scholarly journals Colonization Resistance in the Infant Gut: The Role of B. infantis in Reducing pH and Preventing Pathogen Growth

2020 ◽  
Vol 9 (2) ◽  
pp. 7 ◽  
Author(s):  
Rebbeca M. Duar ◽  
David Kyle ◽  
Giorgio Casaburi

Over the past century, there has been a steady increase in the stool pH of infants from industrialized countries. Analysis of historical data revealed a strong association between abundance of Bifidobacterium in the gut microbiome of breasted infants and stool pH, suggesting that this taxon plays a key role in determining the pH in the gut. Bifidobacterium longum subsp. infantis is uniquely equipped to metabolize human milk oligosaccharides (HMO) from breastmilk into acidic end products, mainly lactate and acetate. The presence of these acidic compounds in the infant gut is linked to a lower stool pH. Conversely, infants lacking B. infantis have a significantly higher stool pH, carry a higher abundance of potential pathogens and mucus-eroding bacteria in their gut microbiomes, and have signs of chronic enteric inflammation. This suggests the presence of B. infantis and low intestinal pH may be critical to maintaining a protective environment in the infant gut. Here, we summarize recent studies demonstrating that feeding B. infantis EVC001 to breastfed infants results in significantly lower fecal pH compared to controls and propose that low pH is one critical factor in preventing the invasion and overgrowth of harmful bacteria in the infant gut, a process known as colonization resistance.

mSphere ◽  
2018 ◽  
Vol 3 (2) ◽  
Author(s):  
Bethany M. Henrick ◽  
Andra A. Hutton ◽  
Michelle C. Palumbo ◽  
Giorgio Casaburi ◽  
Ryan D. Mitchell ◽  
...  

ABSTRACT Historically, Bifidobacterium species were reported as abundant in the breastfed infant gut. However, recent studies in resource-rich countries show an increased abundance of taxa regarded as signatures of dysbiosis. It is unclear whether these differences are the product of genetics, geographic factors, or interventions such as formula feeding, antibiotics, and caesarean section. Fecal pH is strongly associated with Bifidobacterium abundance; thus, pH could be an indicator of its historical abundance. A review of 14 clinical studies published between 1926 and 2017, representing more than 312 healthy breastfed infants, demonstrated a change in fecal pH from 5.0 to 6.5 (adjusted r 2 = 0.61). This trend of increasing infant fecal pH over the past century is consistent with current reported discrepancies in Bifidobacterium species abundance in the gut microbiome in resource-rich countries compared to that in historical reports. Our analysis showed that increased fecal pH and abundance of members of the families Enterobacteriaceae , Clostridiaceae , Peptostreptococcaceae , and Veillonellaceae are associated, indicating that loss of highly specialized Bifidobacterium species may result in dysbiosis, the implications of which are not yet fully elucidated. Critical assessment of interventions that restore this ecosystem, measured by key parameters such as ecosystem productivity, gut function, and long-term health, are necessary to understand the magnitude of this change in human biology over the past century.


2006 ◽  
Vol 144 (6) ◽  
pp. 489-502 ◽  
Author(s):  
J. DIXON ◽  
L. NALLEY ◽  
P. KOSINA ◽  
R. LA ROVERE ◽  
J. HELLIN ◽  
...  

During the past century wheat breeders have produced a large number of genetically improved wheat lines and varieties. This activity has led to widespread adoption of improved varieties, a steady increase in average wheat yields during the past 4–5 decades and major contributions to food security and poverty reduction. The rate of generation and adoption of improved varieties, and therefore the time lag from varietal release to widespread use, varies across regions. The remarkable success of wheat improvement hinges on the decisions of millions of farmers to adopt, or replace older wheat varieties with superior material. The present paper summarizes studies on the determinants of adoption. Because adoption is a necessary but not sufficient condition for economic impact, the present paper synthesizes key assessments of impact from different farming systems in developing countries.


mSphere ◽  
2017 ◽  
Vol 2 (6) ◽  
Author(s):  
Steven A. Frese ◽  
Andra A. Hutton ◽  
Lindsey N. Contreras ◽  
Claire A. Shaw ◽  
Michelle C. Palumbo ◽  
...  

ABSTRACT The gut microbiome in early life plays an important role for long-term health and is shaped in large part by diet. Probiotics may contribute to improvements in health, but they have not been shown to alter the community composition of the gut microbiome. Here, we found that breastfed infants could be stably colonized at high levels by provision of B. infantis EVC001, with significant changes to the overall microbiome composition persisting more than a month later, whether the infants were born vaginally or by caesarean section. This observation is consistent with previous studies demonstrating the capacity of this subspecies to utilize human milk glycans as a nutrient and underscores the importance of pairing a probiotic organism with a specific substrate. Colonization by B. infantis EVC001 resulted in significant changes to fecal microbiome composition and was associated with improvements in fecal biochemistry. The combination of human milk and an infant-associated Bifidobacterium sp. shows, for the first time, that durable changes to the human gut microbiome are possible and are associated with improved gut function. Attempts to alter intestinal dysbiosis via administration of probiotics have consistently shown that colonization with the administered microbes is transient. This study sought to determine whether provision of an initial course of Bifidobacterium longum subsp. infantis (B. infantis) would lead to persistent colonization of the probiotic organism in breastfed infants. Mothers intending to breastfeed were recruited and provided with lactation support. One group of mothers fed B. infantis EVC001 to their infants from day 7 to day 28 of life (n = 34), and the second group did not administer any probiotic (n = 32). Fecal samples were collected during the first 60 postnatal days in both groups. Fecal samples were assessed by 16S rRNA gene sequencing, quantitative PCR, mass spectrometry, and endotoxin measurement. B. infantis-fed infants had significantly higher populations of fecal Bifidobacteriaceae, in particular B. infantis, while EVC001 was fed, and this difference persisted more than 30 days after EVC001 supplementation ceased. Fecal milk oligosaccharides were significantly lower in B. infantis EVC001-fed infants, demonstrating higher consumption of human milk oligosaccharides by B. infantis EVC001. Concentrations of acetate and lactate were significantly higher and fecal pH was significantly lower in infants fed EVC001, demonstrating alterations in intestinal fermentation. Infants colonized by Bifidobacteriaceae at high levels had 4-fold-lower fecal endotoxin levels, consistent with observed lower levels of Gram-negative Proteobacteria and Bacteroidetes. IMPORTANCE The gut microbiome in early life plays an important role for long-term health and is shaped in large part by diet. Probiotics may contribute to improvements in health, but they have not been shown to alter the community composition of the gut microbiome. Here, we found that breastfed infants could be stably colonized at high levels by provision of B. infantis EVC001, with significant changes to the overall microbiome composition persisting more than a month later, whether the infants were born vaginally or by caesarean section. This observation is consistent with previous studies demonstrating the capacity of this subspecies to utilize human milk glycans as a nutrient and underscores the importance of pairing a probiotic organism with a specific substrate. Colonization by B. infantis EVC001 resulted in significant changes to fecal microbiome composition and was associated with improvements in fecal biochemistry. The combination of human milk and an infant-associated Bifidobacterium sp. shows, for the first time, that durable changes to the human gut microbiome are possible and are associated with improved gut function.


2021 ◽  
Author(s):  
Lauren Tso ◽  
Kevin S Bonham ◽  
Alyssa Fishbein ◽  
Sophie Rowland ◽  
Vanja Klepac-Ceraj ◽  
...  

Bifidobacterium longum subsp. infantis (B. infantis) is one of few microorganisms capable of metabolizing human breast milk and is a pioneer colonizer in the guts of breastfed infants. One current challenge is differentiating B. infantis from its close relatives, B. longum and B. suis, by molecular methods. These two organisms are classified in the same species group as B. infantis but do not share the ability to metabolize human milk oligosaccharides (HMOs). Here, we compared HMO-metabolizing genes across different Bifidobacterium genomes to develop B. infantis specific primers and determine if they alone can be used to quickly characterize B. infantis with shotgun metagenomic sequencing data. We showed that B. infantis is uniquely identified by the presence of five HMO-metabolizing gene clusters, used this characterization to test for its prevalence in infants, and validated the results using the B. infantis-specific primers. By examining stool samples from a cohort of US children and pregnant women using shotgun metagenomic sequencing, we observed that only 18 of 204 (8.8%) of children under 2 years old harbored B. infantis. These results highlight the importance of developing and improving approaches to identify B. infantis. A more accurate characterization may provide insights into regional differences of B. infantis prevalence in infant gut microbiota.


2020 ◽  
Vol 4 (Supplement_2) ◽  
pp. 1571-1571
Author(s):  
Jules Larke ◽  
Mark Underwood ◽  
Kara Kuhn-Riordan ◽  
Carolyn Slupsky

Abstract Objectives The acquisition and succession of intestinal microbiota represents a critical factor in early life development. However, infants born prematurely exhibit a delayed assembly of microbial communities. Supplementation of probiotics help to shift the community to a more favorable structure, thereby promoting a functional benefit. We investigated the effect of two different probiotics, Lactobacillus reuteri (L. reuteri) or Bifidobacterium longum subsp. Infantis (B. infantis) on fecal metabolite profiles in premature infants. Methods This study included 43 very low birth weight (< 1500 grams) premature infants. A dose of L. reuteri (1 × 109 CFU) or B. infantis (8 × 1010 CFU) was provided daily as standard of care until 34 weeks corrected gestational age (CGA). Samples of feces and milk were collected at 30 and 32 weeks CGA. Fecal and milk metabolites were assessed by 1H nuclear magnetic resonance spectroscopy at 298K using a NOESY 1H pre-saturation experiment on a Bruker Avance 600 MHz spectrometer (Bruker BioSpin, Germany). Results Fecal metabolite profiles separated by probiotic group revealing distinct compositions at 30 weeks CGA (Adonis R2 = 0.082, p = 0.023) and was maintained at 32 weeks CGA (Adonis R2 = 0.108, p = 0.001). The separation of groups by fecal metabolites was observed despite similar human milk oligosaccharide (HMO) composition from diet at both 30 weeks (Adonis R2 = 0.034, p = 0.378) and 32 weeks CGA (Adonis R2 = 0.016, p = 0.544). Correlations between milk 2’-fucosyllactose and fecal metabolites revealed trends with fecal acetate and pyruvate at 32 weeks (Pearson's, R = 0.41, p = 0.06 and R = 0.4, p = 0.068 respectively) in the B. infantis group only. Conclusions Fecal metabolite profiles in premature infants are affected by probiotic administration despite similar HMO composition in their diet. Infants receiving L. reuteri exhibit an HMO dominant fecal profile, whereas those receiving B. infantis are more closely oriented with HMO fermentation products indicating a saccharolytic gut microbiota. Funding Sources CS would like to acknowledge funding from the Kinsella endowed chair in Food, Nutrition, and Health as well as USDA-NIFA Hatch project 1021411.


2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Zeyu Kan ◽  
Baolong Luo ◽  
Jingjing Cai ◽  
Yan Zhang ◽  
Fengwei Tian ◽  
...  

Abstract Background Bifidobacteria are important probiotics; some of the beneficial effects of bifidobacteria are achieved by the hydrolysis of glycans in the human gut. However, because the diet of breastfed infants typically lacks plant-derived glycans, in the gut environment of mothers and their breastfed infants, the mother will intake a variety of plant-derived glycans, such as from onions and bananas, through her diet. Under this assumption, we are interested in whether the same species of bifidobacteria isolated from mother-infant pairs present a distinction in their hydrolysis of plant-derived carbohydrates. Results Among the 36 Bifidobacterium strains, bifidobacterial carbohydrate utilization showed two trends related to the intestinal environment where the bacteria lived. Compared with infant-type bifidobacterial strains, adult-type bifidobacterial strains preferred to use plant-derived glycans. Of these strains, 10 isolates, 2 Bifidobacterium pseudocatenulatum (B. pseudocatenulatum), 2 Bifidobacterium pseudolongum (B. pseudolongum), 2 Bifidobacterium bifidum (B. bifidum), 2 Bifidobacterium breve (B. breve), and 2 Bifidobacterium longum (B. longum), were shared between the mother-infant pairs. Moreover, the repetitive sequence-based polymerase chain reaction (rep-PCR) results illustrated that B. pseudolongum and B. bifidum showed genotypic similarities of 95.3 and 98.2%, respectively. Combined with the carbohydrate fermentation study, these results indicated that the adult-type strains have a stronger ability to use plant-derived glycans than infant-type strains. Our work suggests that bifidobacterial carbohydrate metabolism differences resulted in the selective adaptation to the distinct intestinal environment of an adult or breastfed infant. Conclusions The present study revealed that the different gut environments can lead to the differences in the polysaccharide utilization in the same strains of bifidobacterial strains, suggesting a further goal of investigating the exact expression of certain enzymes in response to specific carbon sources.


Foods ◽  
2018 ◽  
Vol 7 (12) ◽  
pp. 196 ◽  
Author(s):  
Erinn Quinn ◽  
Helen Slattery ◽  
Aoife Thompson ◽  
Michelle Kilcoyne ◽  
Lokesh Joshi ◽  
...  

Bifidobacteria play a vital role in human nutrition and health by shaping and maintaining the gut ecosystem. In order to exert a beneficial effect, a sufficient population of bifidobacteria must colonise the host. In this study, we developed a miniaturised high-throughput in vitro assay for assessing the colonising ability of bacterial strains in human cells. We also investigated a variety of components isolated from different milk sources for their ability to increase the adherence of Bifidobacterium longum subsp. infantis ATCC 15697, a common member of the gastrointestinal microbiota of breastfed infants, to HT-29 cells. Both conventional and miniaturised colonisation assays were employed to examine the effect of 13 different milk-derived powders on bacterial adherence, including positive controls which had previously resulted in increased bifidobacterial adherence (human milk oligosaccharides and a combination of 3′- and 6′-sialylactose) to intestinal cells. Immunoglobulin G enriched from bovine whey and goat milk oligosaccharides resulted in increased adhesion (3.3- and 8.3-fold, respectively) of B. infantis to the intestinal cells and the miniaturised and conventional assays were found to yield comparable and reproducible results. This study highlights the potential of certain milk components to favourably modulate adhesion of bifidobacteria to human intestinal cells.


2019 ◽  
Vol 20 (17) ◽  
pp. 4275 ◽  
Author(s):  
Valadez-Bustos ◽  
Escamilla-Silva ◽  
García-Vázquez ◽  
Gallegos-Corona ◽  
Amaya-Llano ◽  
...  

The Insulin-like growth factor-I/Insulin-like growth factor-I receptor (IGF-1/IGF-1R) system is a major determinant in colorectal cancer (CRC) pathogenesis. Probiotics (Bifidobacterium longum, BF) and lycopene (LYC) have been individually researched for their beneficial effects in the prevention of CRC. However, the effect of a combined treatment of microencapsulated BF and LYC on IGF-1/IGF-1R/IGFBPs (Insulin-like growth factor-binding proteins) expression in an azoxymethane (AOM)-dextran sulfate sodium (DSS)-induced CRC model have not been demonstrated. BF was microencapsulated by the spray drying technique, with high viability, and daily gavaged with LYC for 16 weeks to CD-1 mice in an AOM-DSS model. The results indicated that BF- and BF + LYC-treated groups had significantly lower inflammation grade, tumor incidence (13–38%) and adenocarcinoma (13–14%) incidence compared to the AOM + DSS group (80%), whereas LYC treatment only protected against inflammation grade and incidence. Caecal, colonic and fecal pH and β-glucuronidase (β-GA) values were significantly normalized by BF and LYC. Similarly, BF and BF + LYC treatments significantly reduced both the positive rate and expression grade of IGF-1 and IGF-1R proteins and normalized Insulin-like growth factor-binding protein-3 (IGFBP3) expression. Based on intestinal parameters related to the specific colon carcinogenesis in an AOM-DSS-induced model, LYC and microencapsulated BF supplementation resulted in a significant chemopreventive potential through the modulation of IGF-1/IGF-1R system.


Sign in / Sign up

Export Citation Format

Share Document