Skip to main content
Top
Published in: Journal of Translational Medicine 1/2023

Open Access 01-12-2023 | Respiratory Microbiota | Research

Unveiling the dynamics of the breast milk microbiome: impact of lactation stage and gestational age

Authors: Parul Singh, Noora Al Mohannadi, Selvasankar Murugesan, Fajr Almarzooqi, Basirudeen Syed Ahamed Kabeer, Alexandra Katharina Marr, Tomoshige Kino, Tobias Brummaier, Annalisa Terranegra, Rose McGready, François Nosten, Damien Chaussabel, Souhaila Al Khodor

Published in: Journal of Translational Medicine | Issue 1/2023

Login to get access

Abstract

Background

Breast milk (BM) provides complete nutrition for infants for the first six months of life and is essential for the development of the newborn’s immature immune and digestive systems. While BM was conventionally believed to be sterile, recent advanced high throughput technologies have unveiled the presence of diverse microbial communities in BM. These insights into the BM microbiota have mainly originated from uncomplicated pregnancies, possibly not reflecting the circumstances of mothers with pregnancy complications like preterm birth (PTB).

Methods

In this article, we investigated the BM microbial communities in mothers with preterm deliveries (before 37 weeks of gestation). We compared these samples with BM samples from healthy term pregnancies across different lactation stages (colostrum, transitional and mature milk) using 16S rRNA gene sequencing.

Results

Our analysis revealed that the microbial communities became increasingly diverse and compositionally distinct as the BM matured. Specifically, mature BM samples were significantly enriched in Veillonella and lactobacillus (Kruskal Wallis; p < 0.001) compared to colostrum. The comparison of term and preterm BM samples showed that the community structure was significantly different between the two groups (Bray Curtis and unweighted unifrac dissimilarity; p < 0.001). Preterm BM samples exhibited increased species richness with significantly higher abundance of Staphylococcus haemolyticus, Propionibacterium acnes, unclassified Corynebacterium species. Whereas term samples were enriched in Staphylococcus epidermidis, unclassified OD1, and unclassified Veillonella among others.

Conclusion

Our study underscores the significant influence of pregnancy-related complications, such as preterm birth (before 37 weeks of gestation), on the composition and diversity of BM microbiota. Given the established significance of the maternal microbiome in shaping child health outcomes, this investigation paves the way for identifying modifiable factors that could optimize the composition of BM microbiota, thereby promoting maternal and infant health.
Appendix
Available only for authorised users
Literature
1.
go back to reference Victora CG, et al. Breastfeeding in the 21st century: epidemiology, mechanisms, and lifelong effect. Lancet. 2016;387(10017):475–90.PubMedCrossRef Victora CG, et al. Breastfeeding in the 21st century: epidemiology, mechanisms, and lifelong effect. Lancet. 2016;387(10017):475–90.PubMedCrossRef
2.
go back to reference WHO and UNICEF issue new guidance to promote breastfeeding in health facilities globally [press release]. (2018). 2018. WHO and UNICEF issue new guidance to promote breastfeeding in health facilities globally [press release]. (2018). 2018.
3.
5.
go back to reference Martín R, et al. Human milk is a source of lactic acid bacteria for the infant gut. J Pediatr. 2003;143(6):754–8.PubMedCrossRef Martín R, et al. Human milk is a source of lactic acid bacteria for the infant gut. J Pediatr. 2003;143(6):754–8.PubMedCrossRef
6.
go back to reference Lackey KA, et al. What’s normal? Microbiomes in human milk and infant feces are related to each other but vary geographically: the INSPIRE study. Front Nutr. 2019;6:45.PubMedPubMedCentralCrossRef Lackey KA, et al. What’s normal? Microbiomes in human milk and infant feces are related to each other but vary geographically: the INSPIRE study. Front Nutr. 2019;6:45.PubMedPubMedCentralCrossRef
7.
8.
go back to reference Aakko J, et al. Human milk oligosaccharide categories define the microbiota composition in human colostrum. Benef Microbes. 2017;8(4):563–7.PubMedCrossRef Aakko J, et al. Human milk oligosaccharide categories define the microbiota composition in human colostrum. Benef Microbes. 2017;8(4):563–7.PubMedCrossRef
9.
go back to reference Fernández L, et al. The human milk microbiota: origin and potential roles in health and disease. Pharmacol Res. 2013;69(1):1–10.PubMedCrossRef Fernández L, et al. The human milk microbiota: origin and potential roles in health and disease. Pharmacol Res. 2013;69(1):1–10.PubMedCrossRef
10.
go back to reference LopezLeyva L, Brereton NJB, Koski KG. Emerging frontiers in human milk microbiome research and suggested primers for 16S rRNA gene analysis. Comput Struct Biotechnol J. 2021;19:121–33.CrossRef LopezLeyva L, Brereton NJB, Koski KG. Emerging frontiers in human milk microbiome research and suggested primers for 16S rRNA gene analysis. Comput Struct Biotechnol J. 2021;19:121–33.CrossRef
11.
12.
go back to reference Fehr K, et al. Breastmilk feeding practices are associated with the co-occurrence of bacteria in mothers’ milk and the infant gut: the CHILD cohort study. Cell Host Microbe. 2020;28(2):285-297.e4.PubMedCrossRef Fehr K, et al. Breastmilk feeding practices are associated with the co-occurrence of bacteria in mothers’ milk and the infant gut: the CHILD cohort study. Cell Host Microbe. 2020;28(2):285-297.e4.PubMedCrossRef
13.
go back to reference Cortes-Macías E, et al. Maternal diet shapes the breast milk microbiota composition and diversity: impact of mode of delivery and antibiotic exposure. J Nutr. 2021;151(2):330–40.PubMedCrossRef Cortes-Macías E, et al. Maternal diet shapes the breast milk microbiota composition and diversity: impact of mode of delivery and antibiotic exposure. J Nutr. 2021;151(2):330–40.PubMedCrossRef
14.
go back to reference Ballard O, Morrow AL. Human milk composition: nutrients and bioactive factors. Pediatr Clin. 2013;60(1):49–74. Ballard O, Morrow AL. Human milk composition: nutrients and bioactive factors. Pediatr Clin. 2013;60(1):49–74.
15.
go back to reference Khodayar-Pardo P, et al. Impact of lactation stage, gestational age and mode of delivery on breast milk microbiota. J Perinatol. 2014;34(8):599–605.PubMedCrossRef Khodayar-Pardo P, et al. Impact of lactation stage, gestational age and mode of delivery on breast milk microbiota. J Perinatol. 2014;34(8):599–605.PubMedCrossRef
16.
go back to reference Gidrewicz DA, Fenton TR. A systematic review and meta-analysis of the nutrient content of preterm and term breast milk. BMC Pediatr. 2014;14(1):1–14.CrossRef Gidrewicz DA, Fenton TR. A systematic review and meta-analysis of the nutrient content of preterm and term breast milk. BMC Pediatr. 2014;14(1):1–14.CrossRef
18.
go back to reference Biagi E, et al. Microbial community dynamics in mother’s milk and infant’s mouth and gut in moderately preterm infants. Front Microbiol. 2018;9:2512.PubMedPubMedCentralCrossRef Biagi E, et al. Microbial community dynamics in mother’s milk and infant’s mouth and gut in moderately preterm infants. Front Microbiol. 2018;9:2512.PubMedPubMedCentralCrossRef
20.
21.
go back to reference Asbury MR, et al. Mothers of preterm infants have individualized breast milk microbiota that changes temporally based on maternal characteristics. Cell Host Microbe. 2020;28(5):669-682.e4.PubMedCrossRef Asbury MR, et al. Mothers of preterm infants have individualized breast milk microbiota that changes temporally based on maternal characteristics. Cell Host Microbe. 2020;28(5):669-682.e4.PubMedCrossRef
22.
go back to reference Moossavi S, et al. Composition and variation of the human milk microbiota are influenced by maternal and early-life factors. Cell Host Microbe. 2019;25(2):324-335.e4.PubMedCrossRef Moossavi S, et al. Composition and variation of the human milk microbiota are influenced by maternal and early-life factors. Cell Host Microbe. 2019;25(2):324-335.e4.PubMedCrossRef
23.
go back to reference Brummaier T, et al. Cohort profile: molecular signature in pregnancy (MSP): longitudinal high-frequency sampling to characterise cross-omic trajectories in pregnancy in a resource-constrained setting. BMJ Open. 2020;10(10): e041631.PubMedPubMedCentralCrossRef Brummaier T, et al. Cohort profile: molecular signature in pregnancy (MSP): longitudinal high-frequency sampling to characterise cross-omic trajectories in pregnancy in a resource-constrained setting. BMJ Open. 2020;10(10): e041631.PubMedPubMedCentralCrossRef
24.
go back to reference Brummaier T, et al. A prospective cohort for the investigation of alteration in temporal transcriptional and microbiome trajectories preceding preterm birth: a study protocol. BMJ Open. 2019;9(1): e023417.PubMedPubMedCentralCrossRef Brummaier T, et al. A prospective cohort for the investigation of alteration in temporal transcriptional and microbiome trajectories preceding preterm birth: a study protocol. BMJ Open. 2019;9(1): e023417.PubMedPubMedCentralCrossRef
25.
26.
29.
go back to reference Team RC. R. (R foundation for statistical computing Vienna, Austria, 2013). Team RC. R. (R foundation for statistical computing Vienna, Austria, 2013).
30.
go back to reference Chao A. Estimating the population size for capture-recapture data with unequal catchability. Biometrics. 1987;43:783–91.PubMedCrossRef Chao A. Estimating the population size for capture-recapture data with unequal catchability. Biometrics. 1987;43:783–91.PubMedCrossRef
31.
go back to reference Shannon CE. A mathematical theory of communication. Bell Syst Techn J. 1948;27(3):379–423.CrossRef Shannon CE. A mathematical theory of communication. Bell Syst Techn J. 1948;27(3):379–423.CrossRef
32.
33.
go back to reference Pielou EC. The measurement of diversity in different types of biological collections. J Theor Biol. 1966;13:131–44.CrossRef Pielou EC. The measurement of diversity in different types of biological collections. J Theor Biol. 1966;13:131–44.CrossRef
34.
go back to reference Faith DP. Conservation evaluation and phylogenetic diversity. Biol Cons. 1992;61(1):1–10.CrossRef Faith DP. Conservation evaluation and phylogenetic diversity. Biol Cons. 1992;61(1):1–10.CrossRef
37.
go back to reference DeSantis TZ, et al. Greengenes, a Chimera-Checked 16S rRNA Gene Database and Workbench Compatible with ARB. Appl Environ Microbiol. 2006;72(7):5069–72.PubMedPubMedCentralCrossRef DeSantis TZ, et al. Greengenes, a Chimera-Checked 16S rRNA Gene Database and Workbench Compatible with ARB. Appl Environ Microbiol. 2006;72(7):5069–72.PubMedPubMedCentralCrossRef
38.
go back to reference Liu C, et al. Microeco: an R package for data mining in microbial community ecology. FEMS Microbiol Ecol. 2021;97(2):255.CrossRef Liu C, et al. Microeco: an R package for data mining in microbial community ecology. FEMS Microbiol Ecol. 2021;97(2):255.CrossRef
40.
go back to reference Pannaraj PS, et al. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. 2017;171(7):647–54.PubMedPubMedCentralCrossRef Pannaraj PS, et al. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. 2017;171(7):647–54.PubMedPubMedCentralCrossRef
42.
go back to reference Jost T, et al. Stability of the maternal gut microbiota during late pregnancy and early lactation. Curr Microbiol. 2014;68(4):419–27.PubMedCrossRef Jost T, et al. Stability of the maternal gut microbiota during late pregnancy and early lactation. Curr Microbiol. 2014;68(4):419–27.PubMedCrossRef
43.
go back to reference Jiménez E, et al. Metagenomic analysis of milk of healthy and mastitis-suffering women. J Hum Lact. 2015;31(3):406–15.PubMedCrossRef Jiménez E, et al. Metagenomic analysis of milk of healthy and mastitis-suffering women. J Hum Lact. 2015;31(3):406–15.PubMedCrossRef
44.
45.
go back to reference Togo A, et al. Repertoire of human breast and milk microbiota: a systematic review. Future Microbiol. 2019;14:623–41.PubMedCrossRef Togo A, et al. Repertoire of human breast and milk microbiota: a systematic review. Future Microbiol. 2019;14:623–41.PubMedCrossRef
46.
go back to reference Gueimonde M, et al. Breast milk: a source of bifidobacteria for infant gut development and maturation? Biol Neonate. 2007;92(1):64–6.CrossRef Gueimonde M, et al. Breast milk: a source of bifidobacteria for infant gut development and maturation? Biol Neonate. 2007;92(1):64–6.CrossRef
47.
go back to reference Martín R, et al. Diversity of the Lactobacillus group in breast milk and vagina of healthy women and potential role in the colonization of the infant gut. J Appl Microbiol. 2007;103(6):2638–44.PubMedCrossRef Martín R, et al. Diversity of the Lactobacillus group in breast milk and vagina of healthy women and potential role in the colonization of the infant gut. J Appl Microbiol. 2007;103(6):2638–44.PubMedCrossRef
48.
go back to reference Delgado S, et al. PCR-DGGE assessment of the bacterial diversity of breast milk in women with lactational infectious mastitis. BMC Infect Dis. 2008;8(1):1–8.CrossRef Delgado S, et al. PCR-DGGE assessment of the bacterial diversity of breast milk in women with lactational infectious mastitis. BMC Infect Dis. 2008;8(1):1–8.CrossRef
49.
go back to reference Collado M, et al. Assessment of the bacterial diversity of breast milk of healthy women by quantitative real-time PCR. Lett Appl Microbiol. 2009;48(5):523–8.PubMedCrossRef Collado M, et al. Assessment of the bacterial diversity of breast milk of healthy women by quantitative real-time PCR. Lett Appl Microbiol. 2009;48(5):523–8.PubMedCrossRef
50.
go back to reference Ward TL, et al. Human milk metagenome: a functional capacity analysis. BMC Microbiol. 2013;13:1–12.CrossRef Ward TL, et al. Human milk metagenome: a functional capacity analysis. BMC Microbiol. 2013;13:1–12.CrossRef
51.
go back to reference Rodríguez JM. The origin of human milk bacteria: is there a bacterial entero-mammary pathway during late pregnancy and lactation? Adv Nutr. 2014;5(6):779–84.PubMedPubMedCentralCrossRef Rodríguez JM. The origin of human milk bacteria: is there a bacterial entero-mammary pathway during late pregnancy and lactation? Adv Nutr. 2014;5(6):779–84.PubMedPubMedCentralCrossRef
52.
go back to reference Goldman AS. The immune system of human milk: antimicrobial, antiinflammatory and immunomodulating properties. Pediatr Infect Dis J. 1993;12(8):664–71.PubMedCrossRef Goldman AS. The immune system of human milk: antimicrobial, antiinflammatory and immunomodulating properties. Pediatr Infect Dis J. 1993;12(8):664–71.PubMedCrossRef
53.
go back to reference Ramsay DT, et al. Ultrasound imaging of milk ejection in the breast of lactating women. Pediatrics. 2004;113(2):361–7.PubMedCrossRef Ramsay DT, et al. Ultrasound imaging of milk ejection in the breast of lactating women. Pediatrics. 2004;113(2):361–7.PubMedCrossRef
56.
go back to reference Cabrera-Rubio R, et al. The human milk microbiome changes over lactation and is shaped by maternal weight and mode of delivery. Am J Clin Nutr. 2012;96(3):544–51.PubMedCrossRef Cabrera-Rubio R, et al. The human milk microbiome changes over lactation and is shaped by maternal weight and mode of delivery. Am J Clin Nutr. 2012;96(3):544–51.PubMedCrossRef
57.
go back to reference Fitzstevens JL, et al. Systematic Review of the Human Milk Microbiota. Nutr Clin Pract. 2017;32(3):354–64.PubMedCrossRef Fitzstevens JL, et al. Systematic Review of the Human Milk Microbiota. Nutr Clin Pract. 2017;32(3):354–64.PubMedCrossRef
58.
go back to reference Sanjulián L, et al. Bacterial diversity of breast milk in healthy spanish women: evolution from birth to five years postpartum. Nutrients. 2021;13(7):2414.PubMedPubMedCentralCrossRef Sanjulián L, et al. Bacterial diversity of breast milk in healthy spanish women: evolution from birth to five years postpartum. Nutrients. 2021;13(7):2414.PubMedPubMedCentralCrossRef
59.
go back to reference Martín R, et al. Cultivation-independent assessment of the bacterial diversity of breast milk among healthy women. Res Microbiol. 2007;158(1):31–7.PubMedCrossRef Martín R, et al. Cultivation-independent assessment of the bacterial diversity of breast milk among healthy women. Res Microbiol. 2007;158(1):31–7.PubMedCrossRef
61.
go back to reference Tian R, et al. Small and mighty: adaptation of superphylum Patescibacteria to groundwater environment drives their genome simplicity. Microbiome. 2020;8(1):51.PubMedPubMedCentralCrossRef Tian R, et al. Small and mighty: adaptation of superphylum Patescibacteria to groundwater environment drives their genome simplicity. Microbiome. 2020;8(1):51.PubMedPubMedCentralCrossRef
62.
go back to reference Murray NJ, et al. Myanmar’s terrestrial ecosystems: status, threats and conservation opportunities. BioRxiv. 2020;252:108834. Murray NJ, et al. Myanmar’s terrestrial ecosystems: status, threats and conservation opportunities. BioRxiv. 2020;252:108834.
63.
go back to reference Arunyawat S, Shrestha RP. Simulating future land use and ecosystem services in Northern Thailand. J Land Use Sci. 2018;13(1–2):146–65.CrossRef Arunyawat S, Shrestha RP. Simulating future land use and ecosystem services in Northern Thailand. J Land Use Sci. 2018;13(1–2):146–65.CrossRef
64.
go back to reference Jost T, et al. Assessment of bacterial diversity in breast milk using culture-dependent and culture-independent approaches. Br J Nutr. 2013;110(7):1253–62.PubMedCrossRef Jost T, et al. Assessment of bacterial diversity in breast milk using culture-dependent and culture-independent approaches. Br J Nutr. 2013;110(7):1253–62.PubMedCrossRef
65.
go back to reference Chen P-W, Lin Y-L, Huang M-S. Profiles of commensal and opportunistic bacteria in human milk from healthy donors in Taiwan. J Food Drug Anal. 2018;26(4):1235–44.PubMedPubMedCentralCrossRef Chen P-W, Lin Y-L, Huang M-S. Profiles of commensal and opportunistic bacteria in human milk from healthy donors in Taiwan. J Food Drug Anal. 2018;26(4):1235–44.PubMedPubMedCentralCrossRef
68.
go back to reference Dzidic M, et al. Gut microbiota and mucosal immunity in the neonate. Med Sci. 2018;6(3):56. Dzidic M, et al. Gut microbiota and mucosal immunity in the neonate. Med Sci. 2018;6(3):56.
70.
go back to reference Jiang T, Savaiano DA. In vitro lactose fermentation by human colonic bacteria is modified by lactobacillus acidophilus supplementation. J Nutr. 1997;127(8):1489–95.PubMedCrossRef Jiang T, Savaiano DA. In vitro lactose fermentation by human colonic bacteria is modified by lactobacillus acidophilus supplementation. J Nutr. 1997;127(8):1489–95.PubMedCrossRef
71.
go back to reference Piwowarek K, et al. Propionibacterium spp.-source of propionic acid, vitamin B12, and other metabolites important for the industry. Appl Microbiol Biotechnol. 2018;102(2):515–38.PubMedCrossRef Piwowarek K, et al. Propionibacterium spp.-source of propionic acid, vitamin B12, and other metabolites important for the industry. Appl Microbiol Biotechnol. 2018;102(2):515–38.PubMedCrossRef
72.
73.
go back to reference Murphy K, et al. The composition of human milk and infant faecal microbiota over the first three months of life: a pilot study. Sci Rep. 2017;7(1):40597.PubMedPubMedCentralCrossRef Murphy K, et al. The composition of human milk and infant faecal microbiota over the first three months of life: a pilot study. Sci Rep. 2017;7(1):40597.PubMedPubMedCentralCrossRef
74.
go back to reference Kunz C, et al. Influence of gestational age, secretor, and lewis blood group status on the oligosaccharide content of human milk. J Pediat Gastroenterol Nutrit. 2017;64(5):789–98.CrossRef Kunz C, et al. Influence of gestational age, secretor, and lewis blood group status on the oligosaccharide content of human milk. J Pediat Gastroenterol Nutrit. 2017;64(5):789–98.CrossRef
75.
go back to reference Zeng S, et al. First 1000 days and beyond after birth: gut microbiota and necrotizing enterocolitis in preterm infants. Front Microbiol. 2022;13:905380.PubMedPubMedCentralCrossRef Zeng S, et al. First 1000 days and beyond after birth: gut microbiota and necrotizing enterocolitis in preterm infants. Front Microbiol. 2022;13:905380.PubMedPubMedCentralCrossRef
76.
go back to reference Bauer J, Gerss J. Variability in human milk composition: benefit of individualized fortification in very-low-birth-weight infants. Clin Nutr. 2011;30(2):215–20.PubMedCrossRef Bauer J, Gerss J. Variability in human milk composition: benefit of individualized fortification in very-low-birth-weight infants. Clin Nutr. 2011;30(2):215–20.PubMedCrossRef
77.
go back to reference Gidrewicz DA, Fenton TR. A systematic review and meta-analysis of the nutrient content of preterm and term breast milk. BMC Pediatr. 2014;14:1–14.CrossRef Gidrewicz DA, Fenton TR. A systematic review and meta-analysis of the nutrient content of preterm and term breast milk. BMC Pediatr. 2014;14:1–14.CrossRef
78.
79.
go back to reference Mehta R, Petrova A. Biologically active breast milk proteins in association with very preterm delivery and stage of lactation. J Perinatol. 2011;31(1):58–62.PubMedCrossRef Mehta R, Petrova A. Biologically active breast milk proteins in association with very preterm delivery and stage of lactation. J Perinatol. 2011;31(1):58–62.PubMedCrossRef
80.
go back to reference Koenig Á, et al. Immunologic factors in human milk: the effects of gestational age and pasteurization. J Hum Lact. 2005;21(4):439–43.PubMedCrossRef Koenig Á, et al. Immunologic factors in human milk: the effects of gestational age and pasteurization. J Hum Lact. 2005;21(4):439–43.PubMedCrossRef
81.
go back to reference Garcia C, et al. Bioactive compounds in human milk and intestinal health and maturity in preterm newborn: an overview. Cell Mol Biol. 2013;59:108–31.PubMed Garcia C, et al. Bioactive compounds in human milk and intestinal health and maturity in preterm newborn: an overview. Cell Mol Biol. 2013;59:108–31.PubMed
82.
go back to reference Dahl C, et al. Gut microbiome of mothers delivering prematurely shows reduced diversity and lower relative abundance of Bifidobacterium and Streptococcus. PLoS ONE. 2017;12(10): e0184336.PubMedPubMedCentralCrossRef Dahl C, et al. Gut microbiome of mothers delivering prematurely shows reduced diversity and lower relative abundance of Bifidobacterium and Streptococcus. PLoS ONE. 2017;12(10): e0184336.PubMedPubMedCentralCrossRef
83.
84.
go back to reference Bianchi-Jassir F, et al. Preterm birth associated with group b streptococcus maternal colonization worldwide: systematic review and meta-analyses. Clin Infect Dis. 2017;65(2):S133-s142.PubMedPubMedCentralCrossRef Bianchi-Jassir F, et al. Preterm birth associated with group b streptococcus maternal colonization worldwide: systematic review and meta-analyses. Clin Infect Dis. 2017;65(2):S133-s142.PubMedPubMedCentralCrossRef
85.
go back to reference Turner C, et al. Group B streptococcal carriage, serotype distribution and antibiotic susceptibilities in pregnant women at the time of delivery in a refugee population on the Thai-Myanmar border. BMC Infect Dis. 2012;12(1):34.PubMedPubMedCentralCrossRef Turner C, et al. Group B streptococcal carriage, serotype distribution and antibiotic susceptibilities in pregnant women at the time of delivery in a refugee population on the Thai-Myanmar border. BMC Infect Dis. 2012;12(1):34.PubMedPubMedCentralCrossRef
87.
go back to reference Stoeva MK, et al. Butyrate-producing human gut symbiont, Clostridium butyricum, and its role in health and disease. Gut Microbes. 2021;13(1):1–28.PubMedCrossRef Stoeva MK, et al. Butyrate-producing human gut symbiont, Clostridium butyricum, and its role in health and disease. Gut Microbes. 2021;13(1):1–28.PubMedCrossRef
88.
go back to reference Leonel AJ, Alvarez-Leite JI. Butyrate: implications for intestinal function. Curr Opin Clin Nutr Metab Care. 2012;15(5):474–9.PubMedCrossRef Leonel AJ, Alvarez-Leite JI. Butyrate: implications for intestinal function. Curr Opin Clin Nutr Metab Care. 2012;15(5):474–9.PubMedCrossRef
89.
go back to reference Soeorg H, et al. Genetic relatedness of Staphylococcus haemolyticus in gut and skin of preterm neonates and breast milk of their mothers. Pediatric Infect Dis J. 2019;38(3):308–13.CrossRef Soeorg H, et al. Genetic relatedness of Staphylococcus haemolyticus in gut and skin of preterm neonates and breast milk of their mothers. Pediatric Infect Dis J. 2019;38(3):308–13.CrossRef
90.
go back to reference Arboleya S, et al. Establishment and development of intestinal microbiota in preterm neonates. FEMS Microbiol Ecol. 2012;79(3):763–72.PubMedCrossRef Arboleya S, et al. Establishment and development of intestinal microbiota in preterm neonates. FEMS Microbiol Ecol. 2012;79(3):763–72.PubMedCrossRef
91.
go back to reference Maestre FT, et al. Species richness effects on ecosystem multifunctionality depend on evenness, composition and spatial pattern. J Ecol. 2012;100(2):317–30.CrossRef Maestre FT, et al. Species richness effects on ecosystem multifunctionality depend on evenness, composition and spatial pattern. J Ecol. 2012;100(2):317–30.CrossRef
92.
go back to reference Suez J, et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell. 2018;174(6):1406-1423. e16.PubMedCrossRef Suez J, et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell. 2018;174(6):1406-1423. e16.PubMedCrossRef
93.
go back to reference Di Renzo GC, et al. Intrapartum GBS screening and antibiotic prophylaxis: a European consensus conference. J Matern Fetal Neonatal Med. 2015;28(7):766–82.PubMedCrossRef Di Renzo GC, et al. Intrapartum GBS screening and antibiotic prophylaxis: a European consensus conference. J Matern Fetal Neonatal Med. 2015;28(7):766–82.PubMedCrossRef
94.
go back to reference Mueller NT, et al. Prenatal exposure to antibiotics, cesarean section and risk of childhood obesity. Int J Obes. 2015;39(4):665–70.CrossRef Mueller NT, et al. Prenatal exposure to antibiotics, cesarean section and risk of childhood obesity. Int J Obes. 2015;39(4):665–70.CrossRef
95.
go back to reference Ong M-S, Umetsu DT, Mandl KD. Consequences of antibiotics and infections in infancy: bugs, drugs, and wheezing. Ann Allergy Asthma Immunol. 2014;112(5):441–4451.PubMedCrossRef Ong M-S, Umetsu DT, Mandl KD. Consequences of antibiotics and infections in infancy: bugs, drugs, and wheezing. Ann Allergy Asthma Immunol. 2014;112(5):441–4451.PubMedCrossRef
96.
go back to reference Ungaro R, et al. Antibiotics associated with increased risk of new-onset Crohn’s disease but not ulcerative colitis: a meta-analysis. Official J Am Coll Gastroenterol. 2014;109(11):1728–38.CrossRef Ungaro R, et al. Antibiotics associated with increased risk of new-onset Crohn’s disease but not ulcerative colitis: a meta-analysis. Official J Am Coll Gastroenterol. 2014;109(11):1728–38.CrossRef
Metadata
Title
Unveiling the dynamics of the breast milk microbiome: impact of lactation stage and gestational age
Authors
Parul Singh
Noora Al Mohannadi
Selvasankar Murugesan
Fajr Almarzooqi
Basirudeen Syed Ahamed Kabeer
Alexandra Katharina Marr
Tomoshige Kino
Tobias Brummaier
Annalisa Terranegra
Rose McGready
François Nosten
Damien Chaussabel
Souhaila Al Khodor
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2023
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-023-04656-9

Other articles of this Issue 1/2023

Journal of Translational Medicine 1/2023 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.