Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Oct 1.
Published in final edited form as: Semin Fetal Neonatal Med. 2017 Jul 15;22(5):284–289. doi: 10.1016/j.siny.2017.07.002

Prenatal and postnatal administration of prebiotics and probiotics

Kristin Sohn 1, Mark A Underwood 1,*
PMCID: PMC5618799  NIHMSID: NIHMS891948  PMID: 28720399

SUMMARY

Colonization of the neonatal gut by beneficial bacteria is important for the establishment and maintenance of the mucosal barrier, thus protecting the neonate from enteric pathogens and local and systemic inflammation. The neonatal microbiome is influenced by infant diet, environment, and the maternal microbiome. Dysbiosis in pregnancy increases the risk of pre-eclampsia, diabetes, infection, preterm labor, and later childhood atopy. Dysbiosis of the neonatal gut plays an important role in colic in the term infant, in the disease processes which plague preterm infants, including necrotizing enterocolitis and sepsis, and in the long-term outcomes of neonates. Administration of enteral prebiotics, probiotics, and synbiotics during pregnancy, lactation, and postnatal life appears to be a safe and feasible method to alter the maternal and neonatal microbiome, thus improving pregnancy and neonatal outcomes.

Keywords: Pregnancy, Microbiota, Prematurity, Necrotizing enterocolitis, Atopic dermatitis, Allergic rhinitis, Metabolic syndrome, Diabetes mellitus

1. Introduction

Novel approaches to analysis of microbial communities have provided evidence that changes in maternal, fetal, and neonatal microbes impact both short- and long-term outcomes. As this field is still in its infancy, we begin with some definitions. The term microbiota is used to describe the microbial community of a given anatomic or environmental niche; microbiome is widely used both as a synonym for microbiota and for the genetic material of the microbiota. We use the term dysbiosis to refer to alterations in the microbiota associated with disease, probiotic to mean a dietary supplement or drug containing live micro-organisms administered with the intent to improve health, prebiotic to mean a dietary supplement that is not digestible by the host and stimulates the growth of desirable or commensal microbes, and synbiotic to mean a dietary supplement that contains both probiotic microbes and prebiotic components. Micro-organisms can modify the human microbiota by reducing luminal pH, competing for nutrients, secreting antimicrobial compounds, preventing bacterial adhesion, and inducing antimicrobial production by the host. Table 1 presents many of the bacteria that discussed below, classified by their phylum, class, order, family, genus, and species.

Table 1.

Key bacterial taxa in pregnancy and during perinatal period.

Phylum Class Order Family Genus
Firmicutes Bacilli Bacillales Staphylococcaceae Staphylococcus
Lactobacillales Streptococcaceae Streptococcus
Enterococcaceae Enterococcus
Lactobacillaceae Lactobacillus
Clostridia Clostridiales Clostridiaceae Clostridium
Negativicutes Selenomonadales Veillonellaceae Veillonella
Tenericutes Mollicutes Mycoplasmatales Mycoplasmataceae Ureaplasma
Mycoplasma
Proteobacteria γ-Proteobacteria Enterobacteriales Enterobacteriaceae Klebsiella
Escherichia
Proteus
Serratia
Enterobacter
Cronobacter
Pseudomonadales Pseudomonadaceae Pseudomonas
Moraxellaceae Acinetobacter
α-Proteobacteria
Bacteroidetes Bacteroidetes Bacteroidales Bacteroidaceae Bacteroides
Prevotellaceae Prevotella
Actinobacteria Actinobacteria Bifidobacteriales Bifidobacteriaceae Bifidobacterium
Gardnerella
Propionibacteriales Propionibacteriaceae Propionibacterium
Coriobacteriales Coriobacteriaceae Atopobium
Actinomycetales
Fusobacteria Fusobacteria Fusobacteriales Leptotrichiaceae Leptotrichia
Sneathia

2. The maternal microbiome: changes during normal pregnancy

2.1. The vaginal microbiome

This changes from the first to the third trimester of pregnancy, with an overall decrease in microbial diversity and a shift toward predominance of Lactobacillus spp., followed by Clostridiales, Bacteroidales, and Actinomycetales [1,2]. Lactobacillus predominance appears to protect from bacterial pathogens by maintenance of a low vaginal pH through lactic acid production. The majority of vaginal microbial community state types (CSTs) are dominated by Lactobacillus species (L. crispatus, L. iners, L. Jensenii, and L. gasseri), whereas some are composed of anaerobic bacteria associated with bacterial vaginosis (e.g. Gardnerella vaginalis, Sneathia spp., Prevotella spp., Megasphaera spp., and Atopobium vaginae) and an increased risk of sexually transmitted infections, preterm birth, chorioamnionitis, and spontaneous abortion. Non-pregnant women fluctuate between CSTs, whereas normal pregnant women who deliver at term maintain CSTs dominated by Lactobacillus species. The enhanced stability of Lactobacillus spp. during pregnancy is thought to provide a protective role against ascending infections [2].

2.2. The maternal gut microbiome

This also changes throughout pregnancy, independent of health status and diet, with a decrease in individual diversity by the third trimester, marked by an increase in Proteobacteria and Actinobacteria, and a decrease in Faecalibacterium spp., butyrate producers with anti-inflammatory effects [3]. These changes are similar to the changes seen with metabolic syndrome in non-pregnant individuals, and are likely beneficial to pregnancy by promoting the physiological insulin resistance that develops during a typical pregnancy course. This insulin resistance fosters adequate energy transfer to the developing fetus and ensures optimal fetal growth. When transferred to germ-free mice, third-trimester maternal fecal samples have been shown to induce greater adiposity and insulin insensitivity when compared to first-trimester fecal samples [3]. Other studies have demonstrated stability in the maternal microbiota, particularly in late pregnancy [4,5].

2.3. The placenta

The placenta maintains its own microbiome, which appears to occupy a niche separate from the maternal vaginal and gut microbiome. Surprisingly, this community most closely correlates with the maternal oral microbiome, as it is largely composed of non-pathogenic commensal organisms from the Firmicutes, Tenericutes, Proteobacteria, Bacteroidetes, and Fusobacteria phyla [6]. Variations of the placental microbiome may be seen with preterm birth, e.g. increased abundance of Actinomycetales and Alphaproteobacteria, and after remote antenatal infection, e.g. increased Streptococcus and Acinetobacter spp. [6].

3. Dysbiosis in pregnancy

Adverse health states during pregnancy may be associated with intestinal microbial changes. Obesity and excessive weight gain during pregnancy are associated with adverse gut microbiota alterations in mothers and their infants. Overweight pregnant women have significantly reduced numbers of intestinal bifidobacteria and significantly increased numbers of staphylococci, Bacteroides, Enterobacteriaceae (e.g. Escherichia coli) and clostridia with similar changes associated with excessive weight gain during pregnancy and with progression from the first to the third trimester of pregnancy [7]. Infants of overweight mothers have significantly higher concentrations of staphylococci, clostridia, and Bacteroides and lower concentrations of bifidobacteria in their fecal samples when compared to infants of normal weight mothers and infants of mothers with normal weight gain during pregnancy [8]. Furthermore, infants born to mothers with pre-gestational diabetes have a significant increase in bacterial diversity and a higher prevalence of Bacteroides, Parabacteroides, and Lachnospiraceae in their meconium when compared to infants born to mothers without diabetes [9]. Decreased Lactobacillus and Bifidobacterium spp. colonization during early infancy is associated with a greater risk for allergies at five years of life [10], and decreased bifidobacterial numbers and increased S. aureus numbers in infancy may predict childhood obesity [11]. These studies provide compelling evidence that the maternal microbiome affects the infant microbiome, which has lasting effects on childhood health.

Two lines of evidence support the hypothesis that maternal and/or placental dysbiosis is a trigger for preterm labor. First, bacterial vaginosis is associated with spontaneous abortion and preterm labor [12]. Bacterial vaginosis represents a marked state of vaginal dysbiosis, the complexity of which is still being characterized. Whether treatment of bacterial vaginosis in pregnancy decreases the risk of preterm birth remains uncertain [13]. Second, periodontal disease in pregnancy is associated with preterm labor and with low infant birth weight. The mechanisms connecting maternal oral dysbiosis and placental and fetal biology have been recently summarized [14]. Whereas treatment of periodontal disease in pregnancy has not resulted in improved pregnancy outcomes [15], animal models present compelling evidence of causality [16].

4. Prebiotic administration in pregnancy

Altering the prenatal microbiome during pregnancy can affect both the health of the mother and fetal outcomes. Animal studies have shown that prebiotic supplementation during pregnancy and lactation confers benefits to offspring, including improved weight gain independent of intake, increased colon length, increased muscle mass, increased bone mass, and decreased incidence of allergies and asthma symptoms [17,18]. Human studies are limited, but supplementation with indigestible oligosaccharide prebiotics, specifically fructo-oligosaccharides (FOS) and galacto-oligosaccharides (GOS) has been shown to significantly increase the number of maternal fecal Bifidobacterium spp., and most importantly Bifidobacterium longum; however, this bifidogenic effect may not be transferred to the neonatal gut [19]. Supplementation with synbiotics has been shown to significantly decrease serum insulin concentrations in women [20] and reduce the risk of pre-eclampsia and dyslipidemia [21].

5. Probiotic administration in pregnancy

Probiotic supplementation during pregnancy is safe and may have a protective role in preeclampsia, gestational diabetes, vaginal infections, maternal and infant weight gain, and later childhood diseases [22]. Studies linking probiotics to improved glycemic control have been mixed. One randomized placebo-controlled study of 256 healthy women showed that dietary interventions plus probiotics (Lactobacillus rhamnosus GG and Bifidobacterium lactis) decreased postpartum waist circumference [23] and significantly reduced the incidence of gestational diabetes from 34–36% to 13% [24]. However, another study of 175 obese women showed that a shorter, four-week administration of probiotics during early third trimester of pregnancy did not improve glycemic control [25]. The mixed results may be due to differences in demographics, genetics, or phenotypes, durations of probiotic intervention, or differences in probiotic strains or doses.

As pre-eclampsia can be thought of as an excessive, severe maternal generalized inflammatory reaction, it is an appealing disease target for probiotics. There have been no controlled trials, but a large prospective cohort study in Norway found an association between intake of milk products containing probiotic lactobacilli and reduced risk of pre-eclampsia, which was most pronounced in severe pre-eclampsia (Table 2) [26]. Additionally, probiotics have been shown to decrease high-sensitivity C-reactive protein [27], a marker of inflammation associated with adverse maternal conditions such as pre-eclampsia and gestational diabetes.

Table 2.

Summary of large cohort studies and meta-analyses of prenatal and postnatal probiotics

Intervention Patient type No. of patients Outcome RR or OR 95% CI
Cohort studies
 Milk containing lactobacilli [26] Primiparous women 33,399 Severe preeclampsia 0.79 0.66–0.96
 Milk containing lactobacilli [30] Pregnant women 18,888 Pre-term delivery 0.86 0.74–0.99
 Probiotic milk products [33] Pregnant women ± their infants 40,614 Eczema 0.94 0.89–0.99
 Probiotic milk products [33] Pregnant women ± their infants 40,614 Rhinoconjunctivitis 0.87 0.78–0.98
 Various probiotics [56] Premature infants 10,800 Necrotizing enterocolitis 0.55 0.39–0.78
 Various probiotics [56] Premature infants 8139 Death 0.72 0.61–0.85
 Various probiotics [56] Premature infants 6893 Sepsis 0.86 0.74–1.0
Randomized controlled trials
 Various probiotics [32] Pregnant women ± their term infants 4755 Eczema 0.78 0.69–0.89
 Various probiotics [55] Premature infants 10,520 Necrotizing enterocolitis 0.53 0.42–0.66
 Various probiotics [55] Premature infants 9507 Death 0.79 0.68–0.93
 Various probiotics [55] Premature infants 8707 Sepsis 0.88 0.77–1.0

RR, relative risk; OR, odds ratio; CI, confidence interval.

Bacterial vaginosis is another target for probiotics with combined benefits of restoring the vaginal microbiota after (or in place of) antibiotic treatment and decreasing the vaginal pH to an optimum value. Supplementation with a probiotic mixture in late pregnancy counteracts the decrease in Bifidobacterium spp., modulates the decrease in anti-inflammatory cytokines interleukin (IL)-4 and IL-10 and induces a decrease in pro-inflammatory cytokines [28]. One meta-analysis showed an 81% reduction in genital infections with oral probiotics, but data were inconclusive on whether this would decrease preterm labor [29]. A cohort study in Norway demonstrated an association between high intake of probiotic dairy products and reduced risk of spontaneous preterm delivery (Table 2) [30]. A more recent study showed no significant difference in the vaginal microbiota after eight weeks of probiotics, starting at the end of the first trimester [31]; however, the incidence of bacterial vaginosis was low (2.8%) in the treatment group.

Perhaps even more compelling are several meta-analyses which have shown that administration of probiotics during pregnancy prevents atopic dermatitis in children (Table 2) [32], and a large cohort study showed that probiotic lactobacilli and bifidobacteria during pregnancy decrease eczema and rhinoconjunctivitis in children [33]. Administration of probiotics to mothers during pregnancy and/or breastfeeding alters the cytokine profile of mother’s milk, increases infant fecal sIgA [34], and may moderate excessive weight gain of children during early childhood [35].

5.1. The intestinal microbiota and dysbiosis in term and preterm infants

Carefully performed studies suggest that term breast-fed infants in developing countries are more likely to be dominated by bifidobacteria compared to breast-fed infants in more industrialized nations and that this early dysbiosis is causally linked to increases in type 1 diabetes and food allergies in the latter [36]. In a cohort in Bangladesh, higher numbers of fecal bifidobacteria were associated with improved growth and vaccine response [37]. In developed countries, dysbiosis is widespread among term infants, even those who are born vaginally, breast-fed, and not treated with antibiotics. In premature infants, dysbiosis is almost universal with pro-inflammatory Proteobacteria in high abundance, particularly from 28–33 weeks corrected gestational age [38]. Dysbiosis and systemic inflammation have been demonstrated in such markedly diverse processes as infantile colic in term infants and necrotizing enterocolitis (NEC) in premature infants [39,40]. The loss of the intestinal microbes that have inhabited the intestinal tract of neonates and infants over millennia through formula feeding, antibiotics, and environmental and hygiene changes may have profound implications.

6. Prebiotics for term infants

Studies of non-human milk prebiotics in term infants are limited. Two recent randomized controlled trials are noteworthy. In the first, infants receiving formula supplemented with GOS had less colic than did infants receiving standard formula, and, among the infants with colic, those receiving the prebiotic formula had lower fecal clostridia and higher fecal lactobacilli and bifidobacteria [41]. In the second, infants receiving a fermented milk formula with added oligosaccharides (synbiotic) had a lower incidence of colic than infants receiving either a fermented milk formula (probiotic) or a formula containing a combination of oligosaccharides (prebiotic) [42].

Human milk oligosaccharides (HMOs) are produced in large diversity and abundance in human milk. These glycans are not digestible by the infant and appear to serve a variety of functions, including prebiotic stimulation of growth of specific bacterial species. More than 100 HMO structures have been characterized, and there is wide variability among the various gut microbes in their capacity to consume intact HMOs. Many bifidobacteria and bacteroides species are able to transport and consume HMOs, whereas Enterobacteriaceae are able to consume non-HMOs, such as GOS and maltodextrin, but not intact HMOs [43]. A careful characterization of changes in the fecal microbiota of the term infant and the ingested HMOs that pass through the intestinal tract to later appear in the infant feces confirms that, as the numbers of bifidobacteria and bacteroides increase in the feces, the amounts of fecal HMOs decrease, suggesting that HMOs play a significant role in shaping the microbiota of the breast-fed infant [44]. A single subspecies, Bifidobacterium longum subsp. infantis has evolved the capacity to transport and consume all the various HMO structures, providing it with a significant advantage in colonization over other gut microbes [45]. It is likely that the prebiotic HMOs, which are produced at significant cost to the mother, play a significant role in many of the observed beneficial effects of human milk. The discovery of novel methods for commercial production of large quantities of simple HMO structures has prompted the recent addition of HMOs to term infant formulas, though evidence of benefit from a single HMO structure is limited.

7. Prebiotics for preterm infants

In premature infants, various mixtures of non-human milk galacto-, fructo- and acidic oligosaccharides have been extensively studied. These prebiotic mixtures alter the fecal microbiome, decrease fecal pH, improve gastric motility, decrease feeding intolerance, and increase fecal sIgA [46]. However, a meta-analysis of seven placebo-controlled randomized clinical trials of prebiotics showed no decrease in NEC, sepsis or death [47]. Infants treated with prebiotic mixtures did not differ in intestinal permeability, vaccine response, neurodevelopmental outcome, or allergic/infectious diseases from placebo infants [48].

The variability of HMOs is greater in milk from mothers delivering preterm compared to milk from mothers delivering at term. Specific HMO structures appear to impact the fecal microbiome in premature infants [49] and may explain a portion of the protective benefit of human milk against NEC. Studies of single HMO structures are thus far limited to animal studies, which demonstrate benefit in prevention of NEC [50,51].

8. Probiotics for term infants

The impact of probiotic administration for the treatment of infant colic has been the subject of several randomized clinical trials. A recent meta-analysis of six studies of L. reuteri demonstrated significant improvement with a mean decrease in crying time of 56 min per day in the infants receiving the probiotic [52]. A single small trial of a synbiotic combination of FOS and seven probiotic bacteria also demonstrated significant improvement in colic symptoms compared to placebo [53]. Several of the clinical trials of probiotic administration during pregnancy included administration of the same probiotic organism to the infants after birth. A recent analysis of four such clinical trials included long-term outcomes and found that perinatal L. rhamnosus was associated with decreased allergic diseases in children without safety concerns [54].

9. Probiotics for preterm infants

To date, 35 randomized controlled trials of probiotics have been published, with mixed results. A recent meta-analysis of these studies concluded that probiotics decrease the risk of NEC and death, but not sepsis (Table 2) [55]. Cohort studies of probiotics in premature infants have yielded strikingly similar results (Table 2) [56]. Although these trials utilized differing doses and probiotic strains, it appears that doses of ≥109 microbes per day are more beneficial than lower doses, that combinations of probiotics may have advantages over single organisms, and that probiotics are more effective in premature infants receiving human milk than formula [57].

10. Safety of prebiotics and probiotics

The safety of probiotics and prebiotics has been extensively reviewed. The most frequent adverse effect of prebiotic oligosaccharides is intestinal discomfort including bloating, flatulence, and diarrhea. Risks associated with probiotic microbes include probiotic sepsis, contamination of the probiotic product, and lack of efficacy related to reduced viability. Probiotic sepsis has been reported for a variety of commercial products, but appears to be rare. In countries where registries of all positive blood cultures are available, marked increases in probiotic sepsis have not been seen in spite of marked increases in consumption of probiotics [58]. Contamination of commercial probiotics with organisms not advertised on the package is frequent [59]. Contamination with pathogens appears to be much less widespread, but this has been associated with nosocomial infection and death in a premature infant [60].

Practice points.

During pregnancy:

  • Prebiotics:

    • May decrease the risk of pre-eclampsia and dyslipidemia.

  • Probiotics:

    • They have beneficial role in modulating gestational diabetes, pre-eclampsia, excessive weight gain, and bacterial vaginosis.

    • Evidence is mixed regarding their role in preventing preterm labor and low birth weight.

    • They decrease the incidence of atopic dermatitis in later childhood. Evidence is compelling and suggests a need for practice change.

For infants:

  • Prebiotics:

    • They decrease symptoms of infantile colic.

    • They do not decrease the risk of NEC, sepsis or death in premature infants.

  • Probiotics:

    • They decrease symptoms of infant colic.

    • They decrease the risk of NEC and death with consistent trends towards prevention of sepsis in premature infants. Evidence is compelling and suggests a need for practice change.

Research directions.

  • Development of optimal probiotic strains for prevention of diseases associated with intestinal dysbiosis (it is likely that different strains will protect against different disease processes).

  • Interactions between ingested probiotic microbes and dietary prebiotic glycans (it is likely that the foods consumed by the probiotics are important in colonization and efficacy).

  • Interactions between host genotype and probiotic or prebiotic response (e.g. individuals with a common mutation in the fucosyl transferase 2 gene, widely referred to as “non-secretors,” likely respond differently than those without the mutation).

  • Novel methods of probiotic delivery (current probiotic products are limited to organisms resistant to gastric acid, bile acids, and digestive enzymes).

Acknowledgments

Funding sources

None.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of interest statement

None declared.

References

  • 1.Aagaard K, Riehle K, Ma J, et al. A metagenomic approach to characterization of the vaginal microbiome signature in pregnancy. PloS One. 2012;7:e36466. doi: 10.1371/journal.pone.0036466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Romero R, Hassan SS, Gajer P, et al. The composition and stability of the vaginal microbiota of normal pregnant women is different from that of non-pregnant women. Microbiome. 2014;2:4. doi: 10.1186/2049-2618-2-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Koren O, Goodrich JK, Cullender TC, et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell. 2012;150:470–80. doi: 10.1016/j.cell.2012.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Avershina E, Storro O, Oien T, Johnsen R, Pope P, Rudi K. Major faecal microbiota shifts in composition and diversity with age in a geographically restricted cohort of mothers and their children. FEMS Microbiol Ecol. 2014;87:280–90. doi: 10.1111/1574-6941.12223. [DOI] [PubMed] [Google Scholar]
  • 5.Jost T, Lacroix C, Braegger C, Chassard C. Stability of the maternal gut microbiota during late pregnancy and early lactation. Curr Microbiol. 2014;68:419–27. doi: 10.1007/s00284-013-0491-6. [DOI] [PubMed] [Google Scholar]
  • 6.Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta harbors a unique microbiome. Sci Transl Med. 2014;6:237ra65. doi: 10.1126/scitranslmed.3008599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Santacruz A, Collado MC, Garcia-Valdes L, et al. Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Br J Nutr. 2010;104:83–92. doi: 10.1017/S0007114510000176. [DOI] [PubMed] [Google Scholar]
  • 8.Collado MC, Isolauri E, Laitinen K, Salminen S. Effect of mother’s weight on infant’s microbiota acquisition, composition, and activity during early infancy: a prospective follow-up study initiated in early pregnancy. Am J Clin Nutr. 2010;92:1023–30. doi: 10.3945/ajcn.2010.29877. [DOI] [PubMed] [Google Scholar]
  • 9.Hu J, Nomura Y, Bashir A, et al. Diversified microbiota of meconium is affected by maternal diabetes status. PloS One. 2013;8:e78257. doi: 10.1371/journal.pone.0078257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sjogren YM, Jenmalm MC, Bottcher MF, Bjorksten B, Sverremark-Ekstrom E. Altered early infant gut microbiota in children developing allergy up to 5 years of age. Clin Exp Allergy. 2009;39:518–26. doi: 10.1111/j.1365-2222.2008.03156.x. [DOI] [PubMed] [Google Scholar]
  • 11.Kalliomaki M, Collado MC, Salminen S, Isolauri E. Early differences in fecal microbiota composition in children may predict overweight. Am J Clin Nutr. 2008;87:534–8. doi: 10.1093/ajcn/87.3.534. [DOI] [PubMed] [Google Scholar]
  • 12.Isik G, Demirezen S, Donmez HG, Beksac MS. Bacterial vaginosis in association with spontaneous abortion and recurrent pregnancy losses. J Cytol. 2016;33:135–40. doi: 10.4103/0970-9371.188050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lamont RF, Keelan JA, Larsson PG, Jorgensen JS. The treatment of bacterial vaginosis in pregnancy with clindamycin to reduce the risk of infection-related preterm birth: a response to the Danish Society of Obstetrics and Gynecology guideline group’s clinical recommendations. Acta Obstet Gynecol Scand. 2017;96:139–43. doi: 10.1111/aogs.13065. [DOI] [PubMed] [Google Scholar]
  • 14.Puertas A, Magan-Fernandez A, Blanc V, et al. Association of periodontitis with preterm birth and low birth weight: a comprehensive review. J Maternal-Fetal Neonatal Med. 2017 Feb 28; doi: 10.1080/14767058.2017.1293023. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 15.Lopez NJ, Uribe S, Martinez B. Effect of periodontal treatment on preterm birth rate: a systematic review of meta-analyses. Periodontology 2000. 2015;67:87–130. doi: 10.1111/prd.12073. [DOI] [PubMed] [Google Scholar]
  • 16.Ebersole JL, Holt SC, Cappelli D. Periodontitis in pregnant baboons: systemic inflammation and adaptive immune responses and pregnancy outcomes in a baboon model. J Periodontal Res. 2014;49:226–36. doi: 10.1111/jre.12099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hogenkamp A, Knippels LM, Garssen J, van Esch BC. Supplementation of mice with specific nondigestible oligosaccharides during pregnancy or lactation leads to diminished sensitization and allergy in the female offspring. J Nutr. 2015;145:996–1002. doi: 10.3945/jn.115.210401. [DOI] [PubMed] [Google Scholar]
  • 18.Bueno-Vargas P, Manzano M, Diaz-Castro J, Lopez-Aliaga I, Rueda R, Lopez-Pedrosa JM. Maternal dietary supplementation with oligofructose-enriched inulin in gestating/lactating rats preserves maternal bone and improves bone microarchitecture in their offspring. PloS One. 2016;11:e0154120. doi: 10.1371/journal.pone.0154120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jinno S, Toshimitsu T, Nakamura Y, et al. Maternal prebiotic ingestion increased the number of fecal bifidobacteria in pregnant women but not in their neonates aged one month. Nutrients. 2017;9(3) doi: 10.3390/nu9030196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Taghizadeh M, Asemi Z. Effects of synbiotic food consumption on glycemic status and serum hs-CRP in pregnant women: a randomized controlled clinical trial. Hormones (Athens) 2014;13:398–406. doi: 10.14310/horm.2002.1489. [DOI] [PubMed] [Google Scholar]
  • 21.Qiu C, Coughlin KB, Frederick IO, Sorensen TK, Williams MA. Dietary fiber intake in early pregnancy and risk of subsequent preeclampsia. Am J Hypertens. 2008;21:903–9. doi: 10.1038/ajh.2008.209. [DOI] [PubMed] [Google Scholar]
  • 22.Gomez Arango LF, Barrett HL, Callaway LK, Nitert MD. Probiotics and pregnancy. Curr Diabet Rep. 2015;15:567. doi: 10.1007/s11892-014-0567-0. [DOI] [PubMed] [Google Scholar]
  • 23.Ilmonen J, Isolauri E, Poussa T, Laitinen K. Impact of dietary counselling and probiotic intervention on maternal anthropometric measurements during and after pregnancy: a randomized placebo-controlled trial. Clin Nutr. 2011;30:156–64. doi: 10.1016/j.clnu.2010.09.009. [DOI] [PubMed] [Google Scholar]
  • 24.Luoto R, Laitinen K, Nermes M, Isolauri E. Impact of maternal probiotic-supplemented dietary counselling on pregnancy outcome and prenatal and postnatal growth: a double-blind, placebo-controlled study. Br J Nutr. 2010;103:1792–9. doi: 10.1017/S0007114509993898. [DOI] [PubMed] [Google Scholar]
  • 25.Lindsay KL, Kennelly M, Culliton M, et al. Probiotics in obese pregnancy do not reduce maternal fasting glucose: a double-blind, placebo-controlled, randomized trial (Probiotics in Pregnancy Study) Am J Clin Nutr. 2014;99:1432–9. doi: 10.3945/ajcn.113.079723. [DOI] [PubMed] [Google Scholar]
  • 26.Brantsaeter AL, Myhre R, Haugen M, et al. Intake of probiotic food and risk of preeclampsia in primiparous women: the Norwegian Mother and Child Cohort Study. Am J Epidemiol. 2011;174:807–15. doi: 10.1093/aje/kwr168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Asemi Z, Jazayeri S, Najafi M, et al. Effects of daily consumption of probiotic yoghurt on inflammatory factors in pregnant women: a randomized controlled trial. Pak J Biol Sci. 2011;14:476–82. doi: 10.3923/pjbs.2011.476.482. [DOI] [PubMed] [Google Scholar]
  • 28.Vitali B, Cruciani F, Baldassarre ME, et al. Dietary supplementation with probiotics during late pregnancy: outcome on vaginal microbiota and cytokine secretion. BMC Microbiol. 2012;12:236. doi: 10.1186/1471-2180-12-236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Othman M, Neilson JP, Alfirevic Z. Probiotics for preventing preterm labour. Cochrane Database Syst Rev. 2007;(1):CD005941. doi: 10.1002/14651858.CD005941.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Myhre R, Brantsaeter AL, Myking S, et al. Intake of probiotic food and risk of spontaneous preterm delivery. Am J Clin Nutr. 2011;93:151–7. doi: 10.3945/ajcn.110.004085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Gille C, Boer B, Marschal M, et al. Effect of probiotics on vaginal health in pregnancy. EFFPRO, a randomized controlled trial. Am J Obstet Gynecol. 2016;215:608.e1–7. doi: 10.1016/j.ajog.2016.06.021. [DOI] [PubMed] [Google Scholar]
  • 32.Zuccotti G, Meneghin F, Aceti A, et al. Probiotics for prevention of atopic diseases in infants: systematic review and meta-analysis. Allergy. 2015;70:1356–71. doi: 10.1111/all.12700. [DOI] [PubMed] [Google Scholar]
  • 33.Bertelsen RJ, Brantsæter AL, Magnus MC, et al. Probiotic milk consumption in pregnancy and infancy and subsequent childhood allergic diseases. J Allergy Clin Immunol. 2014;133:165–71.e8. doi: 10.1016/j.jaci.2013.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Baldassarre ME, Di Mauro A, Mastromarino P, et al. Administration of a multi-strain probiotic product to women in the perinatal period differentially affects the breast milk cytokine profile and may have beneficial effects on neonatal gastrointestinal functional symptoms. A randomized clinical trial. Nutrients. 2016;8(11) doi: 10.3390/nu8110677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Luoto R, Kalliomaki M, Laitinen K, Isolauri E. The impact of perinatal probiotic intervention on the development of overweight and obesity: follow-up study from birth to 10 years. Int J Obes (2005) 2010;34:1531–7. doi: 10.1038/ijo.2010.50. [DOI] [PubMed] [Google Scholar]
  • 36.Vatanen T, Kostic AD, d’Hennezel E, et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell. 2016;165:842–53. doi: 10.1016/j.cell.2016.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Huda MN, Lewis Z, Kalanetra KM, et al. Stool microbiota and vaccine responses of infants. Pediatrics. 2014;134:e362–72. doi: 10.1542/peds.2013-3937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.La Rosa PS, Warner BB, Zhou Y, et al. Patterned progression of bacterial populations in the premature infant gut. Proc Natl Acad Sci USA. 2014;111:12522–7. doi: 10.1073/pnas.1409497111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pärtty A, Kalliomäki M, Salminen S, Isolauri E. Infantile colic is associated with low-grade systemic inflammation. J Pediatr Gastroenterol Nutr. 2017;64:691–5. doi: 10.1097/MPG.0000000000001340. [DOI] [PubMed] [Google Scholar]
  • 40.Pammi M, Cope J, Tarr PI, et al. Intestinal dysbiosis in preterm infants preceding necrotizing enterocolitis: a systematic review and meta-analysis. Microbiome. 2017;5:31. doi: 10.1186/s40168-017-0248-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Giovannini M, Verduci E, Gregori D, et al. Prebiotic effect of an infant formula supplemented with galacto-oligosaccharides: randomized multicenter trial. J Am Coll Nutr. 2014;33:385–93. doi: 10.1080/07315724.2013.878232. [DOI] [PubMed] [Google Scholar]
  • 42.Vandenplas Y, Ludwig T, Bouritius H, et al. Randomised controlled trial demonstrates that fermented infant formula with short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides reduces the incidence of infantile colic. Acta Paediatr (Oslo, Norway: 1992) 2017;106:1150–8. doi: 10.1111/apa.13844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hoeflinger JL, Davis SR, Chow J, Miller MJ. In vitro impact of human milk oligosaccharides on Enterobacteriaceae growth. J Agric Food Chem. 2015;63:3295–302. doi: 10.1021/jf505721p. [DOI] [PubMed] [Google Scholar]
  • 44.De Leoz ML, Kalanetra KM, Bokulich NA, et al. 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. doi: 10.1021/pr500759e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Underwood MA, German JB, Lebrilla CB, Mills DA. Bifidobacterium longum subspecies infantis: champion colonizer of the infant gut. Pediatr Res. 2015;77:229–35. doi: 10.1038/pr.2014.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Westerbeek EA, Slump RA, Lafeber HN, et al. The effect of enteral supplementation of specific neutral and acidic oligosaccharides on the faecal microbiota and intestinal microenvironment in preterm infants. Eur J Clin Microbiol Infect Dis. 2013;32:269–76. doi: 10.1007/s10096-012-1739-y. [DOI] [PubMed] [Google Scholar]
  • 47.Srinivasjois R, Rao S, Patole S. Prebiotic supplementation in preterm neonates: updated systematic review and meta-analysis of randomised controlled trials. Clin Nutr (Edinb) 2013;32:958–65. doi: 10.1016/j.clnu.2013.05.009. [DOI] [PubMed] [Google Scholar]
  • 48.LeCouffe NE, Westerbeek EA, van Schie PE, Schaaf VA, Lafeber HN, van Elburg RM. Neurodevelopmental outcome during the first year of life in preterm infants after supplementation of a prebiotic mixture in the neonatal period: a follow-up study. Neuropediatrics. 2014;45:22–9. doi: 10.1055/s-0033-1349227. [DOI] [PubMed] [Google Scholar]
  • 49.Underwood MA, Gaerlan S, De Leoz ML, et al. Human milk oligosaccharides in premature infants: absorption, excretion, and influence on the intestinal microbiota. Pediatr Res. 2015;78:670–7. doi: 10.1038/pr.2015.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Good M, Sodhi CP, Yamaguchi Y, et al. The human milk oligosaccharide 2′-fucosyllactose attenuates the severity of experimental necrotising enterocolitis by enhancing mesenteric perfusion in the neonatal intestine. Br J Nutr. 2016;116:1175–87. doi: 10.1017/S0007114516002944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Autran CA, Schoterman MH, Jantscher-Krenn E, Kamerling JP, Bode L. Sialylated galacto-oligosaccharides and 2′-fucosyllactose reduce necrotising enterocolitis in neonatal rats. Br J Nutr. 2016;116:294–9. doi: 10.1017/S0007114516002038. [DOI] [PubMed] [Google Scholar]
  • 52.Harb T, Matsuyama M, David M, Hill RJ. Infant colic – what works: a systematic review of interventions for breast-fed infants. J pediatr gastroenterol nutr. 2016;62:668–86. doi: 10.1097/MPG.0000000000001075. [DOI] [PubMed] [Google Scholar]
  • 53.Kianifar H, Ahanchian H, Grover Z, et al. Synbiotic in the management of infantile colic: a randomised controlled trial. J Paediatr Child Health. 2014;50:801–5. doi: 10.1111/jpc.12640. [DOI] [PubMed] [Google Scholar]
  • 54.Lundelin K, Poussa T, Salminen S, Isolauri E. Long-term safety and efficacy of perinatal probiotic intervention: evidence from a follow-up study of four randomized, double-blind, placebo-controlled trials. Pediatr Allergy Immunol. 2017;28:170–5. doi: 10.1111/pai.12675. [DOI] [PubMed] [Google Scholar]
  • 55.Sawh SC, Deshpande S, Jansen S, Reynaert CJ, Jones PM. Prevention of necrotizing enterocolitis with probiotics: a systematic review and meta-analysis. PeerJ. 2016;4:e2429. doi: 10.7717/peerj.2429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Olsen R, Greisen G, Schroder M, Brok J. Prophylactic probiotics for preterm infants: a systematic review and meta-analysis of observational studies. Neonatology. 2016;109:105–12. doi: 10.1159/000441274. [DOI] [PubMed] [Google Scholar]
  • 57.Underwood MA. Impact of probiotics on necrotizing enterocolitis. Semin Perinatol. 2017;41:41–51. doi: 10.1053/j.semperi.2016.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Salminen MK, Tynkkynen S, Rautelin H, et al. Lactobacillus bacteremia during a rapid increase in probiotic use of Lactobacillus rhamnosus GG in Finland. Clin Infect Dis. 2002;35:1155–60. doi: 10.1086/342912. [DOI] [PubMed] [Google Scholar]
  • 59.Lewis ZT, Shani G, Masarweh CF, et al. Validating bifidobacterial species and subspecies identity in commercial probiotic products. Pediatr Res. 2016;79:445–52. doi: 10.1038/pr.2015.244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Vallabhaneni S, Walker TA, Lockhart SR, et al. Notes from the field: fatal gastrointestinal mucormycosis in a premature infant associated with a contaminated dietary supplement – Connecticut, 2014. Morb Mortal Wkly Rep. 2015;64:155–6. [PMC free article] [PubMed] [Google Scholar]

RESOURCES