Colonization by B. infantis EVC001 modulates enteric inflammation in exclusively breastfed infants

Bethany M Henrick, Stephanie Chew, Giorgio Casaburi, Heather K Brown, Steven A Frese, You Zhou, Mark A Underwood, Jennifer T Smilowitz, Bethany M Henrick, Stephanie Chew, Giorgio Casaburi, Heather K Brown, Steven A Frese, You Zhou, Mark A Underwood, Jennifer T Smilowitz

Abstract

Background: Infant gut dysbiosis, often associated with low abundance of bifidobacteria, is linked to impaired immune development and inflammation-a risk factor for increased incidence of several childhood diseases. We investigated the impact of B. infantis EVC001 colonization on enteric inflammation in a subset of exclusively breastfed term infants from a larger clinical study.

Methods: Stool samples (n = 120) were collected from infants randomly selected to receive either 1.8 × 1010 CFU B. infantis EVC001 daily for 21 days (EVC001) or breast milk alone (controls), starting at day 7 postnatal. The fecal microbiome was analyzed using 16S ribosomal RNA, proinflammatory cytokines using multiplexed immunoassay, and fecal calprotectin using ELISA at three time points: days 6 (Baseline), 40, and 60 postnatal.

Results: Fecal calprotectin concentration negatively correlated with Bifidobacterium abundance (P < 0.0001; ρ = -0.72), and proinflammatory cytokines correlated with Clostridiaceae and Enterobacteriaceae, yet negatively correlated with Bifidobacteriaceae abundance. Proinflammatory cytokines were significantly lower in EVC001-fed infants on days 40 and 60 postnatally compared to baseline and compared to control infants.

Conclusion: Our findings indicate that gut dysbiosis (absence of B. infantis) is associated with increased intestinal inflammation. Early addition of EVC001 to diet represents a novel strategy to prevent enteric inflammation during a critical developmental phase.

Conflict of interest statement

B.M.H., S.C., G.C., H.K.B., and S.A.F. are employees of Evolve Biosystems, a company focused on restoring the infant microbiome. J.T.S. previously worked as a consultant for Evolve Biosystems. The other authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Microscopic analysis of the infant gut microbiome. Gram stain light microscopy (a, b) and scanning electron microscopy (c, d) micrographs of diluted fecal samples on day 40 postnatal from the control infants (a, c) and infants fed EVC001 (b, d). Scale bars: 50 µm (a, b) and 5 µm (c, d)
Fig. 2
Fig. 2
Fecal calprotectin levels are dependent on the abundance of Bifidobacteriaceae. Forty fecal samples from day 40 postnatal were evaluated for the concentration of fecal calprotectin and Bifidobacteriaceae abundance (****P < 0.0001; rs = −0.72; a) and subdivided based on Bifidobacteriaceae abundance < or >25% (b). The data set is representative of at least three different experiments completed in duplicate and a non-parametric Wilcoxon rank-sum test was used to determine significance with the corresponding P values adjusted and considered statistically significant if *P < 0.05. **P < 0.01
Fig. 3
Fig. 3
Fecal cytokine signature of infants who received B. infantis EVC001. Radar plot representations of median cytokine concentrations [pg/mg] detected in fecal samples from the controls (n = 20) and infants fed Bifidobacterium infantis EVC001 (n = 20; EVC001) on a day 6 (Baseline), b day 40 postnatal, and c day 60 postnatal. Median values were adjusted to log scale, then normalized within each cytokine group from 0 to 1. Statistical analysis was completed using Wilcoxon rank-sum test. P values were adjusted using Bonferonni–Holm method and considered statistically significant if *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 4
Fig. 4
Principal coordinate analysis (PCoA) of global cytokine profiles according to group status. PCoA based on Bray–Curtis dissimilarity of global cytokine profiles between EVC001-fed infants and controls at a day 6 (Baseline), b day 40, and c day 60 postnatal
Fig. 5
Fig. 5
Correlations between specific gut taxa and intestinal inflammatory cytokine responses. Heatmap shows correlation between bacterial families and specific cytokines computed via Spearman correlation. P values are corrected using Benjamini–Hochberg procedure (false discovery rate) to estimate significant correlations between microbial taxonomic composition and specific cytokine concentration detected in the feces of exclusively breastfed infants at three time points (day 6 (Baseline), day 40, and day 60 postnatal). Each cytokine was tested in duplicate at three different time points. P values were adjusted and considered to be statistically significant if P < 0.05 (empty circle); P < 0.01 (semi-solid circle); P < 0.001 (solid circle)

References

    1. Olin A, et al. Stereotypic immune system development in newborn children. Cell. 2018;174:1277–1292. doi: 10.1016/j.cell.2018.06.045.
    1. Arrieta M-C, et al. Associations between infant fungal and bacterial dysbiosis and childhood atopic wheeze in a nonindustrialized setting. J. Allergy Clin. Immunol. 2018;142:424–434. doi: 10.1016/j.jaci.2017.08.041.
    1. Arrieta M-C, et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci. Transl. Med. 2015;7:307ra152-2. doi: 10.1126/scitranslmed.aab2271.
    1. Rhoads JM, et al. Infant colic represents gut inflammation and dysbiosis. J. Pediatr. 2018;203:55–61. doi: 10.1016/j.jpeds.2018.07.042.
    1. Fujimura KE, et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat. Med. 2016;22:1187–1191. doi: 10.1038/nm.4176.
    1. Kostic AD, et al. The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe. 2015;17:260–273. doi: 10.1016/j.chom.2015.01.001.
    1. Vatanen T, et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell. 2016;165:1–12. doi: 10.1016/j.cell.2016.05.056.
    1. Cox LM, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell. 2014;158:705–721. doi: 10.1016/j.cell.2014.05.052.
    1. Gevers D, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014;15:382–392. doi: 10.1016/j.chom.2014.02.005.
    1. Russell SL, et al. Perinatal antibiotic treatment affects murine microbiota, immune responses and allergic asthma. Gut Microbes. 2013;4:158–164. doi: 10.4161/gmic.23567.
    1. Cahenzli J, et al. Intestinal Microbial diversity during early-life colonization shapes long-term IgE levels. Cell Host Microbe. 2013;14:559–570. doi: 10.1016/j.chom.2013.10.004.
    1. Ho TTB, et al. Enteric dysbiosis and fecal calprotectin expression in premature infants. Pediatr. Res. 2019;85:361–368. doi: 10.1038/s41390-018-0254-y.
    1. Herrera OR, Christensen ML, Helms RA. Calprotectin: clinical applications in pediatrics. J. Pediatr. Pharm. Ther. 2016;21:308–321.
    1. Nakayuenyongsuk W, et al. Point-of-care fecal calprotectin monitoring in preterm infants at risk for necrotizing enterocolitis. J. Pediatr. 2018;196:98–103. doi: 10.1016/j.jpeds.2017.12.069.
    1. Campeotto F, et al. Fecal calprotectin: cutoff values for identifying intestinal distress in preterm infants. J. Pediatr. Gastroenterol. Nutr. 2009;48:507–510.
    1. Kapel N, et al. Faecal calprotectin in term and preterm neonates. J. Pediatr. Gastroenterol. Nutr. 2010;51:542–547. doi: 10.1097/MPG.0b013e3181e2ad72.
    1. Souwer Y, et al. IL-17 and IL-22 in atopic allergic disease. Curr. Opin. Immunol. 2010;22:821–826. doi: 10.1016/j.coi.2010.10.013.
    1. Liwen Z, et al. A low abundance of Bifidobacterium but not Lactobacillius in the feces of Chinese children with wheezing diseases. Medicine. 2018;97:e12745–e12746. doi: 10.1097/MD.0000000000012745.
    1. Sjögren YM, et al. Altered early infant gut microbiota in children developing allergy up to 5 years of age. Clin. Exp. Allergy. 2009;39:518–526. doi: 10.1111/j.1365-2222.2008.03156.x.
    1. Rizzetto L, et al. Connecting the immune system, systemic chronic inflammation and the gut microbiome: the role of sex. J. Autoimmun. 2018;92:12–34. doi: 10.1016/j.jaut.2018.05.008.
    1. Zhang Y, et al. Variations in early gut microbiome are associated with childhood eczema. FEMS Microbiol. Lett. 2019;366:fnz020. doi: 10.1093/femsle/fnz020.
    1. Maffeis C, et al. Association between intestinal permeability and faecal microbiota composition in Italian children with beta cell autoimmunity at risk for type 1 diabetes. Diabetes Metab. Res. Rev. 2016;32:700–709. doi: 10.1002/dmrr.2790.
    1. Hollander D. Crohn’s disease-a permeability disorder of the tight junction? Gut. 1988;29:1621–1624. doi: 10.1136/gut.29.12.1621.
    1. Al-Sadi R, Boivin M, Ma T. Mechanism of cytokine modulation of epithelial tight junction barrier. Front. Biosci. (Landmark Ed.) 2009;14:2765–2778. doi: 10.2741/3413.
    1. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012;336:1268–1273. doi: 10.1126/science.1223490.
    1. Shin N-R, Whon TW, Bae J-W. Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol. 2015;33:496–503. doi: 10.1016/j.tibtech.2015.06.011.
    1. Frese SA, et al. Persistence of supplemented Bifidobacterium longum subsp. infantis EVC001 in breastfed infants. mSphere. 2017;2:e00501-17. doi: 10.1128/mSphere.00501-17.
    1. Huda MN, et al. Bifidobacterium abundance in early infancy and vaccine response at 2 years of age. Pediatrics. 2019;143:e20181489. doi: 10.1542/peds.2018-1489.
    1. Sela DA, et al. The genome sequence of Bifidobacterium longum subsp. infantis reveals adaptations for milk utilization within the infant microbiome. Proc. Natl Acad. Sci. 2008;105:18964–18969. doi: 10.1073/pnas.0809584105.
    1. Henrick BM, et al. Elevated fecal pH indicates a profound change in the breastfed infant gut microbiome due to reduction of Bifidobacterium over the past century. mSphere. 2018;3:e00041-18. doi: 10.1128/mSphere.00041-18.
    1. Karav S, Casaburi G, Frese SA. Reduced colonic mucin degradation in breastfed infants colonized by Bifidobacterium longum subsp. infantis EVC001. FEBS Open Bio. 2018;8:1649–1657. doi: 10.1002/2211-5463.12516.
    1. Smilowitz JT, et al. Safety and tolerability of Bifidobacterium longum subspecies infantis EVC001 supplementation in healthy term breastfed infants: a phase I clinical trial. BMC Pediatr. 2017;17:1–11. doi: 10.1186/s12887-016-0759-7.
    1. Houser MC, et al. Stool immune profiles evince gastrointestinal inflammation in Parkinson’s disease. Mov. Disord. 2018;33:793–804. doi: 10.1002/mds.27326.
    1. Vázquez-Baeza Y, et al. EMPeror: a tool for visualizing high-throughput microbial community data. Gigascience. 2013;2:16. doi: 10.1186/2047-217X-2-16.
    1. Zeng MY, Inohara N, Nunez G. Mechanisms of inflammation-driven bacterial dysbiosis in the gut. Mucosal Immunol. 2016;10:18–26. doi: 10.1038/mi.2016.75.
    1. Orivuori L, et al. High level of fecal calprotectin at age 2 months as a marker of intestinal inflammation predicts atopic dermatitis and asthma by age 6. Clin. Exp. Allergy. 2015;45:928–939. doi: 10.1111/cea.12522.
    1. Wickramasinghe S, et al. Bifidobacteria grown on human milk oligosaccharides downregulate the expression of inflammation-related genes in Caco-2 cells. BMC Microbiol. 2015;15:172. doi: 10.1186/s12866-015-0508-3.
    1. Bergmann KR, et al. Bifidobacteria stabilize claudins at tight junctions and prevent intestinal barrier dysfunction in mouse necrotizing enterocolitis. Am. J. Pathol. 2013;182:1595–1606. doi: 10.1016/j.ajpath.2013.01.013.
    1. Guo S, et al. Secreted metabolites of Bifidobacterium infantis and Lactobacillus acidophilus protect immature human enterocytes from IL-1β-induced inflammation: a transcription profiling analysis. PLoS ONE. 2015;10:e0124549-19.
    1. Aragozzini F, et al. Indole-3-lactic acid as a tryptophan metabolite produced by Bifidobacterium spp. Appl. Environ. Microbiol. 1979;38:544–546.
    1. Ehrlich, A. M. et al. Bifidobacterium grown on human milk oligosaccharides produce tryptophan metabolite Indole-3-lactic acid that significantly decreases inflammation in intestinal cells in vitro. FASEB J. 32(1 Suppl), 359 (2018).

Source: PubMed

3
Abonnere