Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-06-10T11:04:49.965Z Has data issue: false hasContentIssue false

The association between the maternal diet and the maternal and infant gut microbiome: a systematic review

Published online by Cambridge University Press:  04 March 2020

Siofra E. Maher
Affiliation:
UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Republic of Ireland
Eileen C. O’Brien
Affiliation:
UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Republic of Ireland
Rebecca L. Moore
Affiliation:
UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Republic of Ireland
David F. Byrne
Affiliation:
UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Republic of Ireland
Aisling A. Geraghty
Affiliation:
UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Republic of Ireland
Radka Saldova
Affiliation:
The National Institute for Bioprocessing, Research, and Training (NIBRT), Dublin, Republic of Ireland UCD School of Medicine, College of Health and Agricultural Science, University College Dublin, Republic of Ireland
Eileen F. Murphy
Affiliation:
Alimentary Health Group, Cork Airport Business Park, Cork, Republic of Ireland
Douwe Van Sinderen
Affiliation:
APC Microbiome Ireland, National University of Ireland, Cork, Republic of Ireland School of Microbiology, National University of Ireland, Cork, Republic of Ireland
Paul D. Cotter
Affiliation:
APC Microbiome Ireland, National University of Ireland, Cork, Republic of Ireland Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Republic of Ireland
Fionnuala M. McAuliffe*
Affiliation:
UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Republic of Ireland
*
*Corresponding author: Professor Fionnuala M. McAuliffe, fax +353 1 662 7586, email fionnuala.mcauliffe@ucd.ie
Rights & Permissions [Opens in a new window]

Abstract

During pregnancy, changes occur to influence the maternal gut microbiome, and potentially the fetal microbiome. Diet has been shown to impact the gut microbiome. Little research has been conducted examining diet during pregnancy with respect to the gut microbiome. To meet inclusion criteria, dietary analyses must have been conducted as part of the primary aim. The primary outcome was the composition of the gut microbiome (infant or maternal), as assessed using culture-independent sequencing techniques. This review identified seven studies for inclusion, five examining the maternal gut microbiome and two examining the fetal gut microbiome. Microbial data were attained through analysis of stool samples by 16S ribosomal RNA gene-based microbiota assessment. Studies found an association between the maternal diet and gut microbiome. High-fat diets (% fat of total energy), fat-soluble vitamins (mg/d) and fibre (g/d) were the most significant nutrients associated with the gut microbiota composition of both neonates and mothers. High-fat diets were significantly associated with a reduction in microbial diversity. High-fat diets may reduce microbial diversity, while fibre intake may be positively associated with microbial diversity. The results of this review must be interpreted with caution. The number of studies was low, and the risk of observational bias and heterogeneity across the studies must be considered. However, these results show promise for dietary intervention and microbial manipulation in order to favour an increase of health-associated taxa in the gut of the mother and her offspring.

Type
Systematic Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
© The Author(s), 2020. Published by Cambridge University Press on behalf of The Nutrition Society

Advancements in the past decade in next-generation sequencing and associated bioinformatics analyses have facilitated a more in-depth study of the human gut ‘microbiome’; a word coined to describe the overall community of micro-organisms in the gastrointestinal tract(Reference Aagaard, Petrosino and Keitel1). Links between the microbiome and many physiological conditions of the associated host have been made(Reference Serino, S and Trabelsi2Reference Majnik and Lane4). The various components contributing and modulating the microbiome are yet to be truly defined; however, environmental factors such as lifestyle and diet have come to the fore(Reference Hemalatha5,Reference Rothschild, Weissbrod and Barkan6) .

Diet and dietary patterns have been shown to rapidly alter microbial diversity and in turn influence host physiology(Reference Laitinen, Collado and Isolauri7,Reference Sheflin, Melby and Carbonero8) . In non-pregnant cohorts, the dietary macronutrients fat and fibre have most commonly been demonstrated to be able to cause a shift in microbial diversity, with fibre consumption associated with beneficial effects(Reference Chu, Antony and Ma9Reference Valdes, Walter and Segal11).

With respect to dietary patterns, the Mediterranean diet, the Western diet, low-fat and high-fibre diets have been examined in greatest detail, with some research showing Western diet to influence the gut microbiome more considerably than BMI(Reference Sheflin, Melby and Carbonero8,Reference Davis, Yadav and Barrow12) . Diets high in fibre have been shown to have the ability to increase the relevant abundance of SCFA-producing bacteria(Reference LeBlanc, Chain and Martín13). This is in contrast to diets rich in animal fats, high in saturated fat and protein, which have been shown to have a negative impact(Reference Singh, Chang and Yan14). The blueprint for the optimal gut microbiome is still unknown, but the negative association of decreased diversity is commonly observed. Decreased diversity is linked to a phenomenon called dysbiosis (a disruption of normal gut microbiota); diversity is involved in the survival and adaptability of any ecosystem, the microbiome being no exception(Reference Cardinale, Palmer and Collins15). Furthermore, diets such as the Western diet are associated with decreased microbial diversity(Reference Davis, Yadav and Barrow12,Reference Wu, Chen and Hoffmann16) .

Diversity is the method used to assess the gut microbiome. α Diversity (also described as the intra-personal variation) is the individual’s diversity in the microbiota. It has been suggested that a higher α diversity correlates with a healthier microbiome(Reference Kennedy, Naeem and Howe17,Reference Manichanh, Rigottier-Gois and Bonnaud18) . As for many ecosystems, a high species diversity is linked with greater resistance to dysbiosis (disruption of microbiota composition from outside normal ranges) and an overall health within the host(Reference Keesing, Belden and Daszak19).

β Diversity on the other hand describes the interpersonal variation of microbial composition and can be based on collapsing all microbial data to a single coordinate point and measuring the distance (using various metrics, e.g. Bray-Curtis, unweighted and weighted UniFrac, Euclidean) between this point and another, usually another participant, person or collection site.

In pregnancy, the gut microbiome is thought to be dynamic with a change seen in first trimester diversity compared with that of the third trimester(Reference Koren, Goodrich and Cullender20). Mode of delivery, pre-term birth, breast-feeding and maternal diet have been identified as important factors that directly influence the composition of the neonatal gut microbiota(Reference Chu, Valentine and Seferovic21). Likewise, the presence of furry pets in the home has been shown to influence the composition of the gut microbiota of newborns(Reference Tapiainen, Paalanne and Tejesvi22).

There is limited literature examining the association between maternal macronutrient and micronutrient intake and infant and maternal gut microbiome. Without this knowledge, it is impossible to develop a therapeutic use of dietary manipulation to modulate the microbiome and in turn lead to improvements in infant and maternal health.

The aim of this systematic review was to summarise current evidence relating to the association between maternal diet in pregnancy and both the maternal and neonatal gut microbiome.

Methods

Protocol and registration

The PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) Statement for reporting on systematic reviews was followed(Reference Liberati, Altman and Tetzlaff23). A search checklist of items to include methods, strategy, study selection process, a risk of bias tool and summary measures was used and reported.

Eligibility criteria

To be included in the review, the studies had to be observational or cross-sectional in design, subjects needed to be pregnant women and/or infants within the first 6 weeks postpartum. The study needed to include a formal dietary analysis during pregnancy and use a culture-independent sampling technique to assess the gut microbiome. Studies had to include details of ages, ethnicities and demographic characteristics of the women/infants. Studies that evaluated the effect of dietary supplementation or probiotic use only, without formal dietary assessment, were excluded, as book chapters, online abstracts and conference proceedings were not included. Articles had to be published in English, and no time restrictions was imposed.

Outcomes

The main outcomes examined in this review were the maternal or neonatal gut microbiome composition and diversity, as assessed by culture-independent sequencing techniques. These outcomes are expressed as microbial diversity in terms of both α (intra-individual variation) and β (inter-individual variation) diversity and relative abundance of specific microbes. Indices such as Shannon’s index, whole-tree phylogenetic diversity and Simpson’s index, which measure diversity within microbial communities or Unifrac distances, Bayesian models or principal component analysis, which measure diversity between microbial communities were included.

Information sources

The following five electronic databases were searched; MEDLINE (PubMed), Cochrane Library, Web of Science, CINAHL and Ovid. The last search was conducted on 7 October 2019.

Search

Search terms are as follows: human; antenatal; pregnant; pregnancy; maternal; microbiome; microbial; microbiota; microbe; gut bacteria; gut microbiome; nutrient; diet; nutrition; dietary.

Search terms were identified by initial scoping searches and then adjusted depending on the electronic database searched, to better match the key words and indexing terms of each database, and align with MeSH (Medical Subject Headings) terms.

Study selection

Summary measures

It was not possible to carry out a summary analysis or meta-analysis for this systematic review due to heterogeneity across the included studies. This included differences in stage of pregnancy of participants, the stool sample analysed, the dietary assessment tool used and the method of microbiota analysis. An overall description of individual results is therefore provided in the Results section, separated into two sections: maternal gut microbiota and neonatal gut microbiota.

Results

Identified articles were added to a reference manager software package (EndNote version 7.7.1), and duplicates removed. A new file was created minus the duplicates. Studies were then screened based on the study title. Papers were then excluded based on reading an abstract and its fitting of the defined population, comparison, intervention and outcome (PICO) terms. Abstracts were reviewed independently by two researchers (S. E. M and E. C. O’B.), and two individual spreadsheets were created with researchers’ final included abstracts. Full papers of said abstracts were reviewed independently by two researchers (S. E. M. and E. C. O’B.) and both parties selected final papers. Disagreements were resolved by a third party (F. M. M.). A flow chart created based on the PRISMA guidelines can be seen below (Fig. 1).

Fig. 1. Flow diagram of study selection. Flow diagram depicting each stage of the study identification process. PICO, population, comparison, intervention and outcome.

Study characteristics

The study characteristics are described in Table 1.

Table 1. Summary of results

NoMIC, Norwegian microflora study; RCT, randomised controlled trial; SPRING, Study of PRobiotics IN Gestational diabetes; Ow, overweight; Ob, obese; GDM, gestational diabetes mellitus; N/A, not applicable; V, vegetarian; C, control; DSQ, Dietary Screener Questionnaire; rRNA, ribosomal RNA; IDQ, Index of Dietary Quality; PcoA, principal component analysis; LefSe, linear discriminant analysis effect size; PD, phylogenetic diversity; OTU, operational taxonomic unit; PERMANOVA, permutational multivariate ANOVA; ACE, abundance-based coverage estimator; FDR, false discovery rate; C-section; Caesarean section; TEI, total energy intake.

Risk of bias in individual studies

The seven studies were assessed for risk of bias using the 2016 ROBINS-I (‘Risk Of Bias In Non-randomised Studies – of Interventions’) assessment tool(Reference Sterne, Hernan and Reeves31). The ROBINS-I consists of an assessment and a scoring algorithm that ranks studies with little, moderate or severe bias, on contact with the Cochrane Group; this was agreed to be the most suitable risk of bias tool. Three researchers (S. E. M., E. C. O’B. and D. F. B.) independently assessed the included articles.

Risk of bias assessment

All studies were subject to a varying level of bias due to the observational nature of the analysis and potential confounders. Four studies were found to be at serious risk of bias in at least one domain, with three studies at moderate risk of bias (Table 2). No study was judged to be at a critical risk of bias in any domain. Therefore, the seven studies were included in this review(Reference Chu, Antony and Ma24Reference Laitinen and Mokkala30).

Table 2. ROBINS-I (Risk Of Bias In Non-randomised Studies – of Interventions) risk of bias results

Maternal diet and the maternal gut microbiota

The association between maternal diet and the maternal gut microbiome composition in pregnancy was investigated in five studies. All five studies reported that the maternal gut microbiome in pregnancy is influenced by maternal diet to varying degrees. In addition, specific macronutrients are associated with distinct bacterial compositions and relative abundances and can modulate, either positively or negatively, the diversity of the gut microbiome.

Three studies identified an association between dietary fat intake and gut microbiome composition(Reference Mandal, Godfrey and McDonald25,Reference Roytio, Mokkala and Vahlberg26,Reference Barrett, Gomez-Arango and Wilkinson28) . Two of these studies reported a negative correlation between α diversity and intakes of cholesterol(Reference Mandal, Godfrey and McDonald25), total fat and SFA(Reference Roytio, Mokkala and Vahlberg26). The third study(Reference Barrett, Gomez-Arango and Wilkinson28) reported a difference in β diversity, although α diversity did not differ. Furthermore, microbial composition differed by type of fat. Intakes of cholesterol and MUFA were associated with relative increases in Proteobacteria composition(Reference Mandal, Godfrey and McDonald25). In contrast, SFA intake was linked to relative decreases in this phylum and also negatively associated with the genus Roseburia (rho = −0·4, P = 0·038)(Reference Barrett, Gomez-Arango and Wilkinson28). The study by Barrett et al. (Reference Barrett, Gomez-Arango and Wilkinson28) compared the effect of a vegetarian diet v. omnivorous diet in early pregnancy on the maternal microbiome composition. Barrett et al. (Reference Barrett, Gomez-Arango and Wilkinson28) reported that women on the vegetarian diet had a higher intake of PUFA, of which, linoleic acid positively correlated with Holdemania (rho = 0·51, P = 0·006) and Roseburia (rho = 0·40, P = 0·04) abundance, but negatively with Collinsella (rho = −0·50, P = 0·009).

Four studies reported results on dietary carbohydrate intake and gut microbiome composition(Reference Roytio, Mokkala and Vahlberg26,Reference Barrett, Gomez-Arango and Wilkinson28Reference Laitinen and Mokkala30) . Each of these studies reported that higher dietary fibre intakes were positively associated with increased gut microbiota diversity and richness. Moreover, similar associations between dietary fibre intake and relative abundance of specific bacteria were reported in three of these papers(Reference Barrett, Gomez-Arango and Wilkinson28Reference Laitinen and Mokkala30). Higher fibre intakes were positively associated with increased relative abundances of Holdemania, Roseburia, Lachnospira and Coprococcus. In contrast, dietary fibre intake was negatively associated with relative Collinsella (Actinobacteria) and Sutterella (Proteobacteria) abundances.

The study by Mandal et al. (Reference Mandal, Godfrey and McDonald25) reported increased dietary intakes of fat-soluble vitamins, such as vitamin D and retinol are inversely correlated with α diversity. Vitamin D showed the strongest associations for both measures. For Shannon’s diversity, only vitamin D was significantly associated (−5·1 % change in diversity per unit increase in vitamin D intake, P < 0·001). The authors report that associations between dietary components and β diversity did not show any effects (UniFrac (weighted and unweighted; data not shown)). Furthermore, multiple regression modelling was used to assess associations between microbial composition and one standard deviation of nutrient intake for several dietary components. Vitamin D was associated with relative increases in Actinobacteria and Proteobacteria. Retinol was also associated with relative increases in Proteobacteria composition. Conversely, protein and vitamin E correlated with relative decreases in Proteobacteria.

Protein intake was collected and examined by all studies; however, significant findings were not seen(Reference Chu, Antony and Ma24Reference Barrett, Gomez-Arango and Wilkinson28,Reference Sterne, Hernan and Reeves31) .

Maternal diet and the neonatal gut microbiome

Two studies investigated the effect of maternal diet in pregnancy on the neonatal gut microbiome. Both studies reported that maternal diet in pregnancy is associated with distinct changes in the neonatal gut microbiome.

Chu et al. (Reference Chu, Antony and Ma24) identified an association between maternal dietary fat intake and distinct changes in the neonatal gut microbiota, at birth and 4–6 weeks of age. Participants were grouped by extremes of dietary fat intake (1 sd greater or less than the cohort mean), to produce a high-fat maternal diet group (n 13, 43·1 % fat intake) and low-fat group (n 13, 24·4 % fat intake). Significant differences in neonatal microbiome clusters were detected between groups (principal component analysis unweighted UniFrac: P = 0·04). There was an inverse association between high-fat maternal diet and relative abundance of Bacteroides in neonatal stool at delivery, persisting at 6 weeks, whereas Enterococcus abundance was higher in the high-fat group at delivery only.

The study by Lundgren et al. (Reference Lundgren, Madan and Emond27) found that associations between maternal diet and the gut microbiome composition of infant stool samples differed by mode of delivery. Three distinct genera clusters were identified in vaginally born infants (cluster 1: Bifidobacterium; cluster 2: Streptococcus and Clostridium and cluster 3: Bacteroides). Through multinomial logistic regression, the odds of falling within cluster 2 were 2·73 times higher with each additional fruit serving per d. Furthermore, maternal fruit intake was negatively associated with the Bifidobacterium group. The clusters differed in infants delivered by Caesarean section (cluster 1: Bifidobacterium; cluster 2: high Clostridium, low Streptococcus and low Ruminococcus; cluster 3: high Enterobacteriaceae, Ruminococcus and Lachnospiraceae). In this sub-group, the analysis found a 2·36 increase in odds of being in a high Clostridium-low Streptococcus cluster with every increase of dairy portion. Maternal fish intake was positively associated with the Streptococcus genus in both groups of infants. In addition, red meat consumption was positively associated with the Bifidobacterium genus for the Caesarean section group. Likewise, the association between maternal alternative Mediterranean diet score differed slightly by mode of delivery, with positive associations existing with the Enterobacteriaceae family and the genus Streptococcus in the vaginally born group. In the Caesarean section group, a negative association was observed. Taking premature infants out of the analysis did not change results.

Discussion

Main findings in this study

Pregnancy is a unique time point during which improvement to the health of the woman can also benefit the immediate and long-term health of the child. Manipulating the gut microbiome during pregnancy may be beneficial to the health of both mother and baby(Reference Chu, Valentine and Seferovic21). Indeed, each of the studies included in this review demonstrates the important influence of maternal diet in pregnancy in modulating the gut microbiome of mother and infant, both beneficially and detrimentally. They provide evidence that diet quality, determined by factors including amount of fibre, fat, fat-soluble vitamins, fruit and vegetables, and fish and meat consumed, is associated with distinct gut microbiota profiles and diversity of the gut microbiota. Interestingly, the findings from Lundgren et al. (Reference Lundgren, Madan and Emond27) demonstrate that the influence of maternal diet on gut microbiota profiles differ by delivery mode.

The findings from this review align with those of the prevailing literature. Recent studies have shown the influence of diet and the gut–brain axis in the prenatal period, with the gut microbiome potentially playing a role in neurodevelopment(Reference Borre, O’Keeffe and Clarke32). In addition, diet has been shown to change the composition and metabolism of gut microbes(Reference Riaz Rajoka, Shi and Mehwish33). Fibre and to a lesser degree fat have been identified as important modulators of the human gut microbiome(Reference Rothschild, Weissbrod and Barkan6,Reference Valdes, Walter and Segal11) . It is estimated that approximately 20–60 g of undigested carbohydrate reaches the large intestine (the area with the highest density of gut microbes) daily(Reference Silvester, Englyst and Cummings34). This is larger than the amount of fat and protein that reach the colon, which are both readily digested in the upper gastrointestinal tract(Reference Scott, Gratz and Sheridan10), and thus are more likely to impact on the small intestinal microbiota. In high-fat diets (>35 % of total energy intake), a greater proportion of fat will reach the colon and it is hypothesised that this causes reduction of bacteria usually used for carbohydrate degradation, causing a shift in the microbiome as a whole(Reference Rowland, Gibson and Heinken35). In contrast, high-fibre diets (>25 g/d(36)) are associated with greater relative abundances of SCFA-producing bacteria (such as Holdemania and Roseburia) and relative depletion of lactate producers (such as Collinsella), with the former considered directly associated with beneficial metabolic profiles(Reference den Besten, van Eunen and Groen37).

In addition, probiotics have emerged as another promising means by which to manipulate the maternal gut microbiota with a view to improve health and clinical outcomes(Reference Valdes, Walter and Segal11). However, the research behind their use in pregnancy has not shown clear reduction of adverse outcomes such as preterm birth or secondary outcomes such as gestational diabetes or reduction in glucose level(Reference Lindsay, Brennan and Kennelly38,Reference Jarde, Lewis-Mikhael and Moayyedi39) . Jarde et al. (Reference Jarde, Lewis-Mikhael and Moayyedi39) conducted a systematic review with nineteen studies which found no definitive link between probiotic supplementation and improved clinical sequela. Likewise, Lindsay et al. (Reference Lindsay, Brennan and Kennelly38) examined the effect of probiotic supplementation on several important clinical outcomes including birth weight and fasting glucose, with no reported difference in those parameters. Further clarity is required regarding the clinical benefits of probiotic supplementation use during pregnancy. Hence, dietary manipulation of the maternal (and neonatal) gut microbiota may offer more readily available opportunities in the immediate term for improving the health of mother and child.

Environmental determinants have been demonstrated as important mediators of the human gut microbiota, including the shared home environment. Factors such as having other children at home, or having furry pet animals, have been shown to directly influence the composition of the maternal and neonatal gut microbiota(Reference Rothschild, Weissbrod and Barkan6,Reference Tun, Konya and Takaro40) . None of the studies in this review explored these variables.

Significant heterogeneity pervades multiple domains of the studies included in this review. Consequently, the findings of this review should be interpreted with caution and considered in the context of the wider literature. Four of the five studies focusing on maternal gut outcomes studied a cohort of women with overweight and obesity. Although this could be considered a representative sample in the context of rising overweight and obesity rates, a comprehensive well-designed study examining normal-weight and overweight/obese women in pregnancy, nutrients and the microbiome must be conducted first for comparison. BMI was self-reported by participants in the study by Lundgren et al. (Reference Lundgren, Madan and Emond27). It has been shown that self-reported BMI underestimates actual BMI in pregnancy(Reference Natamba, Sanchez and Gelaye41).

In addition, the method of dietary assessment varied considerably across the studies. Five studies assess diet by FFQ, one by 3-d food diaries, and one by Index of Diet Quality. Roytio et al. (Reference Roytio, Mokkala and Vahlberg26) used 3-d food diaries as well as providing participants with oral and written instruction and a portion picture booklet. This would allow for a more accurate correlation between diet and the microbiome. Of the five studies that employed FFQ, there were differences in the period of time assessed (from 4 to 16 weeks) and the time point in pregnancy it was administered (two in first trimester, two in second trimester and one in third). As pregnancy progresses, diet may vary considerably due to increased early satiety, reflux and constipation. There is also potential for misclassification of food groups using FFQ. In the Willett FFQ used in Lundgren et al. (Reference Lundgren, Madan and Emond27), fruit and fruit juices are both in the fruit food group. Fruit juices contain high amounts of free sugar and lower amounts of fibre, and therefore the effect on the gut microbiota could be considerably different(Reference Koutsos, Tuohy and Lovegrove42). Likewise, differences in the temperature at which collected stool samples were stored and the time point at which they were collected across the studies could influence the comparability of the results.

A major strength of this systematic review is the techniques used in the search strategy and the analysis of bias. The PRISMA guidelines recommended by the Cochrane Group were used(Reference Liberati, Altman and Tetzlaff23).

Another strength of this review is that all seven studies used culture-independent analytical techniques. The use of culture-specific sampling technique is now seen as a major risk of bias in the microbiological research. The benefit of culture-independent analytical techniques is that all microbial species present in the microbiome can be identified and therefore analysed(Reference Aagaard, Petrosino and Keitel1).

Future directions of studies

The examination of detailed dietary data in pregnancy and its influence on the microbiome must be conducted in detail in a cohort representative of a normal obstetric population. Without this, findings from subgroups are difficult to interpret. Dietary analysis should be conducted in a systematic manner. Food diaries most accurately capture intake within the last week and therefore may be most appropriate compared with FFQ that capture intake in the last few months. With this said, there is emerging evidence to suggest that long-term food patterns have a stronger role in the metabolism and composition of the human gut microbiome than short-term dietary changes(Reference Wu, Chen and Hoffmann16). Therefore, perhaps both FFQ and food diaries methodologies should be used for each analysis.

Conclusion

In summary, this review demonstrates the important influence of maternal diet in pregnancy in modulating the gut microbiome of mother and infant, both beneficially and detrimentally. The findings provide evidence that diet quality, determined by factors including amount of fibre, fat, fat-soluble vitamins, fruit and vegetables, and fish and meat consumed, is associated with distinct gut microbiota profiles and diversity of the gut microbiota. However, confidence in the quality of this evidence is limited due to methodological limitations within the studies, and variability between studies. Pregnancy is a unique time point during which benefits to the health of the mother can also benefit that of the child. Hence, further high-quality research is required in this area to elucidate the relationship between diet quality and the gut microbiota of mother and child.

Acknowledgements

This publication has emanated from research supported in part by a research grant from Science Foundation Ireland (SFI) under grant nos 12/RC/2273 and 16/SP/3827 and by a research grant from Alimentary Health Ltd.

R. S., E. F. M., D. V. S., P. D. C. and F. M. M. designed the research; S. E. M., A. A. G., R. L. M., E. C. O’B. and D. F. B. conducted the research; S. E. M., E. C. O’B. and D. F. B. analysed the data; S. E. M., E. C. O’B. and D. F. B. wrote the paper; F. M. M. had primary responsibility for final content. All authors read and approved the final manuscript.

Contents are the authors’ own view. E. F. M. is Technical Director at Alimentary Health Group. The authors have no other disclosures to declare.

References

Aagaard, K, Petrosino, J, Keitel, W, et al. (2013) The Human Microbiome Project strategy for comprehensive sampling of the human microbiome and why it matters. FASEB J 27, 10121022.CrossRefGoogle ScholarPubMed
Serino, MN, S, Nicolas, Trabelsi, MS, et al. (2017) Young microbes for adult obesity. Pediatr Obes 12, e28e32.CrossRefGoogle ScholarPubMed
Zmora, NB, Nicolas, S, Levy, M, et al. (2017) The role of the immune system in metabolic health and disease. Cell Metab 25, 506521.CrossRefGoogle ScholarPubMed
Majnik, AV & Lane, RH (2015) The relationship between early-life environment, the epigenome and the microbiota. Epigenomics 7, 11731184.CrossRefGoogle ScholarPubMed
Hemalatha, R (2016) Diet and gut microbiota in human health. Proc Indian Natl Sci Acad 82, 14371447.CrossRefGoogle Scholar
Rothschild, D, Weissbrod, O, Barkan, E, et al. (2018) Environment dominates over host genetics in shaping human gut microbiota. Nature 555, 210.CrossRefGoogle ScholarPubMed
Laitinen, K, Collado, MC & Isolauri, E (2010) Early nutritional environment: focus on health effects of microbiota and probiotics. Benef Microbes 1, 383390.CrossRefGoogle ScholarPubMed
Sheflin, AM, Melby, CL, Carbonero, F, et al. (2017) Linking dietary patterns with gut microbial composition and function. Gut Microbes 8, 113129.CrossRefGoogle ScholarPubMed
Chu, D, Antony, KM, Ma, J, et al. (2016) A maternal high fat diet (HFD) during gestation alters the neonatal gut microbiome in a human population based longitudinal cohort. Am J Obstet Gynecol 214, S79.CrossRefGoogle Scholar
Scott, KP, Gratz, SW, Sheridan, PO, et al. (2013) The influence of diet on the gut microbiota. Pharmacol Res 69, 5260.CrossRefGoogle ScholarPubMed
Valdes, AM, Walter, J, Segal, E, et al. (2018) Role of the gut microbiota in nutrition and health. BMJ 361, k2179.CrossRefGoogle ScholarPubMed
Davis, SC, Yadav, JS, Barrow, SD, et al. (2017) Gut microbiome diversity influenced more by the Westernized dietary regime than the body mass index as assessed using effect size statistic. Microbiologyopen 6, e00476.CrossRefGoogle Scholar
LeBlanc, JG, Chain, F, Martín, R, et al. (2017) Beneficial effects on host energy metabolism of short-chain fatty acids and vitamins produced by commensal and probiotic bacteria. Microb Cell Fact 16, 79.CrossRefGoogle ScholarPubMed
Singh, RK, Chang, H-W, Yan, D, et al. (2017) Influence of diet on the gut microbiome and implications for human health. J Transl Med 15, 73.CrossRefGoogle ScholarPubMed
Cardinale, BJ, Palmer, MA & Collins, SL (2002) Species diversity enhances ecosystem functioning through interspecific facilitation. Nature 415, 426.CrossRefGoogle ScholarPubMed
Wu, GD, Chen, J, Hoffmann, C, et al. (2011) Linking long-term dietary patterns with gut microbial enterotypes. Science 334, 105.CrossRefGoogle ScholarPubMed
Kennedy, TA, Naeem, S, Howe, KM, et al. (2002) Biodiversity as a barrier to ecological invasion. Nature 417, 636638.CrossRefGoogle ScholarPubMed
Manichanh, C, Rigottier-Gois, L, Bonnaud, E, et al. (2006) Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 55, 205211.CrossRefGoogle ScholarPubMed
Keesing, F, Belden, LK, Daszak, P, et al. (2010) Impacts of biodiversity on the emergence and transmission of infectious diseases. Nature 468, 647.CrossRefGoogle ScholarPubMed
Koren, O, Goodrich, JK, Cullender, TC, et al. (2012) Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell 150, 470480.CrossRefGoogle ScholarPubMed
Chu, DM, Valentine, GC, Seferovic, MD, et al. (2019) The development of the human microbiome: why moms matter. Gastroenterol Clin North Am 48, 357375.CrossRefGoogle ScholarPubMed
Tapiainen, T, Paalanne, N, Tejesvi, MV, et al. (2018) Maternal influence on the fetal microbiome in a population-based study of the first-pass meconium. Pediatr Res 84, 371379.CrossRefGoogle Scholar
Liberati, A, Altman, DG, Tetzlaff, J, et al. (2009) The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. PLOS Med 6, e1000100.CrossRefGoogle ScholarPubMed
Chu, DM, Antony, KM, Ma, J, et al. (2016) The early infant gut microbiome varies in association with a maternal high-fat diet. Genome Med 8, 77.CrossRefGoogle ScholarPubMed
Mandal, S, Godfrey, KM, McDonald, D, et al. (2016) Fat and vitamin intakes during pregnancy have stronger relations with a pro-inflammatory maternal microbiota than does carbohydrate intake. Microbiome 4, 55.CrossRefGoogle ScholarPubMed
Roytio, H, Mokkala, K, Vahlberg, T, et al. (2017) Dietary intake of fat and fibre according to reference values relates to higher gut microbiota richness in overweight pregnant women. Br J Nutr 118, 343352.CrossRefGoogle ScholarPubMed
Lundgren, SN, Madan, JC, Emond, JA, et al. (2018) Maternal diet during pregnancy is related with the infant stool microbiome in a delivery mode-dependent manner. Microbiome 6, 109.CrossRefGoogle Scholar
Barrett, HL, Gomez-Arango, LF, Wilkinson, SA, et al. (2018) A vegetarian diet is a major determinant of gut microbiota composition in early pregnancy. Nutrients 10, 890.CrossRefGoogle Scholar
Gomez-Arango, LF, Barrett, HL, Wilkinson, SA, et al. (2018) Low dietary fiber intake increases Collinsella abundance in the gut microbiota of overweight and obese pregnant women. Gut Microbes 9, 189201.CrossRefGoogle ScholarPubMed
Laitinen, K & Mokkala, K (2019) Overall dietary quality relates to gut microbiota diversity and abundance. Int J Mol Sci 20, 1835.CrossRefGoogle ScholarPubMed
Sterne, JA, Hernan, MA, Reeves, B, et al. (2016) ROBINS-I: a tool for assessing risk of bias in non-randomized studies of interventions. BMJ 355, i4919.CrossRefGoogle Scholar
Borre, YE, O’Keeffe, GW, Clarke, G, et al. (2014) Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med 20, 509518.CrossRefGoogle ScholarPubMed
Riaz Rajoka, MS, Shi, J, Mehwish, HM, et al. (2017) Interaction between diet composition and gut microbiota and its impact on gastrointestinal tract health. Food Sci Human Wellness 6, 121130.CrossRefGoogle Scholar
Silvester, KR, Englyst, HN & Cummings, JH (1995) Ileal recovery of starch from whole diets containing resistant starch measured in vitro and fermentation of ileal effluent. Am J Clin Nutr 62, 403411.CrossRefGoogle ScholarPubMed
Rowland, I, Gibson, G, Heinken, A, et al. (2018) Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr 57, 124.CrossRefGoogle ScholarPubMed
European Food Safety Authority (2017) Dietary reference values for nutrients summary report. EFSA Support Publ 14, e15121E.Google Scholar
den Besten, G, van Eunen, K, Groen, AK, et al. (2013) The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 54, 23252340.CrossRefGoogle ScholarPubMed
Lindsay, KL, Brennan, L, Kennelly, MA, et al. (2015) Impact of probiotics in women with gestational diabetes mellitus on metabolic health: a randomized controlled trial. Am J Obstet Gynecol 212, 496.e1496.e11.CrossRefGoogle ScholarPubMed
Jarde, A, Lewis-Mikhael, AM, Moayyedi, P, et al. (2018) Pregnancy outcomes in women taking probiotics or prebiotics: a systematic review and meta-analysis. BMC Pregnancy Childb 18, 14.CrossRefGoogle ScholarPubMed
Tun, HM, Konya, T, Takaro, TK, et al. (2017) Exposure to household furry pets influences the gut microbiota of infant at 3–4 months following various birth scenarios. Microbiome 5, 40.CrossRefGoogle ScholarPubMed
Natamba, BK, Sanchez, SE, Gelaye, B, et al. (2016) Concordance between self-reported pre-pregnancy body mass index (BMI) and BMI measured at the first prenatal study contact. BMC Pregnancy Childb 16, 187.CrossRefGoogle ScholarPubMed
Koutsos, A, Tuohy, KM & Lovegrove, JA (2015) Apples and cardiovascular health – is the gut microbiota a core consideration? Nutrients 7, 39593998.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Flow diagram of study selection. Flow diagram depicting each stage of the study identification process. PICO, population, comparison, intervention and outcome.

Figure 1

Table 1. Summary of results

Figure 2

Table 2. ROBINS-I (Risk Of Bias In Non-randomised Studies – of Interventions) risk of bias results