Analysis of immune, microbiota and metabolome maturation in infants in a clinical trial of Lactobacillus paracasei CBA L74-fermented formula

Paola Roggero, Nadia Liotto, Chiara Pozzi, Daniele Braga, Jacopo Troisi, Camilla Menis, Maria Lorella Giannì, Roberto Berni Canani, Lorella Paparo, Rita Nocerino, Andrea Budelli, Fabio Mosca, Maria Rescigno, Paola Roggero, Nadia Liotto, Chiara Pozzi, Daniele Braga, Jacopo Troisi, Camilla Menis, Maria Lorella Giannì, Roberto Berni Canani, Lorella Paparo, Rita Nocerino, Andrea Budelli, Fabio Mosca, Maria Rescigno

Abstract

Mother's milk is the best choice for infants nutrition, however when it is not available or insufficient to satisfy the needs of the infant, formula is proposed as an effective substitute. Here, we report the results of a randomized controlled clinical trial (NCT03637894) designed to evaluate the effects of two different dietary regimens (standard formula and Lactobacillus paracasei CBA L74-fermented formula) versus breastfeeding (reference group) on immune defense mechanisms (primary endpoint: secretory IgA, antimicrobial peptides), the microbiota and its metabolome (secondary outcomes), in healthy full term infants according to the type of delivery (n = 13/group). We show that the fermented formula, safe and well tolerated, induces an increase in secretory IgA (but not in antimicrobial peptides) and reduces the diversity of the microbiota, similarly, but not as much as, breastmilk. Metabolome analysis allowed us to distinguish subjects based on their dietary regimen and mode of delivery. Together, these results suggest that a fermented formula favors the maturation of the immune system, microbiota and metabolome.

Conflict of interest statement

This work was sponsored by Heinz Italia S.p.A. As an employee of Heinz Italia S.p.A., A.B. has contributed technologically in adapting the fermentation process to the generation of the fermented formula for infant use, and the sponsor has provided the two formulas (standard and fermented) for the study. Moreover, the sponsor has contributed funding for biological assays, including ELISA, kits and –OMICS analyses. A.B. did not play any role in study design, analysis or writing of the paper (except for the methodological section on the formulas). The remaining authors declare no competing interests.

Figures

Fig. 1. Study procedures.
Fig. 1. Study procedures.
Procedures related to collection of biological samples, medical evaluation and anthropometric measurements are shown in a timeline.
Fig. 2. Fermented formula drives sIgA production…
Fig. 2. Fermented formula drives sIgA production similarly to breastfeeding.
a, b sIgA levels according to mode of delivery (vaginal in a and caesarean section in b) and type of feeding were determined in faecal samples. n = 13 (VBM and VF) and n = 12 (VS, CBM, CS and CF) biologically independent samples. Significance determined by one-way ANOVA using TukeyHSD post hoc test. Box plots show the interquartile range (IQR), the horizontal lines show the median values and the whiskers indicate the minimum-to-maximum range. CBM caesarean delivery breast milk feeding, CF caesarean delivery fermented formula feeding, CS caesarean delivery standard formula feeding, VBM vaginal-delivery breast milk feeding, VF vaginal-delivery fermented formula feeding, VS vaginal-delivery standard formula feeding. Source data are provided as a Source Data file.
Fig. 3. Fermented formula but not standard…
Fig. 3. Fermented formula but not standard formula reduces microbiota diversity over time.
a Chao1 index of bacterial α-diversity of the faecal microbiota of infants at enrolment (T0) and visit 2 (T2). Significance determined by Wilcoxon signed-rank test for repeated measurements (two-tailed). Box plots show the interquartile range (IQR), the horizontal lines show the median values and the whiskers extend from the hinge no further than 1.5*IQR. CBM: caesarean delivery breast milk feeding, CF caesarean delivery fermented formula feeding, CS caesarean delivery standard formula feeding, VBM vaginal-delivery breast milk feeding, VF vaginal-delivery fermented formula feeding, VS vaginal-delivery standard formula feeding. b PCoA analysis of weighted UniFrac distance (β-diversity) of gut microbial communities according to the mode of delivery (caesarean—star; vaginal—sphere) and the type of feeding (BM—red; F—blue; S—orange). Left, enrolment; right, visit 2. c Interaction plots of Bifidobacterium abundance according to diets and delivery mode at T0 and T2. Each line represents individual newborns (solid lines caesarean delivery, C; dotted lines vaginal delivery, V). Significance determined accounting for individual baselines with a linear mixed model and ANOVA to test the interaction between groups and time, P = 0.011. BM breast milk feeding, F fermented formula feeding, S standard formula feeding. Source data of panels a and c are provided as a Source Data file.
Fig. 4. Metabolome clearly distinguishes newborns based…
Fig. 4. Metabolome clearly distinguishes newborns based on their dietary regimen.
a, b Metabolomic assessment at enrolment (a) and at visit 2 (b) according to the type of feeding. Left, a PLS-DA analysis is shown separating infants breastfed (BM, red dots), fed formula F (F, blue dots), or formula S (S, orange dots). On the right are shown those metabolites that differentiate the three diets (VIP score > 2) and their mean relative concentration. c, d Metabolomic assessment (PLS-DA 3D) at enrolment (c) and at visit 2 (d) according to the type of feeding (BM—red; F—blue; S—orange) and mode of delivery (caesarean—star; vaginal—sphere). CBM caesarean delivery breast milk feeding, CF caesarean delivery fermented formula feeding, CS caesarean delivery standard formula feeding, VBM vaginal-delivery breast milk feeding, VF vaginal-delivery fermented formula feeding, VS vaginal-delivery standard formula feeding.
Fig. 5. Microbe–metabolite correlation heatmap showing associations…
Fig. 5. Microbe–metabolite correlation heatmap showing associations between bacterial genus and the ANOVA and VIP-score-selected metabolites.
Spearman correlation coefficients were calculated for pairwise combinations of microbial abundances at the genus level (from 16S rRNA gene sequencing data) and metabolite intensities. In red boxes are reported clusters of metabolites either enriched (box 1) or reduced (box 2) in vaginally delivered breastfed infants. Box 2 metabolites are associated with genera highly abundant in breastfed infants; in orange boxes (3 and 4) are highlighted clusters of metabolites that are enriched in formula S-fed infants. The corresponding genera correlating with these metabolites are also highlighted with the same colour and numbering code.

References

    1. Howie PW. Protective effect of breastfeeding against infection in the first and second six months of life. Adv. Exp. Med. Biol. 2002;503:141–147.
    1. Lewis ZT, Mills DA. Differential establishment of bifidobacteria in the breastfed infant gut. Nestle Nutr. Inst. Workshop Ser. 2017;88:149–159.
    1. Jakaitis BM, Denning PW. Human breast milk and the gastrointestinal innate immune system. Clin. Perinatol. 2014;41:423–435.
    1. Stevens EE, Patrick TE, Pickler R. A history of infant feeding. J. Perinat. Educ. 2009;18:32–39.
    1. Thompson AL, Monteagudo-Mera A, Cadenas MB, Lampl ML, Azcarate-Peril MA. Milk- and solid-feeding practices and daycare attendance are associated with differences in bacterial diversity, predominant communities, and metabolic and immune function of the infant gut microbiome. Front Cell Infect. Microbiol. 2015;5:3.
    1. Bokulich NA, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci. Transl. Med. 2016;8:343ra382.
    1. Backhed F, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. 2015;17:690–703.
    1. Dominguez-Bello MG, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl Acad. Sci. USA. 2010;107:11971–11975.
    1. Shively CA, et al. Consumption of mediterranean versus western diet leads to distinct mammary gland microbiome populations. Cell Rep. 2018;25:47–56 e43.
    1. Niimi K, et al. Development of immune and microbial environments is independently regulated in the mammary gland. Mucosal Immunol. 2018;11:643–653.
    1. Parnanen K, et al. Maternal gut and breast milk microbiota affect infant gut antibiotic resistome and mobile genetic elements. Nat. Commun. 2018;9:3891.
    1. Sakwinska O, Bosco N. Host microbe interactions in the lactating mammary gland. Front. Microbiol. 2019;10:1863.
    1. 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;7:40597.
    1. Martin R, et al. Human milk is a source of lactic acid bacteria for the infant gut. J. Pediatr. 2003;143:754–758.
    1. Bezirtzoglou E, Tsiotsias A, Welling GW. Microbiota profile in feces of breast- and formula-fed newborns by using fluorescence in situ hybridization (FISH) Anaerobe. 2011;17:478–482.
    1. Ho NT, et al. Meta-analysis of effects of exclusive breastfeeding on infant gut microbiota across populations. Nat. Commun. 2018;9:4169.
    1. Vendt N, et al. Growth during the first 6 months of life in infants using formula enriched with Lactobacillus rhamnosus GG: double-blind, randomized trial. J. Hum. Nutr. Diet. 2006;19:51–58.
    1. Fransen F, et al. BALB/c and C57BL/6 mice differ in polyreactive IgA abundance, which impacts the generation of antigen-specific IgA and microbiota diversity. Immunity. 2015;43:527–540.
    1. Suez J, et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell. 2018;174:1406–1423 e1416.
    1. Vandenplas Y, De Greef E, Veereman G. Prebiotics in infant formula. Gut Microbes. 2014;5:681–687.
    1. Petherick A. Development: mother’s milk: a rich opportunity. Nature. 2010;468:S5–S7.
    1. Smilowitz JT, et al. The human milk metabolome reveals diverse oligosaccharide profiles. J. Nutr. 2013;143:1709–1718.
    1. Lemas DJ, et al. Exploring the contribution of maternal antibiotics and breastfeeding to development of the infant microbiome and pediatric obesity. Semin Fetal Neonatal Med. 2016;21:406–409.
    1. Vandenplas Y, Zakharova I, Dmitrieva Y. Oligosaccharides in infant formula: more evidence to validate the role of prebiotics. Br. J. Nutr. 2015;113:1339–1344.
    1. Newburg DS, Ko JS, Leone S, Nanthakumar NN. Human milk oligosaccharides and synthetic galactosyloligosaccharides contain 3′-, 4-, and 6′-galactosyllactose and attenuate inflammation in human T84, NCM-460, and H4 cells and intestinal tissue ex vivo. J. Nutr. 2016;146:358–367.
    1. Bertelsen RJ, Jensen ET, Ringel-Kulka T. Use of probiotics and prebiotics in infant feeding. Best. Pr. Res Clin. Gastroenterol. 2016;30:39–48.
    1. Hegar B, et al. The role of two human milk oligosaccharides, 2’-fucosyllactose and lacto-N-neotetraose, in infant nutrition. Pediatr. Gastroenterol. Hepatol. Nutr. 2019;22:330–340.
    1. Tsilingiri K, Rescigno M. Postbiotics: what else? Beneficial microbes. 2013;4:101–107.
    1. Tsilingiri K, et al. Probiotic and postbiotic activity in health and disease: comparison on a novel polarised ex-vivo organ culture model. Gut. 2012;61:1007–1015.
    1. Zagato E, et al. Lactobacillus paracasei CBA L74 metabolic products and fermented milk for infant formula have anti-inflammatory activity on dendritic cells in vitro and protective effects against colitis and an enteric pathogen in vivo. PLoS ONE. 2014;9:e87615.
    1. Nocerino R, et al. Cow’s milk and rice fermented with Lactobacillus paracasei CBA L74 prevent infectious diseases in children: a randomized controlled trial. Clin. Nutr. 2017;36:118–125.
    1. Corsello Giovanni, Carta Maurizio, Marinello Roberto, Picca Marina, De Marco Giulio, Micillo Maria, Ferrara Dante, Vigneri Patrizia, Cecere Gaetano, Ferri Pasqualina, Roggero Paola, Bedogni Giorgio, Mosca Fabio, Paparo Lorella, Nocerino Rita, Berni Canani Roberto. Preventive Effect of Cow’s Milk Fermented with Lactobacillus paracasei CBA L74 on Common Infectious Diseases in Children: A Multicenter Randomized Controlled Trial. Nutrients. 2017;9(7):669.
    1. Kato LM, Kawamoto S, Maruya M, Fagarasan S. Gut TFH and IgA: key players for regulation of bacterial communities and immune homeostasis. Immunol. Cell Biol. 2014;92:49–56.
    1. Pang T, Leach ST, Katz T, Day AS, Ooi CY. Fecal biomarkers of intestinal health and disease in children. Front. Pediatr. 2014;2:6.
    1. Giorgetti G, et al. Interactions between innate immunity, microbiota, and probiotics. J. Immunol. Res. 2015;2015:501361.
    1. Chu DM, et al. Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat. Med. 2017;23:314–326.
    1. Fanos V, Antonucci R, Atzori L. Metabolomics in the developing infant. Curr. Opin. Pediatr. 2013;25:604–611.
    1. Gutierrez-Garcia AG, Contreras CM, Diaz-Marte C. Myristic acid in amniotic fluid produces appetitive responses in human newborns. Early Hum. Dev. 2017;115:32–37.
    1. Contreras CM, et al. Myristic acid produces anxiolytic-like effects in Wistar rats in the elevated plus maze. Biomed. Res. Int. 2014;2014:492141.
    1. Louis P, Flint HJ. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017;19:29–41.
    1. Hamer HM, et al. Review article: the role of butyrate on colonic function. Aliment Pharm. Ther. 2008;27:104–119.
    1. Domej W, et al. Colobronchial fistula: a rare complication of Crohn’s colitis. Am. Rev. Respir. Dis. 1990;142:1225–1227.
    1. Kaakoush NO. Insights into the role of Erysipelotrichaceae in the human host. Front. Cell Infect. Microbiol. 2015;5:84.
    1. Zagato E, et al. Endogenous murine microbiota member Faecalibaculum rodentium and its human homologue protect from intestinal tumour growth. Nat. Microbiol. 2020;5:511–524.
    1. Lei J, et al. Nutritional and regulatory role of branched-chain amino acids in lactation. Front. Biosci. 2012;17:2725–2739.
    1. Wexler AG, Goodman AL. An insider’s perspective: bacteroides as a window into the microbiome. Nat. Microbiol. 2017;2:17026.
    1. Smalley D, Rocha ER, Smith CJ. Aerobic-type ribonucleotide reductase in the anaerobe Bacteroides fragilis. J. Bacteriol. 2002;184:895–903.
    1. Scheiman J, et al. Meta-omics analysis of elite athletes identifies a performance-enhancing microbe that functions via lactate metabolism. Nat. Med. 2019;25:1104–1109.
    1. Agostoni C, et al. Growth patterns of breast fed and formula fed infants in the first 12 months of life: an Italian study. Arch. Dis. Child. 1999;81:395–399.
    1. Villar J, et al. International standards for newborn weight, length, and head circumference by gestational age and sex: the Newborn Cross-Sectional Study of the INTERGROWTH-21st Project. Lancet. 2014;384:857–868.
    1. Ma G, et al. Validation of a new pediatric air-displacement plethysmograph for assessing body composition in infants. Am. J. Clin. Nutr. 2004;79:653–660.
    1. Ellis KJ, et al. Body-composition assessment in infancy: air-displacement plethysmography compared with a reference 4-compartment model. Am. J. Clin. Nutr. 2007;85:90–95.
    1. Fomon SJ, Haschke F, Ziegler EE, Nelson SE. Body composition of reference children from birth to age 10 years. Am. J. Clin. Nutr. 1982;35:1169–1175.
    1. Furet JP, et al. Comparative assessment of human and farm animal faecal microbiota using real-time quantitative PCR. FEMS Microbiol. Ecol. 2009;68:351–362.
    1. Milani C, et al. Assessing the fecal microbiota: an optimized ion torrent 16S rRNA gene-based analysis protocol. PLoS ONE. 2013;8:e68739.
    1. Hall M, Beiko RG. 16S rRNA gene analysis with QIIME2. Methods Mol. Biol. 2018;1849:113–129.
    1. Lozupone C, Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl. Environ. Microbiol. 2005;71:8228–8235.
    1. Quast C, et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013;41:D590–D596.
    1. Sumner LW, Huhman DV, Urbanczyk-Wochniak E, Lei Z. Methods, applications and concepts of metabolite profiling: secondary metabolism. EXS. 2007;97:195–212.

Source: PubMed

3
Se inscrever