Next Article in Journal
Implications of Dietary Guidance for Sport and Exercise
Previous Article in Journal
Dietary Habits and Metabolic Health
Previous Article in Special Issue
Prebiotic Supplementation during Lactation Affects Microbial Colonization in Postnatal-Growth-Restricted Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Effect of Pre- and Perinatal Factors and Infant Nutrition on the Intestinal Microbiota

by
Beata Łoniewska
1 and
Igor Łoniewski
2,3,4,*
1
Department of Neonatology and Intensive Neonatal Care, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
2
Department of Biochemical Science, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
3
Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
4
Sanprobi sp. z o.o. sp. k., 70-535 Szczecin, Poland
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(18), 3977; https://doi.org/10.3390/nu15183977
Submission received: 1 September 2023 / Accepted: 10 September 2023 / Published: 14 September 2023
The intestinal microbiota is an essential determinant of human health. It is responsible for food digestion and plays a significant role in human development and metabolic, immunological, and psychological processes. Research on the gut microbiota of infants and young children still needs to be completed. Children are already in contact with microorganisms in foetal life, and these interactions increase rapidly after birth and during breastfeeding. The increasing number of caesarean section births, the application of antibiotics and other drugs, and formula feeding undoubtedly affect the baby’s microbiome and may influence future health. Therefore, studying factors that may affect the microbiome, the immune system, or the intestinal barrier in early life is essential. It is also important to implement early prevention and modification of the microbiome to prevent adverse consequences from its alterations. In this Special Issue, five articles were published that analysed the influence of various factors during pregnancy, childbirth, and the first years of life on the composition and metabolic functions of the gut microbiota. The gut microbiome changes composition and function during the first 2–3 years of life [1,2]. The most crucial factor involved in the maturation of the microbiome is nutrition. Breast milk feeding has a more favourable effect on the maturation of the microbiome than formula feeding [3,4,5,6]. Infant gut bacterial colonisation is also affected by other factors, including caesarean section [7], antibiotics treatment [8], and maternal body mass index (BMI) [9]. Like with formula feeding, these factors are associated with early gut maturation, which can have an unfavourable effect on the development of the immune system [10,11] and can increase the risk of obesity [9,12]. An essential function of the microbiota is synthesising metabolites, including short-chain fatty acids (SCFAs) [13]. The most important are acetate (C2), propionate (C3), and butyrate (C4) [14]. SCFAs play an important role as a source of energy for various cells [15] and take part in gluconeogenesis and lipogenesis. The aim of the work of Łoniewska et al. [16] was to analyse the influence of perinatal factors, which can affect the gut microbiota, on the concentration of SCFAs in the stool during the first two years of life. The results show dynamic changes in the stool concentrations of SCFAs in children during the first year of life, followed by the stabilisation of their stool concentrations. In addition, the maternal and foetal factors studied, such as antibiotic intake during pregnancy, feeding method, delivery method, pre-pregnancy weight, changes in gestational weight, and children’s weight and sex, influenced the excretion of SCFAs in the stool. However, the study could not determine the long-term health consequences of these observations.
Nevertheless, it can be speculated that breastfeeding and proper weight control in mothers and children benefit health by influencing the composition of SCFAs in the stool and, thus, the development of the gut microbiota. In contrast, antibiotic therapy during pregnancy and caesarean section may harm the functioning of the gut microbiota. However, these speculations should be treated cautiously, and long-term epidemiological studies are warranted to further determine the mechanisms of these changes.
One of the predominant SCFAs—propionate—has antihypertensive properties. In a study on rats, Tain et al. [17] showed that perinatal propionate supplementation in offspring could prevent hypertension induced by the mother’s chronic kidney disease (CKD). The protective effect of propionate was associated with an increased abundance of Clostridium spp. bacteria (which produce propionate) and propionate concentrations in the animals’ plasma, an increased expression of renal G protein-coupled receptor 41 (GPR41, SCFA receptor), increased α-diversity, and changes in the intestinal microbiota composition. The results indicate that CKD-induced hypertension in the offspring can be prevented by SCFA metabolites in early life, which may be helpful for further research on this subject.
Another experimental study analysed the impact of the mother’s metabolism during early pregnancy, which can affect the child’s emotional and behavioural processes, including vulnerability to bullying. This phenomenon results in social anxiety and increases the risk of personality disorders in bullies and their victims. The substrate of this phenomenon is not precisely known, and the influence of genetic, gestational, and environmental factors is suggested [18]. It has been observed that children born to stressed mothers are more prone to bullying [19], which may be associated with increased susceptibility to stress in people with gut microbiota disorders [20]. One of the factors influencing gut microbiota disorders in children and brain development is placental tryptophan fluctuations, which can lead to adverse physiological and socio-behavioural consequences. Huang et al. [21] investigated the hypothesis that the administration of tryptophan during pregnancy could affect children’s mental and physical development by modifying the microbiota–gut–brain axis (MGB), which underlies their mental state. Chicken embryos, whose microbiota are robust and independent from maternal influence, were used for this experiment. The results showed that exposure to tryptophan during the embryonic period reduced the male offspring’s body weight and aggressiveness before and during puberty. In addition, beneficial anatomical changes were found in the gut under the influence of tryptophan. The mechanism for these changes may involve tryptophan-induced reprogramming of the MGB axis, which causes changes in the composition of the intestinal microbiota that may result in increased synthesis of scatole and butyrate, as well as elevated concentrations of catecholamines in the thalamus and hypothalamus. These changes may underlie bullying and warrant further investigation into the possibility of dietary modification and tryptophan supplementation during pregnancy.
Another two studies focused on preterm infants, whose microbiota are less diverse and often enriched with potential pathogens (e.g., members of the Enterobacteriaceae family). In addition, preterm infants are often treated with antibiotics, destabilising the microbiota and increasing the risk of fungal infections. In an experimental study, dietary supplementation of preterm infants with medium-chain triglyceride (MCT) oil was shown to reduce the fungal load of Candida spp. in the gastrointestinal tract [22]. The study of Romo et al. [23] aimed to determine whether MCT supplementation affected the bacterial component of the microbiome. They found that MCT supplementation did not significantly alter the diversity or composition of the gastrointestinal microbiota. Notably, there were no significant changes in the Enterobacteriaceae family, suggesting that MCT supplementation did not enrich the abundance of potential pathogens. MCTs represent a promising therapeutic intervention to reduce fungal colonisation without significantly affecting the bacterial composition of the host gastrointestinal tract. Marousez et al. [24] investigated the impact of consuming early postnatal prebiotic fibre (PF) on growth, intestinal morphology, and the microbiota during weaning in postnatal-growth-restricted mice (PNGR). They found that the PNGR mice had a decreased body weight and ileal crypt depth during weaning compared to the control group and microbiota alterations. The propionate concentrations were also increased with PNGR. PF supplementation did not impact intestinal morphology in the PNGR pups but affected gut microbiota. Moreover, PF supplementation increased the Akkermansia genus in the PF-supplemented control pups compared to the water-supplemented ones. The authors concluded that PF supplementation might improve gut microbiota establishment during the early postnatal period.
Summarising the observed results, the microbiota has an essential metabolic function during early human development. Particular caution is warranted when using drugs, in medical procedures, and in health care during pregnancy, as all of these factors can affect it. On the other hand, early intervention to modify the microbiota in this phase of life can have long-term beneficial health effects. For this reason, despite methodological difficulties, prospective studies should be conducted to assess the distant consequences of altering the microbiota early in life.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bäckhed, F.; Roswall, J.; Peng, Y.; Feng, Q.; Jia, H.; Kovatcheva-Datchary, P.; Li, Y.; Xia, Y.; Xie, H.; Zhong, H.; et al. Dynamics and stabilisation of the human gut microbiome during the first year of life. Cell Host Microbe 2015, 17, 690–703. [Google Scholar] [CrossRef]
  2. Stewart, C.J.; Ajami, N.J.; O’brien, J.L.; Hutchinson, D.S.; Smith, D.P.; Wong, M.C.; Ross, M.C.; Lloyd, R.E.; Doddapaneni, H.; Metcalf, G.A.; et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature 2018, 562, 583–588. [Google Scholar] [CrossRef] [PubMed]
  3. Forbes, J.D.; Azad, M.B.; Vehling, L.; Tun, H.M.; Konya, T.B.; Guttman, D.S.; Field, C.J.; Lefebvre, D.; Sears, M.R.; Becker, A.B.; et al. Association of exposure to formula in the hospital and subsequent infant feeding practices with gut microbiota and risk of overweight in the first year of life. JAMA Pediatr. 2018, 172, e181161. [Google Scholar] [CrossRef]
  4. Baumann-Dudenhoeffer, A.M.; D’Souza, A.W.; Tarr, P.I.; Warner, B.B.; Dantas, G. Infant diet and maternal gestational weight gain predict early metabolic maturation of gut microbiomes. Nat. Med. 2018, 24, 1822–1829. [Google Scholar] [CrossRef]
  5. De Leoz, M.L.A.; Kalanetra, K.M.; Bokulich, N.A.; Strum, J.S.; Underwood, M.A.; German, J.B.; Mills, D.A.; Lebrilla, C.B. Human milk glycomics and gut microbial genomics in infant feces show a correlation between human milk oligosaccharides and gut microbiota: A proof-of-concept study. J. Proteome Res. 2015, 14, 491–502. [Google Scholar] [CrossRef] [PubMed]
  6. Ho, N.T.; Li, F.; Lee-Sarwar, K.A.; Tun, H.M.; Brown, B.; Pannaraj, P.S.; Bender, J.M.; Azad, M.B.; Thompson, A.L.; Weiss, S.T.; et al. Metaanalysis of effects of exclusive breastfeeding on infant gut microbiota across populations. Nat. Commun. 2018, 9, 4169. [Google Scholar] [CrossRef]
  7. Jakobsson, H.E.; Abrahamsson, T.R.; Jenmalm, M.C.; Harris, K.; Quince, C.; Jernberg, C.; Björkstén, B.; Engstrand, L.; Andersson, A.F. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut 2014, 63, 559–566. [Google Scholar] [CrossRef] [PubMed]
  8. Korpela, K.; Salonen, A.; Saxen, H.; Nikkonen, A.; Peltola, V.; Jaakkola, T.; de Vos, W.; Kolho, K.-L. Antibiotics in early life associate with specific gut microbiota signatures in a prospective longitudinal infant cohort. Pediatr. Res. 2020, 88, 438–443. [Google Scholar] [CrossRef] [PubMed]
  9. Soderborg, T.K.; Clark, S.; Mulligan, C.E.; Janssen, R.C.; Babcock, L.; Ir, D.; Young, B.; Krebs, N.; Lemas, D.J.; Johnson, L.K.; et al. The gut microbiota in infants of obese mothers increases inflammation and susceptibility to NAFLD. Nat. Commun. 2018, 9, 4462. [Google Scholar] [CrossRef]
  10. Lazar, V.; Ditu, L.-M.; Pircalabioru, G.G.; Gheorghe, I.; Curutiu, C.; Holban, A.M.; Picu, A.; Petcu, L.; Chifiriuc, M.C. Aspects of gut microbiota and immune system interactions in infectious diseases, immunopathology, and cancer. Front. Immunol. 2018, 9, 1830. [Google Scholar] [CrossRef]
  11. Olin, A.; Henckel, E.; Chen, Y.; Lakshmikanth, T.; Pou, C.; Mikes, J.; Gustafsson, A.; Bernhardsson, A.K.; Zhang, C.; Bohlin, K.; et al. stereotypic immune system development in newborn children. Cell 2018, 174, 1277–1292.e14. [Google Scholar] [CrossRef] [PubMed]
  12. Tun, H.M.; Bridgman, S.L.; Chari, R.; Field, C.J.; Guttman, D.S.; Becker, A.B.; Mandhane, P.J.; Turvey, S.E.; Subbarao, P.; Sears, M.R.; et al. Roles of birth mode and infant gut microbiota in intergenerational transmission of overweight and obesity from mother to offspring. JAMA Pediatr. 2018, 172, 368–377. [Google Scholar] [CrossRef] [PubMed]
  13. Roy, C.C.; Kien, C.L.; Bouthillier, L.; Levy, E. Short-chain fatty acids: Ready for prime time? Nutr. Clin. Pract. 2006, 21, 351–366. [Google Scholar] [CrossRef]
  14. von Martels, J.Z.; Sadabad, M.S.; Bourgonje, A.R.; Blokzijl, T.; Dijkstra, G.; Faber, K.N.; Harmsen, H.J. The role of gut microbiota in health and disease: In vitro modeling of host-microbe interactions at the aerobe-anaerobe interphase of the human gut. Anaerobe 2017, 44, 3–12. [Google Scholar] [CrossRef]
  15. Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef]
  16. Łoniewska, B.; Fraszczyk-Tousty, M.; Tousty, P.; Skonieczna-Żydecka, K.; Maciejewska-Markiewicz, D.; Łoniewski, I. Analysis of Fecal Short-Chain Fatty Acids (SCFAs) in Healthy Children during the First Two Years of Life: An Observational Prospective Cohort Study. Nutrients 2023, 15, 367. [Google Scholar] [CrossRef] [PubMed]
  17. Tain, Y.-L.; Hou, C.-Y.; Chang-Chien, G.-P.; Lin, S.-F.; Hsu, C.-N. Perinatal Propionate Supplementation Protects Adult Male Offspring from Maternal Chronic Kidney Disease-Induced Hypertension. Nutrients 2022, 14, 3435. [Google Scholar] [CrossRef]
  18. Ball, H.A.; Arseneault, L.; Taylor, A.; Maughan, B.; Caspi, A.; Moffitt, T.E. Genetic and environmental influences on victims, bullies and bully-victims in childhood. J. Child Psychol. Psychiatry 2008, 49, 104–112. [Google Scholar] [CrossRef]
  19. Lereya, S.T.; Wolke, D. Prenatal family adversity and maternal mental health and vulnerability to peer victimisation at school. J. Child Psychol. Psychiatry 2013, 54, 644–652. [Google Scholar] [CrossRef]
  20. Misera, A.; Łoniewski, I.; Palma, J.; Kulaszyńska, M.; Czarnecka, W.; Kaczmarczyk, M.; Liśkiewicz, P.; Samochowiec, J.; Skonieczna-Żydecka, K. Clinical significance of microbiota changes under the influence of psychotropic drugs. An updated narrative review. Front. Microbiol. 2023, 14, 1125022. [Google Scholar] [CrossRef]
  21. Huang, X.; Hu, J.; Peng, H.; Cheng, H.-W. Embryonic Exposure to Tryptophan Yields Bullying Victimization via Reprogramming the Microbiota-Gut-Brain Axis in a Chicken Model. Nutrients 2022, 14, 661. [Google Scholar] [CrossRef] [PubMed]
  22. Gunsalus, K.T.; Tornberg-Belanger, S.N.; Matthan, N.R.; Lichtenstein, A.H.; Kumamoto, C.A. Manipulation of Host Diet to Reduce Gastrointestinal Colonisation by the Opportunistic Pathogen Candida albicans. mSphere 2016, 1, e00020-15. [Google Scholar] [CrossRef] [PubMed]
  23. Romo, J.A.; Arsenault, A.B.; Laforce-Nesbitt, S.S.; Bliss, J.M.; Kumamoto, C.A. Minimal Effects of Medium-Chain Triglyceride Supplementation on the Intestinal Microbiome Composition of Premature Infants: A Single-Center Pilot Study. Nutrients 2022, 14, 2159. [Google Scholar] [CrossRef] [PubMed]
  24. Marousez, L.; Tran, L.C.; Micours, E.; Antoine, M.; Gottrand, F.; Lesage, J.; Ley, D. Prebiotic Supplementation during Lactation Affects Microbial Colonization in Postnatal-Growth-Restricted Mice. Nutrients 2023, 15, 2771. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Łoniewska, B.; Łoniewski, I. Effect of Pre- and Perinatal Factors and Infant Nutrition on the Intestinal Microbiota. Nutrients 2023, 15, 3977. https://doi.org/10.3390/nu15183977

AMA Style

Łoniewska B, Łoniewski I. Effect of Pre- and Perinatal Factors and Infant Nutrition on the Intestinal Microbiota. Nutrients. 2023; 15(18):3977. https://doi.org/10.3390/nu15183977

Chicago/Turabian Style

Łoniewska, Beata, and Igor Łoniewski. 2023. "Effect of Pre- and Perinatal Factors and Infant Nutrition on the Intestinal Microbiota" Nutrients 15, no. 18: 3977. https://doi.org/10.3390/nu15183977

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop