Hostname: page-component-8448b6f56d-qsmjn Total loading time: 0 Render date: 2024-04-16T08:23:46.088Z Has data issue: false hasContentIssue false

Longitudinal follow-up of the relationship between dietary intake and growth and development in the Lifeways cross-generation cohort study 2001–2013

Published online by Cambridge University Press:  03 December 2013

Cecily C. Kelleher*
Affiliation:
School of Public Health, Physiotherapy and Population Science, University College Dublin, Woodview House, Belfield, Dublin 4, Republic of Ireland
Karien Viljoen
Affiliation:
School of Public Health, Physiotherapy and Population Science, University College Dublin, Woodview House, Belfield, Dublin 4, Republic of Ireland
Hala Khalil
Affiliation:
School of Public Health, Physiotherapy and Population Science, University College Dublin, Woodview House, Belfield, Dublin 4, Republic of Ireland
Rebecca Somerville
Affiliation:
School of Public Health, Physiotherapy and Population Science, University College Dublin, Woodview House, Belfield, Dublin 4, Republic of Ireland
John O'Brien
Affiliation:
School of Public Health, Physiotherapy and Population Science, University College Dublin, Woodview House, Belfield, Dublin 4, Republic of Ireland
Aakash Shrivastava
Affiliation:
School of Public Health, Physiotherapy and Population Science, University College Dublin, Woodview House, Belfield, Dublin 4, Republic of Ireland
Celine Murrin
Affiliation:
School of Public Health, Physiotherapy and Population Science, University College Dublin, Woodview House, Belfield, Dublin 4, Republic of Ireland
*
*Corresponding author: Professor C. C. Kelleher, fax +353 1 7163421, email Cecily.kelleher@ucd.ie
Rights & Permissions [Opens in a new window]

Abstract

In this paper we will review evidence on the early life and familial influences on childhood growth and development, with particular reference to the Lifeways cross-generation cohort study in the Republic of Ireland. The Lifeways cross-generation cohort study was established in 2001–2013 through two maternity hospitals in the Republic of Ireland and was one of many new cohort studies established worldwide in the millennium period. Mothers were recruited at first booking visit, completing a self-administered questionnaire, which included a 147 item semi-quantitative FFQ. Longitudinal follow-up is ongoing in 2013, with linkage data to hospital and general practice records and examination of children when aged 5 and 9 years. The study is one of very few containing data on grandparents of both lineages with at least one grandparent recruited at baseline. There have been consistent associations between parental and grandparental health status characteristics and children's outcomes, including infant birth-weight, BMI when child was aged 5 years and childhood wheeze or asthma when child was aged 3 and aged 5 years. In conclusion, empirical evidence to date shows consistent familial and cross-generational patterns, particularly in the maternal line.

Type
Conference on ‘Childhood Nutrition and Obesity: Current Status and Future Challenges’
Copyright
Copyright © The Authors 2013 

As the global obesity epidemic continues to pose a major public health challenge, it has become increasingly important to understand how risk for adult chronic disease is transmitted across generations and through the life course of individuals( Reference Barker, Winter and Osmond 1 Reference Gray, Davey Smith and McConnachie 11 ). In the present paper, we will review briefly recent evidence, with particular reference to the Lifeways cross-generation cohort study in the Republic of Ireland. This linkage cohort study was one of several established globally in the millennium period( Reference Larsen, Kamper-Jørgensen and Adamson 12 ) and has followed index children since recruitment during pregnancy in 2001–2013 and also their mothers, fathers and at least one grandparent. The design and data management considerations have been described previously and the original mothers were found to be comparable in demographic terms with women of the same age in the contemporary Survey of Lifestyles, Attitudes and Nutrition (SLAN)( Reference O'Mahony, Fallon and Hannon 13 , Reference Kelleher, Bury and Murphy 14 ). The study objectives were to document health status, diet and lifestyle in the family members and to establish patterns and links across generations. The study continues to follow participants up to 2013 and the data collected in each sweep are summarised in Table 1.

Table 1. Summary of data collection sweeps to date in the Lifeways cross-generation cohort study

MGM, maternal grandmother; MGF, maternal grandfather; PGM, paternal grandmother; PGF, paternal grandfather; GP, general practice.

In the initial 5 years follow-up, primary care utilisation patterns were documented and the study examined how indicators of social position, particularly means-tested eligibility to free healthcare, influenced health status of family members. In the Republic of Ireland, there is a two-tiered means-tested health care system, and access to free primary and community care including general practice is available to those below a certain income level, so-called general medical services (GMS) eligibility. This has been found to be a robust indicator of both social disadvantage and various health outcomes( Reference Fallon, Murphy and Majawit 15 ).

A life-course approach to diet, growth and development

In social epidemiology, a comprehensive account of influences across the life course must be undertaken in order to understand why outcomes such as CVD, cancers or diabetes might develop in adulthood( Reference Kawachi and Berkman 16 ). Such an approach takes account of the immediate proximal pathophysiological processes leading to a specific disease outcome, and more recently how genetic, genomic or nutrigenomic factors acting at different time points might be affecting the outcome in question, which we discuss later ( 4 , Reference Chmurznska 17 Reference Yaghootkar and Freathy 24 ). However, there are also contextual factors to be considered, such as individual lifestyle choices or health behaviours predisposing to risk factor development, and in turn how so-called meso-level situations such as the work environment, occupation or social class determine those health choices at different life stages, all of these being underpinned by the wider sociopolitical structure( Reference Kawachi and Berkman 16 ). This level of understanding is required if changes are to be wrought by public policy strategies. Study designs to take account of these factors may be longitudinal but also have design challenges to record what is measurable at the level of the individual cohort member and how best to record and examine ecological level factors such as a region or area, or group characteristics such as family membership.

The seminal influence of Barker( Reference Barker, Winter and Osmond 1 , Reference Barker, Gluckman and Godfrey 3 , Reference Barker, Larsen and Osmond 5 , Reference Barker 6 ) in reframing our understanding of early life influences on adult chronic disease has created a paradigm shift in our understanding of these processes over the past three decades. In the original Hertfordshire retrospective cohort study, where current health outcomes were linked to original infant anthropometric records, Barker et al. showed the graduated inverse relationship between both birth-weight and weight in infancy and death for CHD at all ages in 10 636 men( Reference Barker, Winter and Osmond 1 ). As they pointed out, there are various reasons for babies to be small at birth and in infancy, including low birth-weight, short stature, placental insufficiencies and failure to thrive( Reference Barker, Gluckman and Godfrey 3 ). Since then many studies have considered cumulative, trajectory and critical period influences on longer term outcomes. It is now well understood that there are two processes at the extremes of birth-weight, whereby larger babies are at greater risk of subsequent diabetes and obesity, in turn related to maternal weight and height( Reference Cnattingus, Villamor and Lagerros 8 , Reference Poston 25 ) and lower birth-weight babies subjected to degrees of intrauterine growth retardation are also susceptible to later adult chronic disease( Reference Barker, Winter and Osmond 1 , Reference Barker, Larsen and Osmond 5 , Reference Adair, Fall and Osmond 7 , Reference Eriksson, Kajantie and Thornburg 10 ). The developmental plasticity hypothesis, whereby there is a critical period during which a system is plastic, followed by loss of plasticity and a fixed functional capacity, is now well accepted as a biological phenomenon, and pregnancy, during which the intrauterine development of all children occurs, is one such critical period shared by all individuals( Reference Barker, Larsen and Osmond 5 , Reference Eriksson, Kajantie and Thornburg 10 , Reference Gray, Davey Smith and McConnachie 11 ).

Barker has since extended this concept to define what he calls 100 years of nutritional flow( Reference Barker 6 ). The 1000 d from conception to toddlerhood not only include maternal influences such as her nutritional supply to the fetus during pregnancy and early childhood feeding practices, but also the quality, size and shape of the placenta are increasingly understood to play a role in successful fetal and early childhood growth and development milestones and subsequent long-term health outcomes. The maternal grandmother is especially influential as it is she who makes the grandchild's egg for subsequent fertilisation by the ovulating mother. In this sense, the developmental investment in the index child dates across generations and an ageing adult owes his health status to his own constitutional and environmental exposures but also to that of his parents and grandparents. There is also the genetic influence of both parents and all grandparents, the X-linked effects of the mother, the Y-linked effects of the paternal line, and notably through the maternal line, the mitochondrial DNA transmission( Reference Pembrey, Bygren and Edvinsson 26 , Reference Fox, Sear and Beise 27 ). Recent studies have shown an association between variants in the ADCY5 gene and birth-weight and up to seven loci identified which account for a similar proportion of the variance in infant birth-weight as smoking( Reference Horikoshi, Yaghootkar and Mook-Kanamori 19 ). This may predispose individuals both to relatively lower birth-weight and a propensity to adult chronic disease.

Cross-generational evidence

In recent years, a number of studies have been published showing the relationship between infant birth-weight and subsequent maternal and paternal morbidity and mortality( Reference Andersen and Osler 28 Reference Wannamethee, Lawlor and Whincup 31 ). Generally such an association has been shown for lower birth-weight, particularly for mothers. Many studies have attempted to examine how children's growth trajectories relate to their parents’ anthropometric characteristics( Reference Durmus, Arends and Ay 9 , Reference Gray, Davey Smith and McConnachie 11 ). Prospects for understanding the genetic, epigenetic or nutrigenomic mechanisms have recently been reviewed( Reference Relton and Davey Smith 23 ). These authors define epigenetics as the regulatory processes that control the transcription of information encoded in the DNA sequence into RNA before their transcription into proteins. The tissue specificity of epigenetic patterns is a well-established phenomenon and not all studies report expected associations( Reference Ivanova, Chen and Segonds-Pichon 21 ), even though in principle nutrigenomic effects may be important as for instance DNA methylation which occurs in utero depends on the availability of several nutrients including methionine, vitamins B6, B12 and folate( Reference Chmurznska 17 ).

A further use of genetic markers is in so-called Mendelian randomisation, whereby an individual receives at random a specific allele from its parent, this process being unlikely to be confounded by any subsequent behavioural characteristic of the individual. It is then possible to adjust for the known influence of this marker on an apparent prospective association between a risk factor and the disease-specific outcome of interest, using genotype as an instrumental variable. This has been shown for the FTO gene in Danish adults in relation to the association of obesity with IHD( Reference Palmer, Nordestgarrard and Benn 32 ) and to attenuate greatly the apparently strong association between maternal BMI and offspring adiposity( Reference Lawlor, Timpson and Harbord 33 ). These studies have attempted to address biased or confounded associations in that parents may share genetic or environmental characteristics, but not the direct health behaviours of their offspring( Reference Bonilla, Lawlor and Ben-Shlomo 34 ).

Several animal studies have provided evidence for transgenerational effects of intrauterine exposure( Reference Harrison and Langley-Evans 18 , Reference Ponzio, Carvalho and Fortes 22 , Reference Zambrano, Martinez-Samayoa and Bautista 35 , Reference Torrens, Poston and Hanson 36 ). Female rats, exposed to protein restriction during pregnancy and lactation, were found to have offspring that delivered a progeny with altered growth and metabolism( Reference Zambrano, Martinez-Samayoa and Bautista 35 ). A similar protein restriction trial in pregnant rats reported that second-generation offspring had raised blood pressure and endothelial dysfunction which was passed through the maternal line( Reference Torrens, Poston and Hanson 36 ). In contrast, Harrison and Langley-Evans( Reference Harrison and Langley-Evans 18 ) reported that protein restriction during pregnancy results in a phenotype of raised blood pressure and reduced nephron number passed through maternal and paternal lines. More recently, Ponzio et al.( Reference Ponzio, Carvalho and Fortes 22 ) have demonstrated that phenotypes resulting from maternal protein restriction are evident in a second and third generation of animals.

Studies of body composition and diet in human subjects have either been limited to one generation or have used proxy markers for diet intake across one or more generations, such as those of the Overkalix studies( Reference Pembrey, Bygren and Edvinsson 26 , Reference Kaati, Bygren and Pembrey 37 ) and the Dutch Winter Hunger study( Reference Heijmans, Tobi and Stein 20 ). By contrast with parental data studies( Reference Andersen and Osler 28 Reference Wannamethee, Lawlor and Whincup 31 , Reference Keane, Layte and Harrington 38 , Reference Davey Smith, Steer and Leary 39 ) there are relatively fewer data on how grandparental characteristics affect their grandchildren's growth and development; all such studies have either been retrospective or data linkage in design and none to date had dietary intake data across generations. To date there are just four well-characterised human studies with three generations( Reference Manor and Koupil 40 Reference Naess, Stoltenberg and Hoff 43 ). Manor and Koupil( Reference Manor and Koupil 40 ) showed a U-shaped association between grandchild's birth-weight and circulatory disease mortality in maternal grandmothers only for infants born prior to 1977. There was a U-shaped association for maternal grandfathers’ overall and circulatory disease mortality and for paternal grandfathers there was an inverse association between grandchild's birth-weight and overall mortality. McCarron et al. ( Reference McCarron, Davey Smith and Hattersley 41 ) found that the grandchildren of maternal grandparents with reported type 2 diabetes were more likely to be in the top tertile of birth-weight. There was evidence for an inverted U-shaped association between birth-weight of grandchildren and diabetes in paternal grandmothers. Smith et al. ( Reference Smith, Wood and White 42 ) have shown an inverse relationship between maternal grandparents’ risk of IHD or cerebrovascular disease and infant birth-weight and did not have data on paternal lineages. In a very large linkage study in Norway, inverse relationships were shown according to both maternal and paternal lineages and cardiovascular outcomes in grandparents; adjusting for maternal smoking accounted for much of the effect on cardiovascular mortality. For grandparental diabetes mortality, U-shaped associations were seen with grandchild birth-weight for the maternal grandmother and inverse associations for all other grandparents( Reference Naess, Stoltenberg and Hoff 43 ).

Prospective studies with information on diet and parental BMI

In an exploration of the literature, we attempted to identify cohort studies that included a measure of nutritional status (BMI or diet assessment) across two or three generations( Reference Alves, Correia, Barros and Azevedo 44 Reference Wickman, Kull and Pershagen 106 ). Prospective or cohort studies that include specifically information on maternal diet or BMI and that of other family members in relation to index children are summarised in Table 2. As Poston has pointed out, many of these are relatively recently established( Reference Poston 25 ). The studies vary in their objectives; several look at offspring size as the outcome of interest; others include diet and body size as factors associated with child and adult morbidities. Maternal pre or postnatal BMI is the variable most frequently reported (63 %) by these studies followed by paternal postnatal BMI (41 %; Table 2). Diet is less frequently reported with only one-third collecting information on prenatal maternal diet; prenatal paternal diet has only been collected by eight studies.

Table 2. Prospective studies with information on either diet or parental BMI, or both

* Prenatal BMI may be estimated before pregnancy or measured during pregnancy.

Cohorts of children born to mothers with gestational diabetes mellitus or previous macrosomia.

Cohorts currently recruiting or have recently completed recruitment.

The 1958 birth cohort has described the relationship between birth-weight of the grandchild and the influence of height by the grandparents( Reference Emanuel, Filakti and Alberman 57 ). Only one other study by Davis et al.( Reference Davis, McGonagle and Schoeni 51 ), has described a measure of BMI for grandparents. This study is limited, however, by the fact that grand maternal and grand paternal body composition was aggregated in the reported analysis. To our knowledge, no other studies have reported data available on either the BMI or diet of the proband child and their individual maternal and paternal grandparents. This makes the Lifeways cross-generation cohort study a highly unusual dataset in the literature, in that it contains information on health status, dietary intakes and adult chronic disease outcomes in the grandparental generation related to the health status of their grandchildren( Reference O'Mahony, Fallon and Hannon 13 , Reference Kelleher, Bury and Murphy 14 ).

Findings to date in the Lifeways study: pregnancy and early years

The Lifeways cross-generation cohort study was established in 2001–2013 through two maternity hospitals in the Republic of Ireland and was one of many new cohort studies established worldwide in the millennium period( Reference Larsen, Kamper-Jørgensen and Adamson 12 , Reference O'Mahony, Fallon and Hannon 13 , Reference Kelleher, Bury and Murphy 14 ). Mothers were recruited at first booking visit, completing a self-administered questionnaire which included a 147 item validated semi-quantitative FFQ. Longitudinal follow-up is ongoing in 2013, with linkage data to hospital and general practice records and examination of children when aged on average 5 and 9 years. The study is one of very few containing data on grandparents of both lineages, with at least one grandparent recruited at baseline. A summary of publications related to diet and cross-generation transmission to date for the Lifeways cohort study is given in Table 3 ( Reference Segonds-Pichon, Hannon and Daly 107 Reference Viljoen, Shrivastava and Murrin 121 ). These analyses in this cohort have been consistent in showing an influence on health outcomes at different time points of lifestyle factors including diet and of cross-generation and familial associations. This is arguably remarkable given that it is not a large cohort study, although characterised in some detail and with linkage to health records.

Table 3. Cross-generational or longitudinal Influences on health outcomes in the Lifeways cohort study 2001–2013

GMS, General medical services.

We have previously shown a social gradient in relation to baseline dietary intake at recruitment for all adult family members, in that those eligible for GMS had lower intakes of vitamin C and higher total fat intakes, having adjusted for other risk factors( Reference Kelleher, Lotya and O'Hara 110 ). Both age and cohort group were important factors, as might be expected, since the parental and grandparental dietary patterns might be likely to differ and sex differences are also apparent, with higher mean energy intakes in males compared with females. Social gradients at the level of food groups have been reported in the Surveys of Lifestyles, Attitudes and Nutrition at each of the three time points 1998, 2002 and 2007( Reference Kelleher, Friel and Nolan 122 , Reference Niedhammer, Kerrad and Schütte 123 ), so the Lifeways study corroborates that observation at the level of selected nutrient intakes.

The pregnancy outcomes were also examined in relation to indicators of maternal social position and health status( Reference Segonds-Pichon, Hannon and Daly 107 Reference Murrin, Fallon and Hannon 109 ). Determinants of mothers’ self-rated health during pregnancy showed a strongly socially graduated pattern and mothers whose own parents were educated to third level had better self-rated health than those whose parents left education early( Reference Segonds-Pichon, Hannon and Daly 107 ). Younger and less socially advantaged mothers tended to have lower birth-weight babies( Reference Murrin, Segonds-Pichon and Fallon 108 ). The dietary patterns of mothers during pregnancy were socially graduated, as was smoking status( Reference Murrin, Fallon and Hannon 109 ). Predictors of pre-term delivery and occupational predictors of pregnancy outcomes were reported in this cohort( Reference Niedhammer, O'Mahony and Daly 111 Reference Niedhammer, Murrin and O'Mahony 112 ). Mothers required to do shiftwork tended to have a higher risk of pre-term delivery, adjusted for other factors( Reference Niedhammer, O'Mahony and Daly 111 ) and this cohort was included in a subsequent meta-analysis, which showed a small effect of shiftwork on risk for this outcome( Reference Bonzini, Palmer and Coggon 124 ).

While many studies report a social gradient to pregnancy outcomes, a few attempts to comprehensively explain the degree to which known health status and lifestyle variables explain that gradient have been made. In the Lifeways study low maternal educational level was associated with an increased risk of this clinical outcome of preterm delivery (hazard ratio 2·14, 95 % CI 1·04, 4·38) and notably the explanatory combination of a material factor such as rented or crowded home, and behavioural factors such as smoking, alcohol consumption and high saturated fat intake during pregnancy reduced the hazard ratio for low education level by 42 %( Reference Niedhammer, Murrin and O'Mahony 112 ). We looked at predictors of low birth-weight of 3 kg or less both in this cohort and in another cohort of disadvantaged Traveller infants, a nomadic disadvantaged group in Ireland and found that maternal smoking and alcohol consumption were important predictors of this outcome, but when these factors were taken account of the birth-weight differential according to social class persisted. We used this cut-off point, as opposed to the clinical cut-off of 2.5 kg, as epidemiological studies show this is associated with later risk of adult chronic disease, providing empirical evidence in an Irish context for findings reported in other cohorts internationally( Reference Hamid, Fitzpatrick and Rowan 113 ).

Follow-up when children averaged age 5 years

A number of analyses have been undertaken across the generations of participating families related to the wave when children now averaged age 5 years( Reference Murrin, Kelly and Tremblay 116 Reference Murrin, Shrivastava and Kelleher 118 ). Although the responses were received for just 669 of 1126 women originally recruited and not all of these had complete examination data, there was no significant difference in baseline maternal BMI in this sweep's participants compared with that of non-participants, although these mothers were older on average than non-participants. We applied mixed effect models to take account of family relatedness, which enables correlation between family members to be modelled and also to take account of incomplete or variable numbers of family participants. Each family could contain up to seven possible members; in the child the height and weight were measured but for other adult family members the data were self-reported. We found consistent correlations for height between family members in both paternal and maternal lines, but the patterns for BMI were significant only for the maternal line. In these statistical models, education level, and also self-rated health and fruit and vegetable consumption patterns were accounted for because they each showed a social gradient and a relationship with BMI in previous analyses, with a strong significant family effect seen across the generations in the maternal line( Reference Murrin, Kelly and Tremblay 116 ).

The reported evidence in the general literature on the relative influence of paternal and maternal lines on children's BMI is mixed( Reference Durmus, Arends and Ay 9 , Reference Keane, Layte and Harrington 38 , Reference Davey Smith, Steer and Leary 39 , Reference Hui, Nelson and Yu 68 , Reference Subramanian, Ackerson and Smith 125 , Reference Kivimaki, Lawlor and Smith 126 ). The Growing Up in Ireland study found that measured parental weight status was correlated with childhood obesity at age 9 years( Reference Keane, Layte and Harrington 38 ). While the Avon Longitudinal Study of Parents and Children study for instance showed no distinction in strength of association between maternal and paternal lines( Reference Davey Smith, Steer and Leary 39 ), the Generation R cohort study also showed a strong maternal pattern, although there was a paternal influence as well( Reference Durmus, Arends and Ay 9 ).

Familial aggregation patterns in dietary intake were also assessed in the Lifeways study. Nuclear family effects were found, with the maternal association being stronger than the paternal association and the maternal grandmothers showed an association both with their own daughters’ diet and with that of their index grandchild's diet at age 5 years. The mother's current diet tended to be more strongly associated with that of her child than her pregnancy consumption, although in the case of non-breastfeeding mothers, dietary fat intake during pregnancy was more strongly associated( Reference Shrivastava, Murrin and Sweeney 117 ). Brion et al. ( Reference Brion, Ness and Rogers 46 ), employing Avon Longitudinal Study of Parents and Children data, found a similar association with regard to prenatal maternal fat intake.

Maternal dietary intake at both time points was also examined in relation to child's BMI at age 5 years. A social gradient in relation to maternal fruit and vegetable intake was found during pregnancy and also when the child averaged age 5 years. Total energy intake in mothers was greater during pregnancy. Increased odds of overweight or obesity were found in mothers with higher intakes of sugar during pregnancy and higher fat intakes when the child was aged 5 years. Mothers with persistently high intakes of SFA and those who had lowered their sugar consumption since pregnancy were more likely to have overweight or obese children and we discuss the possible biological plausibility of these findings in that paper( Reference Murrin, Shrivastava and Kelleher 118 ). Again, cohort data on this issue are rare. The Southampton women's survey( Reference Moon, Harvey and Robinson 127 ) reported that after adjustment for maternal factors including height and duration of breastfeeding maternal n-6 PUFA intake positively predicted fat mass in children aged 4 and 6 years.

Dietary patterns and risk of childhood wheezing or asthma

A specific health outcome of interest that can already be examined in the children themselves is childhood wheeze or asthma, which is strongly suspected to have a dietary basis. The Lifeways study was one of the first to show that general practitioner-reported wheeze or asthma when the child averaged 3 years was inversely associated with maternal fruit and vegetable and fish consumption during pregnancy( Reference Fitz-simon, Fallon and O'Mahony 114 ). In Table 4, we show predictors of reported asthma for children at both time points when they were aged on average 3 and 5 years, replicating the original analysis at the new time point. Various biological, lifestyle and socioeconomic determinants of both child and maternal association that were significant predictors of asthma at the uni-variate level were included as covariates in multivariate models( Reference Viljoen, Shrivastava and O'Brien 115 ). Separate logistic regression models were constructed to examine the effect of food group intake relative to asthma status at each phase of follow-up. Models were adjusted for combinations of birth-weight, sex, region, maternal age, maternal socioeconomic status (measured by education or GMS eligibility), parity, marital status and smoking in pregnancy. Oily fish intake proved to be protective of asthma at both year 3 (odds ratios (OR) = 0·52, 95 % CI 0·30, 0·92) and year 5 (OR = 0·51, 95 % CI 0·28, 0·92). Similarly, vegetable intake in the upper quartile proved protective at year 3 (OR = 0·42, 95 % CI 0·19, 0·93) and year 5 (OR = 0·43, 95 % CI 0·18, 0·99). This shows that mothers of asthmatic children tended to be younger, more disadvantaged and to have a lower oily fish, fruit and vegetable intake during pregnancy. Conversely, high intake of added or spreadable fats were related to asthma at year 3 follow-up (OR = 2·46, 95 % CI 1·34, 4·51). These data are quite consistent over the two time points and notably it is the baseline maternal dietary intake, rather than characteristics at aged 5 years, which show the main effects. The mechanisms through which this may operate are not as yet understood but may be nutrigenomic in origin and several other investigators have examined this question( Reference Devereux and Seaton 128 ). The Lifeways dataset has recently formed part of a pooled analysis as part of the CHICOS (Developing a Research strategy for Child Cohorts in Europe) consortium funded by the FP7 programme to elucidate these pathways further( Reference Larsen, Kamper-Jørgensen and Adamson 12 ); one analysis looked at the influence of maternal fish intake on birth outcomes( Reference Leventakou, Roumeliotaki and Martinez 129 ) and another on how birth outcomes might predict risk of childhood asthma( Reference Sonnenschein-van de Voort, Arends and de Jongste 130 ).

Table 4. Predictors of General Practitioner-reported wheeze (10·4 % of n 614) in children aged 3 years and of maternal reported asthma (14·3 % of n 511) in children aged 5 years in the Lifeways Cohort Study

GMS, General medical services; OR, odds ratio; prim, primary; sec, secondary; multip, multipara; nullip, nullipara.

* P < 0·05

** P = 0·05–0·09

Adjusted OR are reported for nutritional variables; all other values are from the univariate analysis.

Longitudinal follow-up when children were aged 9 years

Because these analyses to date showed such patterns of association across the generations, it was decided to follow-up adults and children again and between 2011 and 2013 further follow-up has taken place of the families. This included a short self-administered health questionnaire for all adults, with 1587 respondents in 593 families, 53·9 % of the original birth cohort families. A note search was undertaken initially in 2011 for grandparent deaths in the General Registry Office. Grandparental morbidity and mortality patterns were then related also to infant birth-weight. We related also the grandparents' blood pressure and lipoprotein profile at baseline recruitment stage to their grandchildren's birth-weight, the first such report to date and examined indicators of morbidity, stroke and diabetes based on self-report from the baseline questionnaire and general practice records for the grandparents at follow-up when the children averaged age 3 years. Maternal grandmothers' likelihood of both stroke and diabetes were inversely related to infant birth-weight and an inverse or U-shaped association was seen for maternal grandparents' mortality patterns, although not statistically significant. Conversely, the patterns for paternal grandparents and birth-weight were positive( Reference Shrivastava, Murrin and O'Brien 120 ) and paternal grandfathers who had now died were more likely to have had higher birth-weight grandchildren, adjusted for grandchildren's gestational age, gender, mothers' age, height, parity, educational status, smoking in pregnancy and grandparents' age, smoking and educational status. Given the novelty of these findings, and the possibility that some form of participant self-selection had influenced the association, we extended the General Registry Office note search in 2012 to all grandparents for whom we had any contact details and not just those who had been examined or completed a questionnaire at baseline. Preliminary analysis of these data reconfirm a positive association between higher birth-weight and paternal grandfathers' mortality( Reference Viljoen, Shrivastava and Murrin 121 ). Our findings in relation to the maternal line were therefore broadly consistent with most other published studies( Reference Smith, Wood and White 42 , Reference Naess, Stoltenberg and Hoff 43 ). Although the Norwegian linkage study shows a contrasting pattern to ours in relation to the paternal lineage( Reference Naess, Stoltenberg and Hoff 43 ), in that both paternal and maternal grandparental cardiovascular mortality patterns were inversely associated with birth-weight, there was an important effect for maternal smoking on that relationship and secular influences on lifestyle may be explaining the differences in patterns in the cohorts to date. There are very few evidenced studies published on this issue, which merits further research and investigation.

During 2012 we conducted examinations through their general practitioners of height and weight of children and their mothers and took salivary and hair samples for future genotyping, 301 were successfully completed and seventy-six of these mainly Dublin-based children also had blood samples taken for lipoprotein profile. Anthropometric measurements at birth, when children were aged 5 and 9 years are summarised in Table 5. We assessed the representativeness of the children who participated in the 2012 follow-up and had valid height and weight measurements. We compared these children with our original baseline cohort in terms of mothers' age, baseline BMI, education level and medical card holder status( Reference Khalil, Viljoen and O'Brien 119 ). A greater proportion of children at follow-up had mothers who did not hold a medical card (90·3 v. 79·2 %, P < 0·001) and achieved a tertiary level of education (60·3 v. 45·4 %, P < 0·001). Mothers were also slightly older than the cohort at baseline (mean age 32·0 years v. 29·0 years, P < 0·001) but did not differ significantly in BMI. The east coast grandparents are undergoing a cardiovascular risk profile assessment in 2013, including also salivary and hair samples and morbidity follow-up of grandparents through general practitioners is ongoing in 2013. In the next stage of this study, a detailed analysis of these biological outcomes will be undertaken, related to the information gathered in previous sweeps of the study.

Table 5. Anthropometric measurements of Lifeways cross-generation cohort study children at birth, at age 5 and at age 9 years

Conclusions

In the present review, we establish that there is renewed and increasing interest in the associations between intrauterine and early childhood development and health outcomes in later life and the possible explanatory mechanisms, particularly in relation to dietary intake. There are contrasting mechanisms for lower birth-weight and higher birth-weight infants, often within the normal clinical range for this parameter. Empirical studies have focused in recent years on possible genetic and epigenetic mechanisms. Although some authors maintain that studies showing observational associations across generations are subject to significant bias, which may be addressed by more robust, un-confounded genetic associations across generations, the evidence base is at an early stage. Human cross-generation studies of parents and children are increasingly common but three-generation studies are rare. The Lifeways cohort study is unusual in that it contains information on three generations of family members and dietary information on all active cohort members including adults and children. Empirical evidence to date shows consistent familial and cross-generational patterns, particularly in the maternal line. The latest sweep includes biological information on children for the first time, including samples for genetic profiling.

Acknowledgements

The Lifways cross-generation cohort study is overseen by an inter-disciplinary steering group chaired by the Principal Investigator, Professor Cecily Kelleher. We thank especially the participating families for their ongoing engagement and support.

Financial Support

The study has been funded in all sweeps since its establishment by peer-reviewed grants received from the Health Research Board of Ireland, presently HRA_PHS/2010/13. The Health Research Board had no role in the design, analysis or writing of this paper. Dr Rebecca Somerville receives an Ad Astra stipend from the UCD School of Public Health, Physiotherapy and Population Science and Ms Hala Khalil receives PhD funding from the Government of Saudi Arabia.

Conflicts of Interest

None.

Authorship

All authors are members of the present Lifeways study team and contributed to the drafting and scientific content of this paper. C. K. has been principal investigator of the study team since its establishment and has overseen all sweeps and data analyses to date. C. M. and A. S. received the PhD degrees for their work on the 2007 sweep when children averaged age 5 years, K. V., H. K. and R. S. are presently completing their PhD degrees on the 2011–2013 sweep. J. O'B. is data manager for the study.

References

1. Barker, DJ, Winter, PD, Osmond, C et al. (1989) Weight in infancy and death from ischaemic heart disease. Lancet 2, 577580.Google Scholar
2. Mossberg, HO (1989) 40-year follow-up of overweight children. Lancet 26, 491493.Google Scholar
3. Barker, DJP, Gluckman, PD, Godfrey, FM et al. (1993) Fetal Nutrition and cardiovascular disease in adult life. Lancet 341, 938941.Google Scholar
4. (GOPEC) Genetics of Pre-eclampsia Consortium (2007) Babies, pre-eclamptic mothers and grandparents: a three-generation phenotyping study. J Hypertens 25, 849854.Google Scholar
5. Barker, DJ, Larsen, G, Osmond, C et al. (2012) The placental origins of sudden cardiac death. Int J Epidemiol 41, 13941399.Google Scholar
6. Barker, DJP (2012) The Developmental Origins of Chronic Disease. Pub Health 126, 185189.Google Scholar
7. Adair, L, Fall, CHD, Osmond, C et al. (2013) Associations of linear growth and relative weight gain during early life with adult height and human capital in countries of low and middle income: findings from five birth cohort studies. Lancet 382, 525534.Google Scholar
8. Cnattingus, S, Villamor, E, Lagerros, YT et al. (2012) High birth weight and obesity-a vicious circle across generations. Int J Obes 36, 13201324.Google Scholar
9. Durmus, B, Arends, LR, Ay, L et al. (2013) Parental anthropometrics, early growth and the risk of overweight in pre-school children: The Generation R study. Ped Obes 8, 339350.Google Scholar
10. Eriksson, JG, Kajantie, E, Thornburg, KL et al. (2011) Mother's body size and placental size predict coronary heart disease in men. Eur Heart J 32, 22972303.Google Scholar
11. Gray, l, Davey Smith, G, McConnachie, A et al. (2012) Parental height in relation to offspring coronary heart disease: examining transgenerational influences on health using the West of Scotland Midspan Family study. Int J Epidemiol 41, 17761785.Google Scholar
12. Larsen, PS, Kamper-Jørgensen, M, Adamson, A et al. (2013) Pregnancy and birth cohort resources in Europe: a large opportunity for aetiological child health research. Paediatr Perinat Epidemiol 27, 393414.Google Scholar
13. O'Mahony, D, Fallon, UB, Hannon, F et al. (2007) The Lifeways Cross-Generation Study: design, recruitment and data management considerations. Ir Med J 100, Suppl., 36.Google Scholar
14. Kelleher, C, Bury, G & Murphy, A (2012) The Lifeways study: an invaluable resource. Forum J Ir Coll Gen Pract 29, 1416.Google Scholar
15. Fallon, UB, Murphy, AW, Majawit, E et al. (2007) Primary care utilisation rates in pre-school children. Ir Med J 100, Suppl., 2327.Google Scholar
16. Kawachi, I & Berkman, L (2000) Social Epidemiology. Oxford: Oxford University Press.Google Scholar
17. Chmurznska, A (2010) Fetal programming: link between early nutrition, DNA methylation and complex disease. Nutr Rev 68, 8798.Google Scholar
18. Harrison, M & Langley-Evans, SC (2009) Intergenerational programming of impaired nephrogenesis and hypertension in rats following maternal protein restriction during pregnancy. Br J Nutr 101, 10201030.Google Scholar
19. Horikoshi, M, Yaghootkar, H, Mook-Kanamori, DO et al. (2013) New Loci associated with birth weight identify genetic links between intrauterine growth and adult height and metabolism. Nat Genet 45, 7682.Google Scholar
20. Heijmans, BT, Tobi, EW, Stein, AD et al. (2008) Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci USA 105, 1704617049.Google Scholar
21. Ivanova, E, Chen, J-H, Segonds-Pichon, A et al. (2012) DNA methylation at differentially methylated regions of imprinted genes is resistant to developmental programming by maternal nutrition. Epigenetics 7, 12001210.Google Scholar
22. Ponzio, BF, Carvalho, MHC, Fortes, ZB et al. (2012) Implications of maternal nutrient restriction in transgenerational programming of hypertension and endothelial dysfunction across F1–F3 offspring. Life Sci 90, 571577.Google Scholar
23. Relton, CL & Davey Smith, G (2010) Epigenetic epidemiology of common complex disease: prospects for prediction, prevention and treatment. PloS Med 7, e1000356.Google Scholar
24. Yaghootkar, H & Freathy, RM (2012) Genetic origins of low birth weight. Curr Opin Clin Nut Metab Care 15, 258264.CrossRefGoogle ScholarPubMed
25. Poston, L (2012) Maternal obesity, gestational weight gain and diet as determinants of offspring long term health. Best Pract Res Clin Endocrinol Metab 26, 627639.Google Scholar
26. Pembrey, ME, Bygren, LO, Edvinsson, S et al. (2006) Sex-specific, male-line transgenerational responses in humans. Eur J Hum Gen 14, 159166.Google Scholar
27. Fox, M, Sear, R, Beise, J et al. (2010) Grandma plays favourites: X-chromosome relatedness and sex-specific childhood mortality. Proc R Soc B 277, 567573.Google Scholar
28. Andersen, AM & Osler, M (2004) Birth dimensions, parental mortality, and mortality in early adult age: a cohort study of Danish men born in 1953. Int J Epidemiol 33, 9299.Google Scholar
29. Smith, GD, Harding, S & Rosato, M (2000) Relation between infants' birth weight and mothers' mortality: prospective observational study. BMJ 320, 839840.Google Scholar
30. Friedlander, Y, Paltiel, O, Manor, O et al. (2007) Birthweight of offspring and mortality of parents: the Jerusalem Perinatal Study Cohort. Ann Epidemiol 17, 914922.Google Scholar
31. Wannamethee, SG, Lawlor, DA, Whincup, PH et al. (2004) Birhweight of offspring and paternal insulin resistance in late adulthood: cross sectional study. Diabetologia 47, 1218.Google Scholar
32. Palmer, TM, Nordestgarrard, BG, Benn, M et al. (2013) Association of plasma uric acid with ischaemic heart disease and blood pressure: Mendelian randomization analysis of two large cohorts. BMJ 347, f4262.Google Scholar
33. Lawlor, DA, Timpson, NJ, Harbord, RM et al. (2008) Exploring the developmental overnutrition hypothesis using Parental-Offspring associations and FTO as an instrumental variable. PLOS Med 5, e33.Google Scholar
34. Bonilla, C, Lawlor, DA, Ben-Shlomo, Y et al. (2012) Maternal and offspring fasting glucose and type 2 diabetes-associated genetic variants and cognitive function at age 8: a Mendelian randomization study in the Avon Longitudinal Study of Parents and Children. BMC Med Genet 13, 90.Google Scholar
35. Zambrano, E, Martinez-Samayoa, PM, Bautista, CJ et al. (2005) Sex differences in transgenerational alterations of growth and metabolism in progeny (F2) of female offspring (F1) of rats fed a low protein diet during pregnancy and lactation. J Physiol 566, 225236.Google Scholar
36. Torrens, C, Poston, L, Hanson, MA (2008) Transmission of raised blood pressure and endothelial dysfunction to the F2 generation induced by maternal protein restriction in the F0, in the absence of dietary challenge in the F1 generation. Br J Nutr 100, 760766.Google Scholar
37. Kaati, G, Bygren, LO, Pembrey, M et al. (2007) Transgenerational response to nutrition, early life circumstances and longevity. Eur J Hum Genet 15, 784790.Google Scholar
38. Keane, E, Layte, R, Harrington, J et al. (2012) Measured parental weight status and familial socio-economic status correlates with childhood overweight and obesity at age 9. PLoS One 7, e43503.Google Scholar
39. Davey Smith, G, Steer, C, Leary, S et al. (2007) Is there an intrauterine influence on obesity? Evidence from parent child associations in the Avon longitudinal Study of Parents and Children (ALSPAC). Arch Dis Child 10, 876880.Google Scholar
40. Manor, O & Koupil, I (2010) Birth weight of infants and mortality in their parents and grandparents: the Uppsala Birth Cohort Study. Int J Epidemiol 39, 12641276.Google Scholar
41. McCarron, P, Davey Smith, G, Hattersley, AT et al. (2004) Type 2 diabetes in grandparents and birth-weight in offspring and grandchildren in the ALSPAC study. J Epidemiol Commun Health 58, 517522.Google Scholar
42. Smith, GCS, Wood, AM, White, IR et al. (2010) Birth weight and the risk of cardiovascular disease in the maternal grandparents. Am J Epidemiol 171, 736744.Google Scholar
43. Naess, O, Stoltenberg, C, Hoff, DA et al. (2012) Cardiovascular mortality in relation to birth weight of children and grandchildren in 500 000 Norwegian families. Eur Heart J. [Epublication ahead of print].Google Scholar
44. Alves, E, Correia, S, Barros, H, Azevedo, A (2012) Prevalence of self-reported cardiovascular risk factors in Portuguese women: a survey after delivery. Int J Pub Health 57, 837847.Google Scholar
45. Bergmann, K, Bergmann, R, Von Kries, R et al. (2003) Early determinants of childhood overweight and adiposity in a birth cohort study: role of breast-feeding. Int J Obesity Relat Metab Disord 27, 162172.Google Scholar
46. Brion, M-JA, Ness, AR, Rogers, I et al. (2010) Maternal macronutrient and energy intakes in pregnancy and offspring intake at 10 y: exploring parental comparisons and prenatal effects. Am J Clin Nutr 91, 748756.Google Scholar
47. Burke, V, Beilin, LJ & Dunbar, D (2001) Family lifestyle and parental body mass index as predictors of body mass index in Australian children: a longitudinal study. Int J Obes Rel Metab Disord 25, 147157.Google Scholar
48. Brunekreef, B, Smit, J, De Jongste, J et al. (2002) The Prevention and Incidence of Asthma and Mite Allergy (PIAMA) birth cohort study: design and first results. Ped Allergy Immunol 13, 5560.Google Scholar
49. Chatzi, L, Plana, E, Daraki, V et al. (2009) Metabolic syndrome in early pregnancy and risk of preterm birth. Am J Epidemiol 170, 829836.Google Scholar
50. Dabelea, D, Hanson, RL, Lindsay, RS et al. (2000) Intrauterine exposure to diabetes conveys risks for type 2 diabetes and obesity: a study of discordant sibships. Diabetes 49, 22082211.Google Scholar
51. Davis, MM, McGonagle, K, Schoeni, RF et al. (2008) Grandparental and parental obesity influences on childhood overweight: implications for primary care practice. J Am Board Fam Med 21, 549554.Google Scholar
52. Dejmek, J, Solansky, I, Benes, I et al. (2000) The impact of polycyclic aromatic hydrocarbons and fine particles on pregnancy outcome. Environ Health Perspect 108, 11591164.Google Scholar
53. Devereux, G, Barker, RN & Seaton, A (2002) Antenatal determinants of neonatal immune responses to allergens. Clin Exp Allergy 32, 4350.Google Scholar
54. Drouillet, P, Forhan, A, De Lauzon-Guillain, B et al. (2009) Maternal fatty acid intake and fetal growth: evidence for an association in overweight women. The ‘EDEN mother–child’ cohort (study of pre- and early postnatal determinants of the child's development and health). Br J Nutr 101, 583591.Google Scholar
55. Dubois, L & Girard, M (2006) Early determinants of overweight at 4·5 years in a population-based longitudinal study. Int J Obes (Lond) 30, 610617.Google Scholar
56. Eggesbo, M, Stigum, H, Longnecker, MP et al. (2009) Levels of hexachlorobenzene (HCB) in breast milk in relation to birth weight in a Norwegian cohort. Environ Res 109, 559566. Epub 2009/05/05.Google Scholar
57. Emanuel, I, Filakti, H, Alberman, E et al. (1992) Intergenerational studies of human birthweight from the 1958 birth cohort. 1. Evidence for a multigenerational effect. Br J Obstet Gynaecol 99, 6774.Google Scholar
58. Gordon-Larsen, P, Adair, LS & Popkin, BM (2002) Ethnic differences in physical activity and inactivity patterns and overweight status. Obes Res 10, 141149.Google Scholar
59. Gracie, S, Lyon, A, Kehler, H et al. (2010) All Our Babies Cohort Study: recruitment of a cohort to predict women at risk of preterm birth through the examination of gene expression profiles and the environment. BMC Preg Childbirth 10, 87.Google Scholar
60. Grandjean, P, Weihe, P, Jorgensen, PJ et al. (1992) Impact of maternal seafood diet on fetal exposure to mercury, selenium, and lead. Arch Environ Health 47, 185195.Google Scholar
61. Guldner, L, Monfort, C, Rouget, F et al. (2007) Maternal fish and shellfish intake and pregnancy outcomes: a prospective cohort study in Brittany, France. Environ Health 6, 33.Google Scholar
62. Guxens, M, Ballester, F, Espada, M et al. (2012) Cohort profile: the INMA – INfancia y Medio Ambiente – (Environment and Childhood) project. Int J Epidemiol 41, 930940.Google Scholar
63. Grazuleviciene, R, Danileviciute, A, Nadisauskiene, R et al. (2009) Maternal smoking, GSTM1 and GSTT1 polymorphism and susceptibility to adverse pregnancy outcomes. Int J Environ Res Pub Health 6, 12821297.Google Scholar
64. Hawkes, CP, Hourihane, JOB, Kenny, LC et al. (2011) Gender- and gestational age-specific body fat percentage at birth. Pediatrics 128, e645ee51.Google Scholar
65. Hawkins, SS, Cole, TJ & Law, C (2008) Maternal employment and early childhood overweight: findings from the UK Millennium Cohort Study. Int J Obes (Lond) 32, 3038.Google Scholar
66. Hill, RA, Brophy, S, Brunt, H et al. (2010) Protocol of the baseline assessment for the Environments for Healthy Living (EHL) Wales cohort study. BMC Pub Health 10, 150.Google Scholar
67. Hryhorczuk, DO, Monaghan, S, Lukyanova, E et al. (1999) Collaborative research and research training through the “Family and Children of Ukraine” research programme. Int J Occup Environ Health 5, 213218.Google Scholar
68. Hui, LL, Nelson, EA, Yu, LM et al. (2003) Risk factors for childhood overweight in 6- to 7-y-old Hong Kong children. Int J Obes Relat Metab Disorders 27, 14111418.Google Scholar
69. Hunt, MS, Katzmarzyk, PT, Perusse, L et al. (2002) Familial resemblance of 7-year changes in body mass and adiposity. Obes Res 10, 507517.Google Scholar
70. Inskip, HM, Godfrey, KM, Robinson, SM et al. (2006) Cohort profile: the Southampton Women's Survey. Int J Epidemiol 35, 4248.Google Scholar
71. Jaddoe, VV, Duijn, C, Heijden, A et al. (2008) The Generation R Study: design and cohort update until the age of 4 years. Eur J Epidemiol 23, 801811.Google Scholar
72. Jedrychowski, W, Perera, FP, Tang, D et al. (2012) Impact of barbecued meat consumed in pregnancy on birth outcomes accounting for personal prenatal exposure to airborne polycyclic aromatic hydrocarbons: birth cohort study in Poland. Nutr 28, 372377.Google Scholar
73. Kaplowitz, HJ, Wild, KA, Mueller, WH et al. (1988) Serial and parent-child changes in components of body fat distribution and fatness in children from the London Longitudinal Growth Study, ages two to eighteen years. Hum Biol 60, 739758.Google Scholar
74. Karvonen, AM, Hyvärinen, A, Roponen, M et al. (2009) Confirmed moisture damage at home, respiratory symptoms and atopy in early life: a birth-cohort study. Pediatrics 124, e329e338.Google Scholar
75. Katier, N, Uiterwaal, CS, de Jong, BM et al. (2004) The Wheezing Illnesses Study Leidsche Rijn (WHISTLER): rationale and design. Eur J Epidemiol 19, 895903.Google Scholar
76. Kroke, A, Manz, F, Kersting, M et al. (2004) The DONALD Study. History, current status and future perspectives. Eur J Nutr 43, 4554.Google Scholar
77. Kummeling, I, Stelma, FF, Dagnelie, PC et al. (2007) Early life exposure to antibiotics and the subsequent development of Eczema, wheeze, and allergic sensitization in the first 2 years of life: the KOALA birth cohort study. Pediatrics 119, e225e231.Google Scholar
78. L'Abee, C, Vrieze, I, Kluck, T et al. (2011) Parental factors affecting the weights of the placenta and the offspring. J Perinat Med 39, 2734.Google Scholar
79. Laitinen, J, Power, C & Jarvelin, MR (2001) Family social class, maternal body mass index, childhood body mass index, and age at menarche as predictors of adult obesity. Am J Clin Nutr 74, 287294.Google Scholar
80. Lagström, H, Rautava, P, Kaljonen, A et al. (2012) Cohort profile: steps to the healthy development and well-being of children (the STEPS Study). Int J Epidemiol, Epublication ahead of print.Google Scholar
81. Li, L, Hardy, R, Kuh, D et al. (2008) Child-to-adult body mass index and height trajectories: a comparison of 2 British birth cohorts. Am J Epidemiol 168, 10081015.Google Scholar
82. Ludvigsson, J (2004) Milk consumption during pregnancy and infant birthweight. Acta Pædiatr 93, 14741478.Google Scholar
83. Magnus, P, Irgens, LM, Haug, K et al. (2006) Cohort profile: The Norwegian Mother and Child Cohort Study (MoBa). Int J Epidemiol 35, 11461150.Google Scholar
84. Mizutani, T, Suzuki, K, Kondo, N et al. (2007) Association of maternal lifestyles including smoking during pregnancy with childhood obesity. Obesity (Silver Spring) 15, 31333139.Google Scholar
85. Moschonis, G, Grammatikaki, E & Manios, Y (2008) Perinatal predictors of overweight at infancy and preschool childhood: the GENESIS study. Int J Obes (Lond) 32, 3947.Google Scholar
86. Nohr, EA, Vaeth, M, Baker, JL et al. (2008) Combined associations of prepregnancy body mass index and gestational weight gain with the outcome of pregnancy. Am J Clin Nutr 87, 17501759.Google Scholar
87. O' Callaghan, MJ, Williams, GM, Andersen, MJ et al. (1997) Prediction of obesity in children at 5 years: a cohort study. J Paediatr Child Health 33, 311316.Google Scholar
88. Oken, E, Huh, SY, Taveras, EM et al. (2005) Associations of maternal prenatal smoking with child adiposity and blood pressure. Obes Res 13, 20212028.Google Scholar
89. Park, HY, Park, JS, Sovcikova, E et al. (2009) Exposure to hydroxylated polychlorinated biphenyls (OH-PCBs) in the prenatal period and subsequent neurodevelopment in eastern Slovakia. Environ Health Perspect 117, 16001606.Google Scholar
90. Parkinson, KN, Pearce, MS, Dale, A et al. (2011) Cohort profile: the Gateshead Millennium Study. Int J Epidemiol 40, 308317.Google Scholar
91. Pirkola, J, Pouta, A, Bloigu, A et al. (2010) Risks of overweight and abdominal obesity at age 16 years associated with prenatal exposures to maternal prepregnancy overweight and gestational diabetes mellitus. Diabet Care 33, 11151121.Google Scholar
92. Polanska, K, Hanke, W, Gromadzinska, J et al. (2009) Polish mother and child cohort study–defining the problem, the aim of the study and methodological assumption. Int J Occup Med Environ Health 22, 383391.Google Scholar
93. Porta, D, Forastiere, F, Di Lallo, D et al. (2007) Enrolment and follow-up of a birth cohort in Rome. Epidemiol Prevenz 31, 303308.Google Scholar
94. Porta, D, Fantini, MP & GaCES (2006) Prospective cohort studies of newborns in Italy to evaluate the role of environmental and genetic characteristics on common childhood disorders. Ital J Ped 32, 350357.Google Scholar
95. Quante, M, Hesse, M, Dohnert, M et al. (2012) The LIFE child study: a life course approach to disease and health. BMC Pub Health 12, 1021.Google Scholar
96. Richiardi, L, Baussano, I, Vizzini, L et al. (2007) Feasibility of recruiting a birth cohort through the Internet: the experience of the NINFEA cohort. Eur J Epidemiol 22, 831837.Google Scholar
97. Schaefer-Graf, UM, Pawliczak, J, Passow, D et al. (2005) Birth weight and parental BMI predict overweight in children from mothers with gestational diabetes. Diab Care 28, 17451750.Google Scholar
98. Sekine, M, Yamagami, T, Hamanishi, S et al. (2002) Parental obesity, lifestyle factors and obesity in preschool children: results of the Toyama Birth Cohort study. J Epidemiol 12, 3339.Google Scholar
99. Skalkidou, A, Kieler, H, Stephansson, O et al. (2010) Ultrasound pregnancy dating leads to biased perinatal morbidity and neonatal mortality among post-term-born girls. Epidemiology 21, 791796.Google Scholar
100. Tsuchiya, KJ, Matsumoto, K, Suda, S et al. (2010) Searching for very early precursors of autism spectrum disorders: the Hamamatsu Birth Cohort for Mothers and Children (HBC). J Dev Orig Health Dis 1, 158173.Google Scholar
101. van Eijsden, M, Vrijkotte, TG, Gemke, RJ et al. (2011) Cohort profile: The Amsterdam Born Children and their Development (ABCD) Study. Int J Epidemiol 40, 11761186.Google Scholar
102. Vandentorren, S, Bois, C, Pirus, C et al. (2009) Rationales, design and recruitment for the Elfe longitudinal study. BMC Pediatr 9, 58.Google Scholar
103. Vecchi Brumatti, L, Montico, M, Russian, S et al. (2013) Analysis of motivations that lead women to participate (or not) in a newborn cohort study. BMC Pediatr 13, 53.Google Scholar
104. West, J, Manchester, B, Wright, J et al. (2011) Reliability of routine clinical measurements of neonatal circumferences and research measurements of neonatal skinfold thicknesses: findings from the Born in Bradford study. Paed Peri Epidemiol 25, 164171.Google Scholar
105. Albertson, AM, Anderson, GH, Crockett, SJ et al. (2003) Ready-to-eat cereal consumption: its relationship with BMI and nutrient intake of children aged 4 to 12 years. J Am Diet Assoc 103, 16131619.Google Scholar
106. Wickman, M, Kull, I, Pershagen, G et al. (2002) The BAMSE Project: presentation of a prospective longitudinal birth cohort study. Ped Allerg Immunol 13, 1113.Google Scholar
107. Segonds-Pichon, A, Hannon, F, Daly, S et al. (2007) Socio-demographic, lifestyle and cross-generation predictors of self-rated health in mothers during pregnancy. Ir Med J 100, Suppl., 712.Google Scholar
108. Murrin, C, Segonds-Pichon, A, Fallon, UB et al. (2007a) Self-reported pre-pregnancy maternal body mass index and infant birth-weight. Ir Med J 100, Suppl., 2023.Google Scholar
109. Murrin, C, Fallon, UB, Hannon, F et al. (2007b) Dietary habits of pregnant women in Ireland. Ir Med J 100, Suppl., 1215.Google Scholar
110. Kelleher, CC, Lotya, J, O'Hara, MC et al. (2008) Session 1: Public health nutrition. Nutrition and social disadvantage in Ireland. Proc Nutr Soc 67, 363370.Google Scholar
111. Niedhammer, I, O'Mahony, D, Daly, S et al. (2009) Occupational predictors of pregnancy outcomes in Irish working women in the Lifeways cohort. BJOG 116, 943952.Google Scholar
112. Niedhammer, I, Murrin, C, O'Mahony, D et al. (2012) Explanations for social inequalities in preterm delivery in the prospective Lifeways cohort in the Republic of Ireland. Eur J Pub Health 22, 533538.Google Scholar
113. Hamid, NA, Fitzpatrick, P, Rowan, A et al. (2013) Does the developmental plasticity hypothesis have application to Irish travellers? Findings from the All Ireland Traveller Health Study birth cohort 2008–2011. J Dev Origins Health Dis 4, 307316.Google Scholar
114. Fitz-simon, N, Fallon, U, O'Mahony, D et al. (2007) Mothers’ dietary patterns during pregnancy and risk of asthma symptoms in children at 3 years. Ir Med J 100, Suppl., 2732.Google Scholar
115. Viljoen, K, Shrivastava, A, O'Brien, J et al. (2012) Early determinants and prospective risk of Asthma symptoms in children at 5 years of age: the Lifeways Cross-Generation Cohort study. Ir J Med Sci 181, 116116.Google Scholar
116. Murrin, CM, Kelly, GE, Tremblay, RE et al. (2012) Body mass index and height over three generations: evidence from the Lifeways cross-generational cohort study. BMC Pub Health 25, 1281.Google Scholar
117. Shrivastava, A, Murrin, C, Sweeney, MR et al. (2012) Familial intergenerational and maternal aggregation patterns in nutrient intakes in the Lifeways Cross-Generation Cohort Study. Pub Health Nutr 13, 111.Google Scholar
118. Murrin, C, Shrivastava, A, Kelleher, CC (2013) Maternal macronutrient intake during pregnancy and 5 years postpartum and associations with child weight status aged five. Eur J Clin Nutr 67, 670679.Google Scholar
119. Khalil, H, Viljoen, K, O'Brien, J et al. (2012) Impact of the Irish recession on mothers' weight status: findings from the Lifeways Cross-generation Cohort Study 2011–2012. Faculty of Public Health Medicine 2012 Winter Scientific Meeting, Dublin, December 5th, 2012. Ir J Med Sci (In the Press).Google Scholar
120. Shrivastava, A, Murrin, C, O'Brien, J et al. (2012) Grandparental morbidity and mortality patterns are associated with infant birthweight in the Lifeways cross-generation cohort study. J Dev Orig Health Dis 3, 458468.Google Scholar
121. Viljoen, K, Shrivastava, A, Murrin, C et al. (2013) Paternal grandfathers' mortality association with grandchildren's birth-weight differs from that of other grandparents: 2012 mortality follow-up of the Lifeways cross-generation cohort study in the Republic of Ireland. Final Program for Society for Pediatric and Perinatal Epidemiological Research, 26th Annual Meeting, Boston, Mass, USA. Available at: http://www.sper.org/meeting/FinalProgram2013.pdf (accessed June 2013).Google Scholar
122. Kelleher, C, Friel, S, Nolan, G et al. (2002) Effect of social variation on the Irish diet. Proc Nutr Soc 61, 527536.Google Scholar
123. Niedhammer, I, Kerrad, S, Schütte, S et al. (2013) Material, psychosocial and behavioural factors associated with self-reported health in the Republic of Ireland: cross-sectional results from the SLAN survey. BMJ Open 3.Google Scholar
124. Bonzini, M, Palmer, KT, Coggon, D et al. (2011) Shift work and pregnancy outcomes: a systematic review with meta-analysis of currently available epidemiological studies. BJOG 118, 14291437.Google Scholar
125. Subramanian, SV, Ackerson, LK & Smith, GD (2010) Parental BMI and childhood undernutrition in India: an assessment of intrauterine influence. Pediatrics 3, e663e671.Google Scholar
126. Kivimaki, M, Lawlor, DA, Smith, GD et al. (2007) substantial intergenerational increases in body mass index are not explained by the fetal under-nutrition hypothesis: the Cardiovascular Risk in Young Finns Study. Am J Clin Nutr 5, 15091514.Google Scholar
127. Moon, RJ, Harvey, NC, Robinson, SM et al. (2013) Maternal plasma polyunsaturated fatty acid status in late pregnancy is associated with offspring body composition in childhood. J Clin Endocrinol Metab 98, 299307.Google Scholar
128. Devereux, G & Seaton, A (2005) Diet as a risk factor for atopy and asthma. J Allerg Clin Immunol 115, 11091117.Google Scholar
129. Leventakou, V, Roumeliotaki, T, Martinez, D et al. (2013) Fish intake during pregnancy, fetal growth and gestational length: a meta-analysis within 19 European Birth cohort studies. J Dev Orig Health Dis 4 (Suppl 2), S211.Google Scholar
130. Sonnenschein-van de Voort, AMM, Arends, LR, de Jongste, JC et al. (2013) Preterm birth, birth weight and infant growth and the risk of childhood asthma: a meta-analysis of 31 birth cohorts. Eur Resp Soc conference abstract proceedings 2013, 715S, www.ers-education.org/search/quick-search.aspx (accessed 27 November 2013).Google Scholar
Figure 0

Table 1. Summary of data collection sweeps to date in the Lifeways cross-generation cohort study

Figure 1

Table 2. Prospective studies with information on either diet or parental BMI, or both

Figure 2

Table 3. Cross-generational or longitudinal Influences on health outcomes in the Lifeways cohort study 2001–2013

Figure 3

Table 4. Predictors of General Practitioner-reported wheeze (10·4 % of n 614) in children aged 3 years and of maternal reported asthma (14·3 % of n 511) in children aged 5 years in the Lifeways Cohort Study

Figure 4

Table 5. Anthropometric measurements of Lifeways cross-generation cohort study children at birth, at age 5 and at age 9 years