher5). There is increasing interest in the role of regulatory T-cells in the pathogenesis of asthma and allergic disease because of their ability to directly inhibit both Th1 and Th2 responses(Reference Robinson6). Although immunologically-mediated inflammation plays a central role in asthma, it appears that the airway epithelium contributes to the airway inflammatory response(Reference Davies, Wicks and Powell7, Reference Holgate, Davies and Lackie8). The airway epithelium of individuals with asthma appears to be intrinsically abnormal, with increased susceptibility to injury and impaired ability to repair resulting in a chronic pro-inflammatory milieu and airway remodelling(Reference Davies, Wicks and Powell7, Reference Holgate, Davies and Lackie8).

The recent increase in asthma prevalence

Since the 1960s there has been a marked increase in asthma prevalence throughout the world, particularly in Westernised countries(Reference Burney, Barnes, Grunstein, Leff and Woolcock3, Reference Asher, Montefort and Bjorksten9). For example, in the UK the lifetime prevalence of parentally-reported asthma in schoolchildren living in Aberdeen was approximately 4% in 1964, whereas by 2004 it had increased to approximately 26%(Reference Devenny, Wassall and Ninan10, Reference McNeill, Tagiyeva-Milne and Helms11). Although recent studies suggest that asthma prevalence has reached a plateau and might even be declining, asthma remains the most common chronic disease of childhood requiring medication in the developed world(Reference Asher, Montefort and Bjorksten9, Reference Ronchetti, Villa and Barreto12). The recent trends in asthma are most likely to be a consequence of changing environmental exposures rather than changing genetic susceptibility, and current research into the environmental aetiology of asthma is driven by four hypotheses. It has been hypothesised that the increase in asthma is a consequence of increasing exposure to major perennial allergens such as house-dust mite (Dermatophagoides pteronyssinus), because modern housing in general favours the mite and more time is being spent indoors(Reference Platts-Mills, Blumenthal and Perzanowski13). The ‘hygiene hypothesis’ emphasises altered exposure to micro-organisms because of improved hygiene, cleanliness and widespread antibiotic usage(Reference Liu and Leung14). Recently, it has been suggested that the increase in asthma has resulted from increasing obesity and declining physical activity, especially in children(Reference Platts-Mills, Blumenthal and Perzanowski13). In the past decade, changing diet has emerged as a somewhat surprising contributory environmental factor to the increase in asthma.

Asthma: dietary hypotheses

Antioxidants

Four hypotheses relate increasing asthma prevalence to changing diet. In 1994 it was hypothesised that the increase in asthma is a consequence of declining dietary intake of antioxidants(Reference Seaton, Godden and Brown15). This hypothesis was based on the observed increase in asthma at the same time as the decline in dietary intakes of antioxidant-rich foods. European Se intake and blood Se concentrations have declined, probably because of increased use of European grain and changes in bread-making technology(Reference Rayman16). In the UK average Se intake has fallen from 60 μg/d in 1975 to 30–40 μg/d currently(Reference Rayman16, Reference Shortt, Duthie and Robertson17). In the UK vitamin C intake remained relatively stable between 1980 and 2000(18). The limited data on UK vitamin E intake suggest little change from 10·8 mg/d in 1994 to 10·7 mg/d in 2004–5(19). However, extrapolation of the 2000–1 National Diet and Nutrition Survey using the UK National Food Surveys suggests that vitamin E intake in the early 1950s was probably higher at approximately 13–15 mg/d, perhaps because of increased consumption of green vegetables and bread and cereals(18, Reference Henderson, Irving and Gregory20). The mechanism proposed in the original antioxidant hypothesis relates declining lung antioxidant defences to increased oxidant-induced airway damage, airway inflammation and asthma(Reference Seaton, Godden and Brown15). Whilst plausible, the evidence now suggests that this oxidant–antioxidant model is most probably a minor mechanism and that biological effects independent of antioxidant properties are more plausible.

PUFA

In 1997 changes in dietary fat intake were highlighted and it was suggested that changes in dietary fat intake may have contributed to the increase in asthma(Reference Black and Sharpe21). Dietary intake of saturated fats (butter and lard) has decreased and consumption of n-6 PUFA present in margarine and vegetable oils has increased, probably in response to public health measures to reduce CHD(Reference Black and Sharpe21). Also highlighted was the decreasing dietary intake of oily fish rich in long-chain n-3 PUFA, and it was proposed that increasing intake of n-6 PUFA and decreasing intake of n-3 PUFA, particularly those derived from oily fish, had contributed to the increase in asthma. A suggested mechanism relates the increased dietary n-6:n-3 PUFA to increased arachidonic acid levels in inflammatory cell membranes and consequent increased synthesis of PGE2 by the action of cyclooxygenase-2 on arachidonic acid(Reference Black and Sharpe21). In vitro, PGE2 suppresses Th1 differentiation and promotes the Th2 phenotype associated with asthma and allergic disease. Although elegant, this proposed mechanism is probably an oversimplification because the immunological consequences of changing PUFA intake are far more complex than the hypothesis suggests. PGE2 has been reported to have other actions, e.g. suppressing dendritic cell leukotriene B4 secretion by an IL-10-dependent mechanism(Reference Harizi, Juzan and Moreau22). The enzyme pathways that metabolise arachidonic acid also metabolise n-3 PUFA such as EPA to 3-series PG and 5-series leukotrienes that possess biological activity, albeit less potent than arachidonic acid metabolites. Furthermore, both n-3 and n-6 PUFA can modulate T-cell function directly through effects on cell membrane fluidity, cell signalling and gene transcription(Reference Calder, Yaqoob and Thies23) and n-3 PUFA influence the ability of antigen-presenting cells to present antigen to T-cells(Reference Sanderson, MacPherson and Jenkins24).

Vitamin D

Two contradictory hypotheses relate vitamin D to asthma and allergy. In 1999 it was hypothesised that the increase in asthma and allergy is a consequence of widespread rickets prophylaxis with vitamin D supplements in Westernised countries(Reference Wjst and Dold25), highlighting the effects of vitamin D in promoting Th-cell differentiation towards the Th2 phenotype(Reference Lemire, Archer and Beck26, Reference Jirapongsananuruk, Melamed and Leung27). In contrast, in 2007 the widespread vitamin D insufficiency in Westernised countries was highlighted and it was hypothesised that the increase in asthma is a consequence of widespread vitamin D insufficiency(Reference Litonjua and Weiss28). The increasing tendency to stay indoors and the promotion of sunshine avoidance to prevent skin cancer and the inability to compensate by diet-sourced vitamin D are the most likely reasons for inadequate vitamin D status. The effects of vitamin D on promoting regulatory T-cell populations were also highlighted(Reference Xystrakis, Kusumakar and Boswell29, Reference Penna, Roncari and Amuchastegui30) and it was suggested that vitamin D insufficiency promotes the development of asthma and allergy by reducing the inhibitory effect of regulatory T-cells on Th2 immune differentiation(Reference Litonjua and Weiss28).

Observational studies of antioxidants

Many observational epidemiological studies have related dietary antioxidants to asthma and allergic outcomes(Reference Tricon, Willers and Smit31). Most studies have been of cross-sectional or case–control design and studies in adults predominate. These observational studies are beset with the usual problems of quantifying dietary intake, but in addition individuals with asthma and allergies may alter their diet because of their disease. Furthermore, reduced blood antioxidant levels may be a consequence of the systematic oxidant stress associated with the inflammatory processes of asthma(Reference Katsoulis, Kontakiotis and Leonardopoulos32).

Several studies have reported associations between asthma and reduced vitamin E status(Reference Tricon, Willers and Smit31). The Nurses' Health Study has reported low vitamin E intake to be associated with an increased incidence of asthma over a 10-year period(Reference Troisi, Willett and Weiss33) and in Saudia Arabia a case–control study has reported childhood asthma to be associated with reduced dietary vitamin E intake(Reference Hijazi, Abalkhail and Seaton34). Reduced dietary and blood vitamin C has been associated with reduced lung function in adults(Reference Hu and Cassano35) and an increased likelihood of asthma in adults(Reference Huang and Pan36) and children and adolescents(Reference Harik-Khan, Muller and Wise37, Reference Rubin, Navon and Cassano38). Asthma has also been reported to be associated with reduced blood carotenoid concentrations in adults(Reference Riccioni, Bucciarelli and Mancini39, Reference Wood, Garg and Blake40) and children(Reference Harik-Khan, Muller and Wise37, Reference Rubin, Navon and Cassano38). Asthma has been associated with reduced Se status in case–control studies. Dietary Se intake has been reported to be reduced in adults with asthma(Reference Shaheen, Sterne and Thompson41) and blood Se levels and glutathione peroxidase activity has been reported to be reduced in adults(Reference Omland, Deguchi and Sigsgaard42, Reference Qujeq, Hidari and Bijani43) and children(Reference Kocyigit, Armutcu and Gurel44) with asthma. Several studies reporting associations between foods and asthma have highlighted the flavonoid content of the foods as possible antioxidants of interest. In adults apple consumption has been associated with a reduced likelihood of asthma(Reference Shaheen, Sterne and Thompson41) and in children daily consumption of bananas and apple juice from concentrate have been independently associated with a reduced likelihood of wheezing symptoms(Reference Okoko, Burney and Newson45). A high intake of ‘fruity vegetables’, citrus fruit and kiwi fruit has been reported to be associated with a reduced likelihood of wheezing outcomes in children(Reference Chatzi, Torrent and Romieu46Reference Forastière, Pistelli and Sestini48). The Mediterranean diet has been associated with a reduced likelihood of asthma and wheezing in children(Reference Chatzi, Torrent and Romieu46, Reference De Batlle, Garcia-Aymerich and Barraza-Villarreal49).

Antioxidant intervention in established asthma

The epidemiological data suggest that asthma is associated with reduced blood levels and/or reduced dietary intake of the antioxidants vitamin C, vitamin E, some carotenoids, Se, flavonoids and antioxidant-rich fruits(Reference Kaur, Rowe and Arnold50Reference Pearson, Lewis and Britton52). However, these studies cannot differentiate between the possible roles of antioxidants in the aetiology of asthma and the effects of asthma on antioxidant intake and metabolism. Whilst these issues have been partly addressed by studies of antioxidant supplementation in adults with asthma, there remains the need for intervention studies investigating the potential for antioxidant supplementation to primarily prevent asthma.

A Cochrane review of vitamin C supplementation in asthma has concluded that there is insufficient evidence to recommend vitamin C in asthma treatment(Reference Kaur, Rowe and Arnold50). A randomised controlled trial of Se supplementation in subjects with mild asthma has reported no beneficial effects on any of the variables measured(Reference Shaheen, Newson and Rayman51) and vitamin E supplementation was not found to improve any variable of asthma control in a randomised controlled trial conducted in subjects with mild to moderate asthma(Reference Pearson, Lewis and Britton52). Whilst these studies do not support the use of single antioxidant supplements as adjuncts to conventional therapy for asthma, there is still scope for studies of antioxidant combinations and studies in children.

Observational studies of PUFA

Many observational epidemiological studies have related lipid status to asthma and allergic outcomes. The majority of studies have investigated children, perhaps reflecting the focus of the original lipid hypothesis on the effects of PUFA on the first interactions between Th-cells and allergens. Several studies have reported beneficial associations between fish intake and reduced risk of asthma in adults(Reference Laerum, Wentzel-Larsen and Gulsvik53, Reference Miyamoto, Miyake and Sasaki54) and children(Reference Hodge, Salome and Peat55, Reference Kim, Elfman and Mi56). The risk of current asthma is markedly reduced in children who eat fresh oily fish(Reference Hodge, Salome and Peat55) and in adults wheeze and asthma are more likely if they infrequently consume fish(Reference Laerum, Wentzel-Larsen and Gulsvik53). Fish intake has been associated with an increased risk of asthma in children; however, the reported association was not adjusted for potential confounding by socio-economic factors(Reference Takemura, Sakurai and Honjo57).

In addition to fish, surrogate markers for PUFA intake that have been studied include oils, butter and margarine. Although an increased dietary intake of margarine has been associated with an increased likelihood of allergic outcomes(Reference Bolte, Frye and Hoelscher58), several studies have reported associations between asthma outcomes and margarine and butter intake(Reference Bolte, Winkler and Holscher59Reference Foliaki, Annesi-Maesano and Tuuau-Potoi61). In adults high margarine intake has been associated with an increased likelihood of adult-onset asthma(Reference Nagel and Linseisen60) and in children regular margarine consumption has been associated with an increased likelihood of wheezing symptoms(Reference Foliaki, Annesi-Maesano and Tuuau-Potoi61). The use of PUFA in spreads and cooking oils has been linked to recent asthma in children(Reference Haby, Peat and Marks62), and polyunsaturated oil intake has been associated with an increased risk of childhood wheezing symptoms(Reference Kim, Elfman and Mi56).

Associations have been reported between asthma outcomes and dietary intakes and blood concentrations of individual PUFA(Reference Miyake, Sasaki and Arakawa63Reference Woods, Raven and Walters66); however, many of these studies are characterised by numerous comparisons and a relative paucity of significant associations. A study that has quantified the dietary intake of individual PUFA by children has reported childhood wheezing to be more likely with increasing intakes of total PUFA, total n-3, total n-6 and linoleic acid(Reference Miyake, Sasaki and Arakawa63). Current asthma in children has been reported to be more likely with higher n-6 PUFA:n-3 PUFA(Reference Oddy, de Klerk and Kendall64). Blood PUFA concentrations in adults and children have similarly been related to asthma and wheeze, with increasing serum levels of linoleic acid being associated with reduced childhood wheeze and asthma, whereas increasing arachidonic acid levels have been associated with an increased risk of childhood wheeze and asthma(Reference Bolte, Kompauer and Fobker65). The same study has shown increasing linolenic acid levels and linolenic acid:linoleic acid to be associated with increased wheeze and asthma outcomes(Reference Bolte, Kompauer and Fobker65). In adults no consistent associations between plasma n-3 PUFA levels and asthma outcomes have been found, but current asthma have been shown to be associated with increasing plasma dihomo-γ-linolenic acid levels(Reference Woods, Raven and Walters66).

The observational data consistently report beneficial associations between fish intake and asthma; however, the studies of individual PUFA suggest that both n-3 and n-6 PUFA appear to be adversely and beneficially associated with asthma outcomes. Whilst these studies support an association between lipids and asthma, they provide little evidence to support the original lipid hypothesis.

PUFA intervention in established asthma

Whilst the observational studies suggest an association between PUFA and asthma they cannot elaborate on the temporal associations between diet and asthma and whether the associations observed have contributed to the aetiology of asthma or are a consequence of disease. These issues have been addressed to some extent by intervention studies that have investigated the therapeutic potential of n-3 PUFA supplements (usually in the form of fish oil capsules) in established asthma and in the primarily prevention of childhood asthma.

A Cochrane Review of marine n-3 PUFA supplementation in asthma has concluded that there is no convincing evidence that marine n-3 PUFA supplementation leads to an improvement in asthma control(Reference Thien, De Luca and Woods67). The review highlights two exemplary randomised controlled trials of n-3 PUFA supplementation that are nevertheless excluded from the review. In elite athletes with exercise-induced bronchoconstriction n-3 PUFA supplementation reduces exercise-induced bronchoconstriction and urinary and blood inflammatory markers(Reference Mickleborough, Murray and Ionescu68). It is unlikely that these observations can be translated to non-elite non-athletic individuals with asthma who are not exercising to volitional exhaustion. In a subsequent study subjects with asthma with exercise-induced bronchoconstriction were supplemented with n-3 PUFA, which was found to reduce rescue bronchodilator use, exercise-induced bronchoconstriction and sputum inflammatory markers(Reference Mickleborough, Lindley and Ionescu69). The clinical applicability of the latter study is limited by the cessation of all asthma maintenance therapies during the study.

Studies of n-3 PUFA supplementation conducted since the Cochrane review(Reference Thien, De Luca and Woods67) have reported conflicting results. n-3 PUFA supplementation has been reported to reduce markers of bronchial inflammation but to have no clinical benefit in subjects with house-dust mite-sensitised asthma(Reference Schubert, Kitz and Beerman70). In another small study n-3 PUFA supplementation of women with asthma was not found to be associated with any improvement in inflammatory markers, asthma control or lung function(Reference Moreira, Moreira and Delgado71).

Most studies of n-3 PUFA (and antioxidant) supplementation have been conducted in adults and there is a need for well-conducted studies in children. Recently, it has been reported that n-3 PUFA, vitamin C and Zn supplementation of children with asthma, either individually or in combination, improves asthma control, lung function and inflammatory markers(Reference Biltagi, Baset and Bassiouny72). At the present time, however, there is little evidence to support the use of n-3 PUFA as adjuncts to conventional therapy in asthma.

Early-life influences in the aetiology of asthma

Although the current evidence suggests that antioxidant and/or n-3 PUFA supplementation have little clinical beneficial effect, there is increasing interest in the notion that early-life diet influences the development of asthma. It is now evident that early-life factors play a critical role in the development of asthma and allergy(Reference Holt, Devereux, Sly, Holgate, Church and Lichenstein73). Anthropometric and lung function measurements at birth are associated with the development of asthma and allergic disease during childhood and adulthood(Reference Håland, Lødrup Carlsen and Sandvik74, Reference Katz, Pocock and Strachan75) and maternal smoking during pregnancy increases the risk of childhood asthma(Reference Gilliland, Li and Peters76). In addition, neonatal in vitro cord blood mononuclear cell (CBMC) responses after stimulation with allergens have been shown to be associated with recognised risk factors for asthma(Reference Devereux and Barker77) and the subsequent development of childhood asthma and allergic disease(Reference Prescott78).

Maternal intakes of some antioxidants, PUFA and vitamin D during pregnancy have been highlighted as potential determinants of childhood asthma. The concept that diet primarily influences asthma aetiology during fetal development might explain the ineffectiveness of dietary supplementation in adults with asthma discussed earlier. It has also been suggested that the associations reported between antioxidants and asthma in older children and adults are indirect reflections of associations with maternal diet during pregnancy(Reference Devereux and Seaton79). Habitual dietary patterns are established in early childhood and are influenced by parental dietary habits such that there are correlations between the diets of parents and their children, and within individuals between adult and childhood diet(Reference Lynch, Kaplan and Salonen80, Reference Talvia, Rasanen and Lagstrom81).

The postulated mechanisms by which maternal diet during pregnancy could influence the development of asthma in children have focused on fetal airway and immune development because of the potential for small changes in nutrient status to exert disproportionately potent irreversible long-term influences on the development of childhood asthma. Airway development occurs predominantly antenatally and is nearly complete by birth; subsequent development is limited to increases in length and diameter(Reference Stick82). Disturbances of respiratory epithelial and mesenchymal development, particularly branching morphogenesis, have been implicated in the pathogenesis of asthma(Reference Davies, Wicks and Powell7, Reference Holgate, Davies and Lackie8) and it is possible that suboptimal fetal nutrient status adversely affects respiratory epithelial and mesenchymal development resulting in suboptimal early-life airway function. Such an adverse effect on fetal airway development will be associated with an increased risk of developing asthma because reduced neonatal lung function is associated with an increased risk of wheezing and asthma in later childhood(Reference Håland, Lødrup Carlsen and Sandvik74, Reference Turner, Palmer and Rye83).

Early-life dietary influences on the initial interactions between the Th-cells and allergens have the potential for long-term irreversible effects. Any suboptimal nutritional deviation of initial encounters between allergen and Th-cells towards the Th2 phenotype are likely to be disproportionately exaggerated by the natural immunoregulatory mechanisms that promote further Th-cell differentiation towards the Th2 phenotype and inhibit development of the reciprocal Th1 phenotype(Reference Abbas, Murphy and Sher5). Whilst it seems highly likely that neonates are exposed to allergen fairly soon after delivery, there is debate as to whether the fetus is exposed to allergens and, if so, whether such exposure influences the immune development, asthma and allergic disease(Reference Prescott and Jones84). The concept that PUFA influence the earliest interactions between the immune system and allergens is inherent to the original lipid hypothesis and may be the reason why most studies in this field have focused on the early-life influences of PUFA. There are relatively few studies of early-life influences of antioxidants and vitamin D.

Early-life lipid status and asthma

Maternal and infant dietary intakes of foods pertinent to the lipid hypothesis during pregnancy have been associated with childhood asthma and allergic disease. In the Children's Health Study maternal intake of oily fish during pregnancy was found to be associated with a reduced likelihood of asthma in children at age 5 years; however, this association was limited to mothers with asthma and maternal diet was assessed >10 years after the pregnancy(Reference Salem, Li and Langholz85). In the prospective Dutch Prevention and Incidence of Asthma and Mite Allergy birth cohort study the daily intake of foods containing milk fat (butter, full cream milk, milk products) at age 2 years was shown to be associated with reduced likelihood of asthma and/or wheeze at the age of 3 years(Reference Wijga, Smit and Kerkhof86).

PUFA concentrations of umbilical cord blood have been related to the subsequent development of childhood asthma and allergic disease with complex and somewhat contradictory results. The UK Avon Longitudinal Study of Parents and Children Study is the largest study relating wheeze and eczema in the first 42 months of life to erythrocyte-membrane PUFA composition of cord blood and maternal blood at 20 weeks of gestation(Reference Newson, Shaheen and Henderson87). Although no associations were demonstrated with maternal PUFA concentrations, several associations with cord blood PUFA concentrations were demonstrated; however, because of the large number of statistical comparisons performed, the authors conclude that it seems unlikely that fetal exposure to n-3 and n-6 PUFA is an important determinant of early-childhood wheezing and allergic disease(Reference Newson, Shaheen and Henderson87). The potential for PUFA to influence the first interactions between the immune system and allergens has been investigated in the US Project Viva birth cohort, which has reported increased cord plasma EPA and arachidonic acid concentrations to be associated with reduced CBMC proliferative and interferon-γ (IFN-γ) responses(Reference Gold, Willwerth and Tantisira88). In addition, higher cord plasma linoleic acid was shown to be associated with increased IL-13 responses(Reference Gold, Willwerth and Tantisira88). The similarity of the associations with n-3 and n-6 PUFA, whilst consistent with some of the observational studies outlined earlier(Reference Miyake, Sasaki and Arakawa63) are at variance with the lipid hypothesis. The findings are however consistent with the observation that IFN-γ responses of murine splenic lymphocytes are reduced by both n-3 and (less potently) n-6 PUFA supplementation(Reference Wallace, Miles and Evans89). Possible explanations for the observations could be the direct effects of n-3 and n-6 PUFA on Th-cells and/or non-PGE2 metabolites. Alternatively, n-3 and n-6 PUFA may be influencing regulatory T-cells(Reference Robinson6).

Although further observational work is required, a number of early-life intervention studies based on the original lipid hypothesis have been conducted. As a follow-up to a Danish intervention study investigating the effects of fish oil supplementation on pregnancy outcomes, registry data were used to ascertain the asthma status of children born to women who had supplemented their diets from 30 weeks of gestation with fish oil or olive oil and a further group of women who had been randomised to take no capsules. When compared with the children whose mothers received olive oil, the children of women receiving the fish oil supplement were less likely to develop asthma(Reference Olsen, Osterdal and Salvig90). Unfortunately, it cannot be concluded from this study that fish oil supplementation during pregnancy reduces the likelihood of childhood asthma because the children of women nominally taking no capsules during pregnancy had asthma outcomes remarkably similar to those of the fish oil-supplemented group(Reference Olsen, Osterdal and Salvig90). This surprising association may have been a consequence of cross-contamination of study groups, with women allocated to ‘no capsules’ choosing to take commercially-available fish oil capsules.

The potential for PUFA to influence the first interactions between the immune system and allergens has also been investigated by an intervention study in which the CBMC responses of children of women supplemented with n-3 PUFA-rich fish oil from 20 weeks of gestation were compared with those of women who had received control olive oil capsules(Reference Dunstan, Mori and Barden91). Maternal fish oil supplementation was found to be associated with a general reduction in CBMC cytokine responses; however, only the reduction in IL-10 response after stimulation with cat allergen was found to be significant (P=0·046). These observations suggest that n-3 PUFA in pregnancy may influence neonatal immune responses to allergens; however, it remains to be seen whether the immunological associations are translated into clinical effects.

The Childhood Asthma Prevention Study is a randomised controlled trial that has investigated the potential for preventing asthma by supplementing the diets of infants with n-3 PUFA(Reference Marks, Mihrshahi and Kemp92). Infants were supplemented with n-3 PUFA-rich fish oil at the commencement of formula feed or at 6 months if breast-fed. Although n-3 PUFA supplementation was found to be associated with reductions in wheeze outcomes in the first 18 months of life, at follow-up at 3 and 5 years minimal, if any, beneficial effect was found to be indicated and it was concluded that n-3 PUFA-rich fish oil supplementation reduces the likelihood of early-childhood wheeze but has no effect on the likelihood of asthma, wheeze and allergic disease in later childhood(Reference Marks, Mihrshahi and Kemp92). Although a negative trial, the study highlights a number of issues relating to PUFA supplementation trials. It is possible that n-3 PUFA intervention commenced ‘too late’, and given the evidence that n-3 and n-6 PUFA may have similar effects on Th-cells(Reference Miyake, Sasaki and Arakawa63, Reference Gold, Willwerth and Tantisira88, Reference Wallace, Miles and Evans89) the use of n-6 PUFA supplementation in the control group may have masked any beneficial effect of n-3 PUFA supplementation.

Maternal antioxidant intake during pregnancy and childhood asthma

The potential for maternal antioxidant intake during pregnancy to modify the risk of childhood asthma has been highlighted by several birth cohort studies. The reported associations also support the notion that biological properties of antioxidants independent of antioxidant activity influence fetal airway and immune development. The UK Avon Longitudinal Study of Parents and Children Study has reported low umbilical cord Se concentrations to be associated with an increased likelihood of persistent wheeze in children up to 42 months(Reference Shaheen, Newson and Henderson93). A subsequent Scottish birth cohort study has similarly reported that low maternal plasma Se concentrations in pregnant women and neonates are associated with an increased likelihood of wheezing at 2 years(Reference Devereux, McNeill and Newman94). However, by age 5 years there is an absence of associations between maternal and/or neonatal Se status and asthma outcomes, perhaps reflecting a short-term effect of maternal Se intake during pregnancy on early-life immune responses to viral infection(Reference Devereux, McNeill and Newman94).

The Project Viva cohort study has reported associations that suggest that maternal diet during pregnancy influences neonatal immune responses to allergens and childhood wheezing symptoms. Low maternal intake of vitamin E and some carotenoids (β-carotene, leutein+zeaxanthin) during pregnancy is associated with increased CBMC proliferative responses(Reference Litonjua, Tantisira and Finn95). The study also reports that low maternal vitamin E intake during pregnancy is associated with an increased likelihood of wheeze outcomes in children at age 2 years(Reference Litonjua, Rifas-Shiman and Ly96). A Scottish birth cohort study has also reported that low maternal vitamin E intake during pregnancy is associated with increased neonatal CBMC proliferative responses(Reference Devereux, Barker and Seaton97) and an increased likelihood of wheezing and asthma outcomes in 5-year-old children, together with increased exhaled breath NO (a marker of asthmatic airway inflammation)(Reference Devereux, Turner and Craig98). In addition, low maternal plasma α-tocopherol levels during pregnancy are associated with reduced lung function at age 5 years(Reference Devereux, Turner and Craig98). Furthermore, there is an association between reduced maternal apple intake during pregnancy and an increased likelihood of wheeze and asthma outcomes in children at 5 years. However, there are no associations with other fruits, suggesting an apple-specific effect, possibly because of their phytochemical content, such as flavonoids(Reference Willers, Devereux and Craig99). The Irish Life-ways Cross-Generation Cohort Study has reported that high maternal fruit and vegetable intake during pregnancy are associated with a reduced likelihood of asthma in children at age 3 years(Reference Fitzsimon, Fallon and O'Mahony100). A Menorcan birth cohort study has reported that wheeze in children aged 6–7 years is less frequent if mothers consume a Mediterranean diet during pregnancy(Reference Chatzi, Torrent and Romieu101). The Menorcan and the Scottish studies quantified the diets of the children and it was found that whilst maternal and child diets are correlated there are no associations with children's diets(Reference Devereux, Turner and Craig98, Reference Chatzi, Torrent and Romieu101).

The associations reported by these studies suggest that maternal nutrient intake during pregnancy (particularly vitamin E and probably flavonoids) influences the development of childhood asthma, possibly by influencing fetal airway development and the first interactions between the neonatal immune system and allergens(Reference Litonjua, Tantisira and Finn95, Reference Devereux, Barker and Seaton97). The absence of a general association with antioxidants such as vitamin C and carotenoids and the associations with immune and lung function suggest that biological properties of vitamin E independent of antioxidant activity may be relevant(Reference Azzi, Gysin and Kempna102). Vitamin E and flavonoids have been reported to have complex effects on immunological and inflammatory pathways that may be relevant to the development of asthma and allergic disease(Reference Azzi, Gysin and Kempna102Reference Manthey104). The anti-inflammatory effects of vitamin E are believed to be mediated by the inhibition of protein kinase B and C activity, which may inhibit the activation and DNA binding of the transcription factor NF-κB, with consequent effects on the regulation of inflammatory cytokines, adhesion molecules and other pro-inflammatory molecules.

Vitamin E has many effects on the immune system, with reduced vitamin E status promoting the Th2 phenotype either by reducing IFN-γ secretion and/or increasing IL-4 or IL-5 secretion by Th-cells(Reference Okamoto, Murata and Tamai105Reference Meydani, Han and Wu109). Several mechanisms have been proposed for these immunomodulatory effects. Vitamin E influences PGE2 secretion by macrophages(Reference Wang, Huang and Eskelson108), with reduced vitamin E status being associated with increased cyclooxygenase-2 activity and increased production of PGE2, which has been shown to promote the Th2 phenotype. Vitamin E has also been reported to inhibit the binding of NF-κB and activator protein-1 to the IL-4 promoter region, down regulating IL-4 mRNA expression in human Th-cells(Reference Li-Weber, Giasisi and Trieber106). Vitamin E may also influence the development of regulatory T-cells(Reference Tan, Sagoo and Chan110). The effects of vitamin E on the immune system appear to be age related, enhancing the IFN-γ responses of young but not old mice(Reference Wakikawa, Utsuyama and Wakabayashi111), and in human subjects naïve Th-cells are more responsive than memory Th-cells to vitamin E(Reference Okamoto, Murata and Tamai105, Reference Malmberg, Lenkei and Petersson107Reference Meydani, Han and Wu109). These age-dependent associations may explain the ineffectiveness of vitamin E supplementation in adults with asthma; conversely, any putative effects of vitamin E on asthma may be most potent in early infancy during initial encounters with allergens rather than after disease has been established.

Although flavonoids are best known for their powerful antioxidant properties, they also have anti-inflammatory and immunomodulatory properties that appear analogous to those of vitamin E. Flavonoids have been reported to inhibit protein kinase B and C and the activation and DNA binding of the transcription factor NF-κB with consequent effects on the regulation of inflammatory cytokines, adhesion molecules and other pro-inflammatory molecules. Flavonoids have also been reported to inhibit the synthesis of eicosanoid metabolites by inhibiting cyclooxygenase-2 and 5-lipoxygenase activity(Reference Manthey104, Reference Xagorari, Papapetropoulos and Mauromatis112). In animal models flavonoid supplementation inhibits Th2 differentiation and allergic responses by enhancing the Th-cell IFN-γ responses and inhibiting the IL-4 and IL-5 secretion(Reference Rogerio, Kanashiro and Fontanari113). Supplementation of human peripheral mononuclear cells with the flavonoid quercetin has been reported to increase gene and protein expression of IFN-γ and inhibit the expression of IL-4(Reference Nair, Kandaswami and Mahajan114).

The reported association between low maternal plasma α-tocopherol levels at 12 weeks of gestation and reduced lung function in children at age 5 years(Reference Devereux, Turner and Craig98) is consistent with the notion that vitamin E influences fetal airway development. In rat models maternal vitamin E supplementation accelerates growth in hypoplastic lungs, increasing lung complexity, surface area and bud count(Reference Islam, Narra and Cote115). Vitamin E also modulates the expression of some genes implicated in cell signalling, cell-cycle regulation and extracellular matrix proteins that could be potentially relevant to fetal airway development(Reference Azzi, Gysin and Kempna116, Reference Ricciarelli, Maroni and Ozer117).

Vitamin D

The investigation of associations between vitamin D and asthma has focused on possible associations with early-life exposure, probably reflecting the proposed influences of vitamin D on early-life interactions between the immune system and allergens. The few observational studies have reported conflicting results. Two birth cohort studies have reported potentially beneficial associations between maternal dietary vitamin D intake during pregnancy and childhood wheeze; however, no account was made of sunlight-derived vitamin D. In Project Viva low maternal vitamin D intake during pregnancy was found to be associated with an increased risk of recurrent wheeze at age 3 years(Reference Camargo, Rifas-Shiman and Litonjua118) and in a Scottish study low maternal vitamin D intake was reported to be associated with an increase in wheezing outcomes in children at age 5 years(Reference Devereux, Litonjua and Turner119). Two cohort studies have reported potentially adverse associations. In a large Northern Finnish study regular high-dose (≥50 μg/d) vitamin D supplementation during infancy has been associated with an increased likelihood of allergic outcomes at 31 years(Reference Hypponen, Sovio and Wjst120). However, many of the characteristics predictive of reduced compliance with vitamin D supplementation were shown to be associated with reduced risk of asthma and allergies and after adjustment for these factors no association between vitamin D supplementation and asthma was found. A UK birth cohort has reported that elevated maternal blood 25-hydroxyvitamin D in the third trimester is associated with an increased likelihood of childhood asthma at 9 years; however, the study response rate was low (30%) and there was no adjustment for potential confounding(Reference Gale, Robinson and Harvey121).

Conclusions

There is now evidence of an association between nutrition and asthma; however, the clinical and public health relevance of the associations remains unclear. At the present time there is insufficient evidence to advise individuals with asthma, pregnant women, parents and children to change or supplement their diet in order to treat or reduce the risk of developing asthma. There is also little evidence that antioxidant or PUFA supplementation can be used as an adjunct to conventional therapy for asthma, although there is still a need to conduct well-designed randomised controlled trials of nutrient intervention in children with asthma and for trials of combinations of nutrients. The emerging associations between maternal intake of vitamin E, vitamin D and PUFA during pregnancy and childhood asthma provides tantalising evidence that perhaps nutritional supplementation in the developmental context of pregnancy may have a role in the primary prevention of asthma in children. This concept is already established for folic acid supplementation during pregnancy for the prevention of neural-tube defects(122). Before pregnant women can be advised to modify their diet, further work is needed. Particularly important will be well-designed intervention studies, and at the present time trials of vitamin E, vitamin D and PUFA supplementation seem the most promising. Whilst nutritional supplementation of pregnant women in a double-blind placebo-controlled manner is relatively straightforward, nutrients are seldom eaten in isolation and it is possible that for optimal effects a single-nutrient supplement may require other nutrients, this problem is probably mostly easily addressed by providing the nutrient in its natural form, food. Food also addresses the issue of the identified nutrient being a marker for other nutrients or combinations of nutrients. Even if dietary modification can be shown to reduce the risk of asthma, it is extremely unlikely that dietary modifications will reduce the prevalence of asthma by >30–40%, because asthma is a multifactoral disease requiring multiple exposures in genetically-predisposed individuals and dietary exposures are likely to be only one of many risk factors within an individual.

Acknowledgements

The author declares no conflicts of interest.

References

1.Asthma UK (2004) Where do we stand? http://www.asthma.org.uk/document.rm?id=18 (accessed April 2009).Google Scholar
2.Asthma UK (2004) Living on a knife edge. http://www.asthma.org.uk/document.rm?id=98 (accessed April 2009).Google Scholar
3.Burney, P (1997) Epidemiological trends. In Asthma, pp. 3547 [Barnes, P, Grunstein, M, Leff, A and Woolcock, AJ, editors]. Philadelphia, PA: Lippincott-Raven.Google Scholar
4.Georas, SN, Guo, J, De Fanis, U et al. (2005) T-helper cell type-2 regulation in allergic disease. Eur Respir J 26, 11191137.CrossRefGoogle ScholarPubMed
5.Abbas, AK, Murphy, KM & Sher, A (1996) Functional diversity of helper T lymphocytes. Nature 383, 787793.CrossRefGoogle ScholarPubMed
6.Robinson, DS (2004) Regulation: the art of control? Regulatory T cells and asthma and allergy. Thorax 59, 640643.CrossRefGoogle ScholarPubMed
7.Davies, DE, Wicks, J, Powell, RM et al. (2003) Airway remodelling in asthma: new insights. J Allergy Clin Immunol 111, 215225.Google Scholar
8.Holgate, ST, Davies, DE, Lackie, PM et al. (2000) Epithelial-mesenchymal interactions in the pathogenesis of asthma. J Allergy Clin Immunol 105, 193204.CrossRefGoogle ScholarPubMed
9.Asher, MI, Montefort, S, Bjorksten, B et al. (2006) Worldwide time trends in the prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and eczema in childhood: ISAAC Phases One and Three repeat multicountry cross-sectional surveys. Lancet 368, 733743.CrossRefGoogle ScholarPubMed
10.Devenny, A, Wassall, H, Ninan, T et al. (2004) Respiratory symptoms and atopy in children in Aberdeen: questionnaire studies of a defined school population repeated over 35 years. Br Med J 329, 489490.CrossRefGoogle ScholarPubMed
11.McNeill, G, Tagiyeva-Milne, N, Helms, PJ et al. (2009) Changes in the prevalence of asthma, eczema and hayfever in prepubertal children: a 40 year perspective. Paed Perinatal Epidemiol (In the Press).Google Scholar
12.Ronchetti, R, Villa, MP, Barreto, M et al. (2002) Is the increase in childhood asthma coming to an end? Findings from three surveys of schoolchildren in Rome, Italy. Eur Respir J 17, 881886.CrossRefGoogle Scholar
13.Platts-Mills, TAE, Blumenthal, K, Perzanowski, M et al. (2000) Determinants of clinical allergic disease. The relevance of indoor allergens to the increase in asthma. Am J Respir Crit Care Med 162, S128S133.Google Scholar
14.Liu, AH & Leung, DYM (2006) Renaissance of the hygiene hypothesis. J Allergy Clin Immunol 117, 10631066.Google Scholar
15.Seaton, A, Godden, DJ & Brown, KM (1994) Increase in asthma: a more toxic environment or a more susceptible population? Thorax 49, 171174.CrossRefGoogle ScholarPubMed
16.Rayman, MP (1997) Dietary selenium: time to act. Br Med J 314, 387388.CrossRefGoogle ScholarPubMed
17.Shortt, CT, Duthie, GG, Robertson, JD et al. (1997) Selenium status of a group of Scottish adults. Eur J Clin Nutr 51, 400404.CrossRefGoogle ScholarPubMed
18.Department of Health (1991) Dietary Reference Values for Food Energy and Nutrients for the United Kingdom. Report on Health and Social Subjects no. 41. London: H. M. Stationery Office.Google Scholar
19.Department of Environmental Farming and Rural Affairs (2005) National Food Survey. Trends in household nutrient intake. http://statistics.defra.gov.uk/esg/publications/efs/2005/chapter5.pdf (accessed April 2009).Google Scholar
20.Henderson, L, Irving, K, Gregory, J et al. (2003) The National Diet and Nutrition Survey: Adults Aged 19 to 64 Years. London: The Stationery Office; available at. http://www.food.gov.uk/multimedia/pdfs/ndnsv3.pdf (accessed March 2009).Google Scholar
21.Black, PN & Sharpe, S (1997) Dietary fat and asthma: is there a connection? Eur Respir J 10, 6–12.CrossRefGoogle ScholarPubMed
22.Harizi, H, Juzan, M, Moreau, JF et al. (1997) Prostaglandins inhibit 5-lipoxygenase-activating protein expression and leukotriene B4 production from dendritic cells via an IL-10 dependent mechanism. J Immunol 159, 2835.Google Scholar
23.Calder, PC, Yaqoob, P, Thies, F et al. (2002) Fatty acids and lymphocyte functions. Br J Nutr 87, Suppl. 1, S31S48.CrossRefGoogle ScholarPubMed
24.Sanderson, P, MacPherson, GG, Jenkins, CH et al. (1997) Dietary fish oil diminishes the antigen presentation activity of rat dendritic cells. J Leukocyte Biol 62, 771777.Google Scholar
25.Wjst, M & Dold, S (1999) Genes, factor X, and allergens: what causes allergic diseases? Allergy 54, 757759.CrossRefGoogle ScholarPubMed
26.Lemire, JM, Archer, DC, Beck, L et al. (1995) Immunosuppressive actions of 1,25-dihydroxy vitamin D3: preferential inhibition of Th1 functions. J Nutr 125, 1704S1708S.Google Scholar
27.Jirapongsananuruk, O, Melamed, I & Leung, DY (2000) Additive immunosuppressive effects of 1,25-dihydroxyvitamin D3 and corticosteroids on TH1, but not TH2, responses. J Allergy Clin Immunol 106, 981985.Google Scholar
28.Litonjua, AA & Weiss, ST (2007) Is vitamin D deficiency to blame for the asthma epidemic? J Allergy Clin Immunol 120, 10311035.Google Scholar
29.Xystrakis, E, Kusumakar, S, Boswell, S et al. (2006) Reversing the defective induction of IL-10-secreting regulatory T cells in glucocorticoid-resistant asthma patients. J Clin Invest 116, 146155.CrossRefGoogle ScholarPubMed
30.Penna, G, Roncari, A, Amuchastegui, S et al. (2005) Expression of the inhibitory receptor ILT3 on dendritic cells is dispensable for induction of CD4+Foxp3+ regulatory T cells by 1,25-dihydroxyvitamin D3. Blood 106, 34903497.CrossRefGoogle Scholar
31.Tricon, S, Willers, S, Smit, HA et al. (2006) Nutrition and allergic disease. Clin Exp Allergy Rev 6, 117188.CrossRefGoogle Scholar
32.Katsoulis, K, Kontakiotis, T, Leonardopoulos, I et al. (2003) Serum total antioxidant status in severe exacerbation of asthma: correlation with the severity of the disease. J Asthma 40, 847854.CrossRefGoogle ScholarPubMed
33.Troisi, RJ, Willett, WC, Weiss, ST et al. (1995) A prospective study of diet and adult-onset asthma. Am J Respir Crit Care Med 151, 14011408.Google Scholar
34.Hijazi, N, Abalkhail, B & Seaton, A (2000) Diet and childhood asthma in a society in transition: a study in urban and rural Saudi Arabia. Thorax 55, 775779.Google Scholar
35.Hu, G & Cassano, P (2000) Antioxidants and pulmonary function: the third National Health and Nutrition Examination Survey (NHANES III). Am J Epidemiol 151, 975981.Google Scholar
36.Huang, S-L & Pan, W-H (2001) Dietary fats and asthma in teenagers: analyses of the first Nutrition and Health Survey in Taiwan (NAHSIT). Clin Exp Allergy 31, 18751880.CrossRefGoogle ScholarPubMed
37.Harik-Khan, RI, Muller, DC & Wise, RA (2004) Serum vitamin levels and the risk of asthma in children. Am J Epidemiol 159, 351357.CrossRefGoogle ScholarPubMed
38.Rubin, RN, Navon, L & Cassano, PA (2004) Relationship of serum antioxidants to asthma prevalence in youth. Am J Respir Crit Care Med 169, 393398.CrossRefGoogle ScholarPubMed
39.Riccioni, G, Bucciarelli, T, Mancini, B et al. (2007) Plasma lycopene and antioxidant vitamins in asthma: the PLAVA Study. J Asthma 44, 429432.Google Scholar
40.Wood, LG, Garg, ML, Blake, RJ et al. (2005) Airway and circulating levels of carotenoids in asthma and healthy controls. J Am Coll Nutr 24, 448455.Google Scholar
41.Shaheen, SO, Sterne, JAC, Thompson, RL et al. (2001) Dietary antioxidants and asthma in adults population-based case-control study. Am J Respir Crit Care Med 164, 18231828.CrossRefGoogle ScholarPubMed
42.Omland, Ø, Deguchi, Y, Sigsgaard, T et al. (2002) Selenium serum and urine is associated with mild asthma and atopy. The SUS study. J Trace Elem Med Biol 16, 123127.CrossRefGoogle ScholarPubMed
43.Qujeq, D, Hidari, B, Bijani, K et al. (2003) Glutathione peroxidase activity and serum selenium concentration in intrinsic asthmatic patients. Clin Chem Lab Med 41, 200202.CrossRefGoogle ScholarPubMed
44.Kocyigit, A, Armutcu, F, Gurel, A et al. (2004) Alterations in plasma essential trace elements selenium, manganese, zinc, copper, and iron concentrations and the possible role of these elements on oxidative status in patients with childhood asthma. Biol Trace Elem Res 97, 3141.Google Scholar
45.Okoko, BJ, Burney, PG, Newson, RB et al. (2007) Childhood asthma and fruit consumption. Eur Respir J 29, 11611168.Google Scholar
46.Chatzi, L, Torrent, M, Romieu, I et al. (2007) Diet, wheeze, and atopy in school children in Menorca, Spain. Pediatric Allergy Immunol 18, 480485.CrossRefGoogle ScholarPubMed
47.Chatzi, L, Apostolaki, G, Bibakis, I et al. (2007) Protective effect of fruits, vegetables and the Mediterranean diet on asthma and allergies among children in Crete. Thorax 62, 677683.Google Scholar
48.Forastière, F, Pistelli, R, Sestini, P et al. (2000) Consumption of fresh fruit rich in vitamin C and wheezing symptoms in children. Thorax 55, 283288.Google Scholar
49.De Batlle, J, Garcia-Aymerich, J, Barraza-Villarreal, A et al. (2008) Mediterranean diet is associated with reduced asthma and rhinitis in Mexican children. Allergy 63, 13101316.CrossRefGoogle ScholarPubMed
50.Kaur, B, Rowe, BH & Arnold, E (2009) Vitamin C supplementation for asthma. Cochrane Database of Systematic Reviews, issue 1, CD000993. doi:10.1002/14651858.CD000993.pub3. http://mrw.interscience.wiley.com/cochrane/clsysrev/articles/CD000993/frame.htmlGoogle Scholar
51.Shaheen, SO, Newson, RB, Rayman, MP et al. (2007) Randomised, double blind, placebo-controlled trial of selenium supplementation in adult asthma. Thorax 62, 483490.CrossRefGoogle ScholarPubMed
52.Pearson, PJK, Lewis, SA, Britton, J et al. (2004) Vitamin E supplements in asthma: a parallel group randomised placebo controlled trial. Thorax 59, 652656.CrossRefGoogle ScholarPubMed
53.Laerum, BN, Wentzel-Larsen, T, Gulsvik, A et al. (2007) Relationship of fish and cod oil intake with adult asthma. Clin Exp Allergy 37, 16161623.Google Scholar
54.Miyamoto, S, Miyake, Y, Sasaki, S et al. (2007) Fat and fish intake and asthma in Japanese women: baseline data from the Osaka Maternal and Child Health Study. Int J Tuberc Lung Dis 11, 103109.Google ScholarPubMed
55.Hodge, L, Salome, CM, Peat, JK et al. (1996) Consumption of oily fish and childhood asthma risk. Med J Aust 164, 137140.Google Scholar
56.Kim, JL, Elfman, L, Mi, Y et al. (2005) Current asthma and respiratory symptoms among pupils in relation to dietary factors and allergens in the school environment. Indoor Air 15, 170182.CrossRefGoogle ScholarPubMed
57.Takemura, Y, Sakurai, Y, Honjo, S et al. (2002) The relationship between fish intake and the prevalence of asthma: The Tokorozawa Childhood asthma and Pollinosis Study. Prev Med 34, 221225.CrossRefGoogle ScholarPubMed
58.Bolte, G, Frye, C, Hoelscher, B et al. (2001) Margarine consumption and allergy in children. Am J Respir Crit Care Med 163, 277279.CrossRefGoogle ScholarPubMed
59.Bolte, G, Winkler, G, Holscher, B et al. (2005) Margarine consumption, asthma, and allergy in young adults: Results of the German National Health Survey 1998. Ann Epidemiol 15, 207213.Google Scholar
60.Nagel, G & Linseisen, J (2005) Dietary intake of fatty acids, antioxidants and selected food groups and asthma in adults. Eur J Clin Nutr 59, 8–15.Google Scholar
61.Foliaki, S, Annesi-Maesano, I, Tuuau-Potoi, N et al. (2008) Risk factors for symptoms of childhood asthma, allergic rhinoconjunctivitis and eczema in the Pacific: an ISAAC Phase III study. Int J Tuberc Lung Dis 12, 799806.Google ScholarPubMed
62.Haby, MM, Peat, JK, Marks, GB et al. (2001) Asthma in pre-school children: prevalence and risk factors. Thorax 56, 589595.CrossRefGoogle ScholarPubMed
63.Miyake, Y, Sasaki, S, Arakawa, M et al. (2008) Fatty acid intake and asthma symptoms in Japanese children: The Ryukyus Child Health Study. Clin Exp Allergy 38, 16441650.CrossRefGoogle ScholarPubMed
64.Oddy, WH, de Klerk, NH, Kendall, GE et al. (2005) Ratio of omega-6 to omega-3 fatty acids and childhood asthma. J Asthma 41, 319326.CrossRefGoogle Scholar
65.Bolte, G, Kompauer, I, Fobker, M et al. (2006) Fatty acids in serum cholesterol esters in relation to asthma and lung function in children. Clin Exp Allergy 36, 293302.Google Scholar
66.Woods, RK, Raven, JM, Walters, EH et al. (2004) Fatty acid levels and risk of asthma in young adults. Thorax 59, 105110.CrossRefGoogle ScholarPubMed
67.Thien, FCK, De Luca, S, Woods, R et al. (2002) Dietary marine fatty acids (fish oil) for asthma in adults and children. Cochrane Database of Systematic Reviews, issue 2, CD001283. doi: 10.1002/14651858.CD001283. http://www.mrw.interscience.wiley.com/cochrane/clsysrev/articles/CD001283/frame.htmlGoogle Scholar
68.Mickleborough, TD, Murray, RL, Ionescu, AA et al. (2003) Fish oil supplementation reduces severity of exercise-induced bronchoconstriction in elite athletes. Am J Respir Crit Care Med 168, 11811189.Google Scholar
69.Mickleborough, TD, Lindley, MR, Ionescu, AA et al. (2006) Protective effect of fish oil supplementation. Chest 129, 3949.CrossRefGoogle ScholarPubMed
70.Schubert, R, Kitz, R, Beerman, C et al. (2009) Effect of n-3 polyunsaturated fatty acids in asthma after low dose allergen challenge. Int Arch Allergy Immunol 148, 321329.CrossRefGoogle ScholarPubMed
71.Moreira, A, Moreira, P, Delgado, L et al. (2007) Pilot study of the effects of n-3 polyunsaturated fatty acids on exhaled nitric oxide in patients with stable asthma. J Investig Allergol Clin Immunol 17, 309313.Google Scholar
72.Biltagi, MA, Baset, AA, Bassiouny, M et al. (2009) Omega-3 fatty acids, vitamin C and Zn supplementation in asthmatic children: a randomized self-controlled study. Acta Paediatr 98, 737742.Google Scholar
73.Holt, PG, Devereux, G & Sly, P (2006) Early life origins of allergy and asthma. In Allergy, 3rd ed., pp. 223232 [Holgate, ST, Church, MK and Lichenstein, LM, editors]. Philadelphia, PA: Mosby Elsevier.Google Scholar
74.Håland, G, Lødrup Carlsen, KC, Sandvik, L et al. (2006) Reduced lung function at birth and the risk of asthma at 10 years of age. N Eng J Med 355, 16821689.Google Scholar
75.Katz, KA, Pocock, SJ & Strachan, DP (2003) Neonatal head circumference, neonatal weight, and risk of hayfever, asthma and eczema in a large cohort of adolescents from Sheffield, England. Clin Exp Allergy 33, 737745.CrossRefGoogle Scholar
76.Gilliland, FD, Li, YF & Peters, JM (2001) Effects of maternal smoking during pregnancy and environmental tobacco smoke on asthma and wheezing in children. Am J Respir Crit Care Med 163, 429436.CrossRefGoogle ScholarPubMed
77.Devereux, G & Barker, R (2002) Studies of cord blood mononuclear cell responses and allergy: still in their infancy? Clin Exp Allergy 32, 331334.CrossRefGoogle ScholarPubMed
78.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
79.Devereux, G & Seaton, A (2001) Why don't we give chest patients dietary advice? Thorax 56, Suppl. 2, S15S22.Google ScholarPubMed
80.Lynch, JW, Kaplan, GA & Salonen, JT (1997) Why do poor people behave poorly? Variation in adult health behaviours and psychosocial characteristics by stages of the socioeconomic lifecourse. Soc Sci Med 44, 809819.CrossRefGoogle ScholarPubMed
81.Talvia, S, Rasanen, L, Lagstrom, H et al. (2006) Longitudinal trends in consumption of vegetables and fruit in Finnish children in an atherosclerosis prevention study(STRIP). Eur J Clin Nutr 60, 172180.Google Scholar
82.Stick, S (2000) The contribution of airway development to paediatric and adult lung disease. Thorax 55, 587594.Google Scholar
83.Turner, SW, Palmer, LJ, Rye, PJ et al. (2004) The relationship between infant airway function childhood airway responsiveness, and asthma. Am J Respir Crit Care Med 169, 921927.Google Scholar
84.Prescott, SL & Jones, CA (2001) Cord blood memory responses: are we being naive? Clin Exp Allergy 31, 16531656.Google Scholar
85.Salem, MT, Li, YF, Langholz, B et al. (2005) Maternal fish consumption during pregnancy and risk of early childhood asthma. J Asthma 42, 513518.CrossRefGoogle Scholar
86.Wijga, AH, Smit, HA, Kerkhof, M et al. (2003) Association of consumption of products containing milk fat with reduced asthma risk in pre-school children: the PIAMA birth cohort study. Thorax 58, 567572.Google Scholar
87.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.Google Scholar
88.Gold, DR, Willwerth, BM, Tantisira, KG et al. (2006) Associations of cord blood fatty acids with lymphocyte proliferation, IL-13 and IFN-γ. J Allergy Clin Immunol 117, 931938.Google Scholar
89.Wallace, FA, Miles, EA, Evans, C et al. (2001) Dietary fatty acids influence the production of Th1- but not Th2-type cytokines. J Leukocyte Biol 69, 449457.CrossRefGoogle Scholar
90.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.CrossRefGoogle ScholarPubMed
91.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.CrossRefGoogle ScholarPubMed
92.Marks, GB, Mihrshahi, S, Kemp, AS et al. (2006) Prevention of asthma during the first 5 years of life: A randomized controlled trial. J Allergy Clin Immunol 118, 5361.Google Scholar
93.Shaheen, SO, Newson, RB, Henderson, AJ et al. (2004) Umbilical cord trace elements and minerals and risk of early childhood wheezing and eczema. Eur Respir J 24, 292297.CrossRefGoogle ScholarPubMed
94.Devereux, G, McNeill, G, Newman, G et al. (2007) Early childhood wheezing symptoms in relation to plasma selenium in pregnant mothers and neonates. Clin Exp Allergy 37, 10001008.Google Scholar
95.Litonjua, AA, Tantisira, KG, Finn, PW et al. (2004) Maternal antioxidant intake during pregnancy and cord blood lymphoproliferative responses. Am J Resp Crit Care Med 169, A501.Google Scholar
96.Litonjua, AA, Rifas-Shiman, S, Ly, NP et al. (2006) Maternal antioxidant intake in pregnancy and wheezing illnesses in children at 2 y of age. Am J Clinical Nutr 84, 903911.CrossRefGoogle ScholarPubMed
97.Devereux, G, Barker, RN & Seaton, A (2002) Ante-natal determinants of neonatal immune responses to allergens. Clin Exp Allergy 32, 4350.Google Scholar
98.Devereux, G, Turner, SW, Craig, LCA et al. (2006) Reduced 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
99.Willers, SM, Devereux, G, Craig, LCA et al. (2007) Maternal food consumption during pregnancy and asthma, respiratory and atopic symptoms in 5-year-old children. Thorax 62, 772778.CrossRefGoogle Scholar
100.Fitzsimon, N, Fallon, UB, O'Mahony, D et al. (2007) Mothers' dietary patterns during pregnancy and the risk of asthma symptoms in children at 3 years. Irish Med J 100, 2732.Google ScholarPubMed
101.Chatzi, L, Torrent, M, Romieu, I et al. (2008) Mediterranean diet in pregnancy is protective for wheeze and atopy in childhood. Thorax 63, 507513.CrossRefGoogle ScholarPubMed
102.Azzi, A, Gysin, R, Kempna, P et al. (2003) The role of α-tocopherol in preventing disease: from epidemiology to molecular events. Molec Aspects Dis 24, 325336.CrossRefGoogle ScholarPubMed
103.Reiter, E, Jiang, Q & Christen, S (2007) Anti-inflammatory properties of α- and γ-tocopherol. Mol Aspects Med 28, 668691.CrossRefGoogle ScholarPubMed
104.Manthey, JA (2000) Biological properties of flavonoids pertaining to inflammation. Microcirculation 7, S29S34.Google Scholar
105.Okamoto, N, Murata, T, Tamai, H et al. (2006) Effects of alpha tocopherol and probucol supplements on allergen-induced airway inflammation and hyperresponsiveness in a mouse model of allergic asthma. Int Arch Allergy Immunol 141, 172180.Google Scholar
106.Li-Weber, M, Giasisi, M, Trieber, MK et al. (2002) Vitamin E inhibits IL-4 gene expression in peripheral blood T-cells. Eur J Immunol 32, 24012408.3.0.CO;2-S>CrossRefGoogle ScholarPubMed
107.Malmberg, KJ, Lenkei, R, Petersson, M et al. (2002) A short-term dietary supplementation of high doses of vitamin E increases T helper 1 cytokine production in patients with advanced colorectal cancer. Clin Cancer Res 8, 17721778.Google ScholarPubMed
108.Wang, Y, Huang, DS, Eskelson, CD et al. (1994) Long term dietary vitamin E retards development of retrovirus induced disregulation in cytokine production. Clin Immunol 72, 7075.Google Scholar
109.Meydani, SN, Han, SN & Wu, D (2005) Vitamin E and immune response in the aged: molecular mechanisms and clinical implications. Imm Rev 205, 269281.Google Scholar
110.Tan, PH, Sagoo, P, Chan, C et al. (2005) Inhibition of NF-κB and oxidative pathways in human dendritic cells by antioxidative vitamins generates regulatory T cells. J Immunol 174, 76337644.Google Scholar
111.Wakikawa, A, Utsuyama, M, Wakabayashi, A et al. (1999) Vitamin E enhances the immune functions of young but not old mice under restraint stress. Exp Gerontology 34, 853862.CrossRefGoogle Scholar
112.Xagorari, A, Papapetropoulos, A, Mauromatis, A et al. (2001) Luteolin inhibits endotoxin stimulated phosphorylation cascade and proinflammatory cytokine production in macrophages. J Pharm Exp Therap 296, 181187.Google Scholar
113.Rogerio, AP, Kanashiro, A, Fontanari, C et al. (2007) Anti-inflammatory activity of quercetin and isoquercitrin in experimental murine allergic asthma. Inflamm Res 56, 402408.Google Scholar
114.Nair, MP, Kandaswami, C, Mahajan, S et al. (2002) The flavonoid, quercetin, differentially regulates Th-1(IFNgamma) and Th-2 (IL-4) cytokine gene expression by normal peripheral blood mononuclear cells. Biochim Biophys Acta 1593, 2936.Google Scholar
115.Islam, S, Narra, V, Cote, GM et al. (1999) Prenatal vitamin E treatment improves lung growth in fetal rats with congenital diaphragmatic hernia. J Ped Surg 34, 172176.CrossRefGoogle ScholarPubMed
116.Azzi, A, Gysin, R, Kempna, P et al. (2004) Vitamin E mediates cell signaling and regulation of gene expression. Ann N Y Acad Sci 1031, 8695.CrossRefGoogle ScholarPubMed
117.Ricciarelli, R, Maroni, P, Ozer, N et al. (1999) Age-dependent increase of collagenase expression can be reduced by alpha-tocopherol via protein kinase C inhibition. Free Rad Biol Med 27, 729737.Google Scholar
118.Camargo, CA, Rifas-Shiman, SL, Litonjua, AA et al. (2007) Maternal intake of vitamin D during pregnancy and reports of recurrent wheezing in children at 3 y of age. Am J Clin Nutr 85, 788795.CrossRefGoogle Scholar
119.Devereux, G, Litonjua, AA, Turner, SW et al. (2007) Maternal vitamin D intake during pregnancy and early childhood wheeze. Am J Clin Nutr 85, 853859.CrossRefGoogle Scholar
120.Hypponen, E, Sovio, U, Wjst, M et al. (2004) Infant vitamin D supplementation and allergic conditions in adulthood: northern Finland birth cohort 1966. Ann N Y Acad Sci 1037, 8495.Google Scholar
121.Gale, CR, Robinson, SM, Harvey, NC et al. (2008) Maternal vitamin D status during pregnancy and childhood outcomes. Eur J Clin Nutr 62, 6877.CrossRefGoogle Scholar
122.MRC Vitamin Study Research Group (1991) Prevention of neural tube defects: results of the Medical Research Council Vitamin Study. Lancet 338, 131137.Google Scholar