Probiotic supplementation and associated infant gut microbiome and health: a cautionary retrospective clinical comparison

C Quin, M Estaki, D M Vollman, J A Barnett, S K Gill, D L Gibson, C Quin, M Estaki, D M Vollman, J A Barnett, S K Gill, D L Gibson

Abstract

While probiotics are a multi-billion dollar industry, there is little evidence to show that supplementing infants provides any health benefits. We conducted an observational study where 35 of 86 participating mothers self-administered probiotics during breastfeeding, as well as directly to their infants. The primary objective was to determine if probiotic exposure influenced the infants' fecal microbiome while the secondary objective assessed associated changes to the mothers' breast milk immunity and infant health. Analysis of infant fecal microbiome throughout the first 6 months of life revealed that probiotics were associated with higher abundances of Bifidobacterium at week 1 only. Short-chain fatty acid production and predicted metagenomic functions of the microbial communities were not altered. While probiotics did not alter breast milk immune markers, fecal sIgA responses were higher among probiotic supplemented infants. Surprisingly, this was not associated with better health outcomes, as the probiotic cohort had higher incidences of mucosal-associated illnesses as toddlers. This retrospective clinical comparison suggests that probiotic exposure during infancy has limited effects on gut microbial composition yet is associated with increased infection later in life. These correlative findings caution against probiotic supplementation during infancy until rigorous controlled follow-up studies determining their safety and efficacy have occurred.

Trial registration: ClinicalTrials.gov NCT03297801.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Flow of participants through the trial.
Figure 2
Figure 2
Alpha diversity measures of fecal microbiota over a 6-month period of probiotics and no probiotics exposed infants. The alpha diversity in the infants’ stool did not differ between the two groups at any time point (1 week to 6 months of age). While the two groups were not different from each other, Faith’s phylogenetic diversity in the no probiotics group significantly increased at months 4 and 5 when compared to the first week, whereas the probiotics group showed no significant increases. *Denotes P < 0.05.
Figure 3
Figure 3
Beta diversity assessment of microbial communities over a 6-month period of probiotics and no probiotics exposed infants. (A) PCoA plots based on the Bray-Curtis dissimilarity distance show no distinct clustering of microbial communities at any age except for at 1 week. (B) Table summary of PERMANOVA results ran on the Bray-Curtis dissimilarity, and weighted and unweighted UniFrac distances between the two groups. The values at the intersect of a blue cell (probiotics group) and an orange cell (no probiotics group) show the estimated P value of the corresponding time points.
Figure 4
Figure 4
Differences in the abundance of taxa between the groups were assessed using two complimentary approaches at each time-point. (A) Differences in the relative abundance of Bifidobacterium spp. in the probiotics group at one week of age was detected as significantly different using ANCOM (P < 0.05, F-statistic = 7.5). (B) LEfSe results showing significantly different taxa between fecal samples between probiotics and no probiotics infants at one week of age. The cladogram reports the taxa showing different abundance values according to LEfSe. Colors indicate the lineages that are encoded within corresponding samples. Higher taxa abundance in the probiotics group is colored blue whereas higher taxa abundance in the no probiotics group is colored orange. (C) Infants directly supplemented with BioGaia according to the manufacturers recommendations show similar results to (A and B). P < 0.05.
Figure 5
Figure 5
Abundances of fecal SCFAs acetic acid, butyric acid, and propionic acid between probiotics and no probiotics exposed infants at 5 months of age expressed as mass % (g SCFA/g dry weight stool).
Figure 6
Figure 6
Immune markers in breast milk of mothers with or without probiotic supplementation at 5 months. The scatter dot plot shows the mean and standard error mean.
Figure 7
Figure 7
Infant exposure to probiotics correlates with increased mucosal illness during the first 2 years of life and corresponding immune responses at 5 months of age. (A) Infants exposed to probiotics had significantly higher reported mucosal infections compared to the no probiotics group but were able to clear infections at similar rates. (B) Probiotic exposure during the first 6 months is associated with no changes in breast milk sIgA but modest increases in fecal sIgA. (C) There were no differences in either breast milk or infant stool IgE levels. *Denotes P < 0.05.

References

    1. Abrahamsson TR, et al. Low gut microbiota diversity in early infancy precedes asthma at school age. Clin Exp Allergy. 2014;44(6):842–850. doi: 10.1111/cea.12253.
    1. Frank DNST, et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(34):13780–13785. doi: 10.1073/pnas.0706625104.
    1. Ott SJ, et al. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004;53(5):685–693. doi: 10.1136/gut.2003.025403.
    1. Manichanh C, et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut. 2006;55(2):205–211. doi: 10.1136/gut.2005.073817.
    1. Wang M, et al. Reduced diversity in the early fecal microbiota of infants with atopic eczema. J Allergy Clin Immunol. 2008;121(1):129–134. doi: 10.1016/j.jaci.2007.09.011.
    1. Geuking MB, McCoy KD, Macpherson AJ. Metabolites from intestinal microbes shape Treg. Cell Res. 2013;23(12):1339–1340. doi: 10.1038/cr.2013.125.
    1. Ahern PP, Faith JJ, Gordon JI. Mining the human gut microbiota for effector strains that shape the immune system. Immunity. 2014;40(6):815–823. doi: 10.1016/j.immuni.2014.05.012.
    1. Wu W, et al. Microbiota metabolite short-chain fatty acid acetate promotes intestinal IgA response to microbiota which is mediated by GPR43. Mucosal Immunol. 2017;10(4):946–956. doi: 10.1038/mi.2016.114.
    1. Ishikawa T, Nanjo F. Dietary cycloinulooligosaccharides enhance intestinal immunoglobulin A production in mice. Biosci Biotechnol Biochem. 2009;73(3):677–682. doi: 10.1271/bbb.80733.
    1. Renz H, Brandtzaeg P, Hornef M. The impact of perinatal immune development on mucosal homeostasis and chronic inflammation. Nature reviews. Immunology. 2011;12(1):9–23. doi: 10.1038/nri3112.
    1. FAO/WHO. Guidelines for evaluating probiotics in food. Report of a joint FAO/WHO working group on drafting guidelines for the evaluations of probiotics in food; Ontario, Canada. (2002).
    1. D.O.H. Government of the District of Columbia, Department of Health. Fatal Gastrointestinal Mucormycosis in an Infant Following Ingestion of Conaminated Dietary Supplement; (2014).
    1. Zuccotti G, et al. Probiotics for prevention of atopic diseases in infants: systematic review and meta-analysis. Allergy. 2015;70(11):1356–1371. doi: 10.1111/all.12700.
    1. Topcuoglu S, Gursoy T, Ovali F, Serce O, Karatekin G. A new risk factor for neonatal vancomycin-resistant Enterococcus colonisation: bacterial probiotics. J Matern Fetal Neonatal Med. 2015;28(12):1491–1494. doi: 10.3109/14767058.2014.958462.
    1. Abe AM, Gregory PJ, Hein DJ, Cochrane ZR, Wilson AF. Survey and Systematic Literature Review of Probiotics Stocked in Academic Medical Centers within the United States. Hosp Pharm. 2013;48(10):834–847. doi: 10.1310/hpj4810-834.
    1. Williams MD, Ha CY, Ciorba MA. Probiotics as therapy in gastroenterology: a study of physician opinions and recommendations. J Clin Gastroenterol. 2010;44(9):631–636.
    1. Hickey Y, Corish C. Irish dietitians’ opinions and use of probiotics in patients with Clostridium difficile-associated disease. J Clin Gastroenterol. 2011;45(6):568–569. doi: 10.1097/MCG.0b013e318209cb43.
    1. Hammerman C, Bin-Nun A, Kaplan M. Safety of probiotics: comparison of two popular strains. BMJ. 2006;333(7576):1006–1008. doi: 10.1136/.
    1. Reuter G. The Lactobacillus and Bifidobacterium microflora of the human intestine: composition and succession. Curr Issues Intest Microbiol. 2001;2(2):43–53.
    1. Gasbarrini, G., Bonvicini, F., Gramenzi, A. Probiotics History. J Clin Gastroenterol. Nov/Dec 2016;50 Suppl 2, Proceedings from the 8th Probiotics, Prebiotics & New Foods for Microbiota and Human Health meeting held in Rome, Italy on September 13–15, S116–S119 (2015).
    1. Lozupone C, Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl Environ Microbiol. 2005;71(12):8228–8235. doi: 10.1128/AEM.71.12.8228-8235.2005.
    1. Mandal S, et al. Analysis of composition of microbiomes: a novel method for studying microbial composition. Microb Ecol Health Dis. 2015;26:27663.
    1. Segata N, et al. Metagenomic biomarker discovery and explanation. Genome biology. 2011;12(6):R60. doi: 10.1186/gb-2011-12-6-r60.
    1. Langille MG, et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol. 2013;31(9):814–821. doi: 10.1038/nbt.2676.
    1. Smith PM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569–573. doi: 10.1126/science.1241165.
    1. Turfkruyer M, Verhasselt V. Breast milk and its impact on maturation of the neonatal immune system. Current opinion in infectious diseases. 2015;28(3):199–206. doi: 10.1097/QCO.0000000000000165.
    1. Newburg DS, Walker WA. Protection of the neonate by the innate immune system of developing gut and of human milk. Pediatr Res. 2007;61(1):2–8. doi: 10.1203/01.pdr.0000250274.68571.18.
    1. Bron PA, van Baarlen P, Kleerebezem M. Emerging molecular insights into the interaction between probiotics and the host intestinal mucosa. Nat Rev Microbiol. 2011;10(1):66–78. doi: 10.1038/nrmicro2690.
    1. Jain N, Walker WA. Diet and host-microbial crosstalk in postnatal intestinal immune homeostasis. Nature reviews. Gastroenterology & hepatology. 2015;12(1):14–25. doi: 10.1038/nrgastro.2014.153.
    1. Goldsmith F, O’Sullivan A, Smilowitz JT, Freeman SL. Lactation and Intestinal Microbiota: How Early Diet Shapes the Infant Gut. J Mammary Gland Biol Neoplasia. 2015;20(3-4):149–158. doi: 10.1007/s10911-015-9335-2.
    1. Martin R, et al. Cultivation-independent assessment of the bacterial diversity of breast milk among healthy women. Res Microbiol. 2007;158(1):31–37. doi: 10.1016/j.resmic.2006.11.004.
    1. Donnet-Hughes A, et al. Potential role of the intestinal microbiota of the mother in neonatal immune education. Proc Nutr Soc. 2010;69(3):407–415. doi: 10.1017/S0029665110001898.
    1. Scholtens PA, Oozeer R, Martin R, Amor KB, Knol J. The early settlers: intestinal microbiology in early life. Annu Rev Food Sci Technol. 2012;3:425–447. doi: 10.1146/annurev-food-022811-101120.
    1. Dotterud CK, et al. Does Maternal Perinatal Probiotic Supplementation Alter the Intestinal Microbiota of Mother and Child? J Pediatr Gastroenterol Nutr. 2015;61(2):200–207. doi: 10.1097/MPG.0000000000000781.
    1. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nature reviews. Immunology. 2009;9(5):313–323. doi: 10.1038/nri2515.
    1. Gibson GR, Wang X. Regulatory effects of bifidobacteria on the growth of other colonic bacteria. J Appl Bacteriol. 1994;77(4):412–420. doi: 10.1111/j.1365-2672.1994.tb03443.x.
    1. Ouwehand A, Isolauri E, Salminen S. The role of the intestinal microflora for the development of the immune system in early childhood. Eur J Nutr. 2002;41(Suppl 1):I32–37.
    1. Baldassarre, M. E. 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. 8(11) (2016).
    1. Cash HL, Whitham CV, Behrendt CL, Hooper LV. Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science. 2006;313(5790):1126–1130. doi: 10.1126/science.1127119.
    1. Sonnenburg JL, Chen CT, Gordon JI. Genomic and metabolic studies of the impact of probiotics on a model gut symbiont and host. PLoS Biol. 2006;4(12):e413. doi: 10.1371/journal.pbio.0040413.
    1. Panigrahi P, et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017;548(7668):407–412. doi: 10.1038/nature23480.
    1. Panigrahi P, et al. Long-term colonization of a Lactobacillus plantarum synbiotic preparation in the neonatal gut. J Pediatr Gastroenterol Nutr. 2008;47(1):45–53. doi: 10.1097/MPG.0b013e31815a5f2c.
    1. Janda JM, Abbott SL. 16S rRNA gene sequencing for bacterial identification in the diagnostic laboratory: pluses, perils, and pitfalls. J Clin Microbiol. 2007;45(9):2761–2764. doi: 10.1128/JCM.01228-07.
    1. Awad A, Al-Shaye D. Public awareness, patterns of use and attitudes toward natural health products in Kuwait: a cross-sectional survey. BMC Complement Altern Med. 2014;14:105. doi: 10.1186/1472-6882-14-105.
    1. Health Canada. Natural Health Product Tracking Survey - 2010 Final Report. 2010.
    1. Arthur JC, et al. VSL#3 probiotic modifies mucosal microbial composition but does not reduce colitis-associated colorectal cancer. Sci Rep. 2013;3:2868. doi: 10.1038/srep02868.
    1. Besselink MG, et al. Probiotic prophylaxis in predicted severe acute pancreatitis: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;371(9613):651–659. doi: 10.1016/S0140-6736(08)60207-X.
    1. Kennedy J, Wang CC, Wu CH. Patient Disclosure about Herb and Supplement Use among Adults in the US. Evid Based Complement Alternat Med. 2008;5(4):451–456. doi: 10.1093/ecam/nem045.
    1. Kemper KJ, O’Connor KG. Pediatricians’ recommendations for complementary and alternative medical (CAM) therapies. Ambul Pediatr. 2004;4(6):482–487. doi: 10.1367/A04-050R.1.
    1. Sawni A, Thomas R. Pediatricians’ attitudes, experience and referral patterns regarding Complementary/Alternative Medicine: a national survey. BMC Complement Altern Med. 2007;7:18. doi: 10.1186/1472-6882-7-18.
    1. Government of Canada. ARCHIVED - Charting A Course: Refining Canada’s Approach to Regulating Natural Health Products (2010).
    1. Faul F, Erdfelder E, Lang AG, Buchner A. G*Power 3: a flexible statistical power analysis program for the social, behavioral, and biomedical sciences. Behavior research methods. 2007;39(2):175–191. doi: 10.3758/BF03193146.
    1. Faul F, Erdfelder E, Buchner A, Lang AG. Statistical power analyses using G*Power 3.1: tests for correlation and regression analyses. Behavior research methods. 2009;41(4):1149–1160. doi: 10.3758/BRM.41.4.1149.
    1. Azad MB, et al. Gut microbiota of healthy Canadian infants: profiles by mode of delivery and infant diet at 4 months. CMAJ. 2013;185(5):385–394. doi: 10.1503/cmaj.121189.
    1. STROBE. STROBE Statement, strengthening the reporting of observational studies in epidemiology. STROBE Statement, strengthening the reporting of observational studies in epidemiology; (2009).
    1. Ramage SM, McCargar LJ, Berglund C, Harber V, Bell RC. Assessment of Pre-Pregnancy Dietary Intake with a Food Frequency Questionnaire in Alberta Women. Nutrients. 2015;7(8):6155–6166. doi: 10.3390/nu7085277.
    1. Ramakrishnan R, Venkatarao T, Koya PK, Kamaraj P. Influence of recall period on estimates of diarrhoea morbidity in infants in rural Tamilnadu. Indian J Public Health. 1999;43(4):136–139.
    1. Martorell R, Habicht JP, Yarbrough C, Lechtig A, Klein RE. Underreporting in fortnightly recall morbidity surveys. J Trop Pediatr Environ Child Health. 1976;22(3):129–134.
    1. Imhoff-Kunsch B, et al. Prenatal docosahexaenoic acid supplementation and infant morbidity: randomized controlled trial. Pediatrics. 2011;128(3):e505–512.
    1. Gorzelak MA, et al. Methods for Improving Human Gut Microbiome Data by Reducing Variability through Sample Processing and Storage of Stool. PLoS One. 2015;10(8):e0134802. doi: 10.1371/journal.pone.0134802.
    1. Klindworth A, et al. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic acids research. 2013;41(1):e1. doi: 10.1093/nar/gks808.
    1. Nextera. Nextera XT Sample Preparation Guide (2012).
    1. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nature methods. 7(5), 335–336 (2010).
    1. Callahan, B. J. et al. DADA2: High-resolution sample inference from Illumina amplicon data. Nature methods. 13(7), 581–583 (2016).
    1. Katoh, K. & Standley, D. M. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol. 30(4), 772–780 (2013).
    1. Price MN, Dehal PS, Arkin AP. FastTree 2–approximately maximum-likelihood trees for large alignments. PloS one. 2010;5(3):e9490. doi: 10.1371/journal.pone.0009490.
    1. Rognes T, Flouri T, Nichols B, Quince C, Mahe F. VSEARCH: a versatile open source tool for metagenomics. PeerJ. 2016;4:e2584. doi: 10.7717/peerj.2584.
    1. Parks DH, Tyson GW, Hugenholtz P, Beiko RG. STAMP: statistical analysis of taxonomic and functional profiles. Bioinformatics. 2014;30(21):3123–3124. doi: 10.1093/bioinformatics/btu494.
    1. Estaki M, et al. Cardiorespiratory fitness as a predictor of intestinal microbial diversity and distinct metagenomic functions. Microbiome. 2016;4(1):42. doi: 10.1186/s40168-016-0189-7.
    1. Brown, K. et al. Prolonged antibiotic treatment induces a diabetogenic intestinal microbiome that accelerates diabetes in NOD mice. The ISME journal. (2015).
    1. Wang YNU, Wright ST, Warton DI. mvabund- an R package for model-based analysis of multivariate abundance data. methods in Ecology and Evolution. 2012;3(3):471–474. doi: 10.1111/j.2041-210X.2012.00190.x.
    1. R Core Team. R: A language and environment for statistical computing. R Foundation for Statistical Computing. Vienna, Austria (2014).
    1. Burnham, K. P., Anderson, D. R. Model Selection and Multimodel Inference. In: Springer, ed. Model Selection and Multimodel Inference. A PracticalInformation-Theoretic Approach. seconded (2002).
    1. Bartoñ, K. MuMIn: multi-model inference (R package version 0.40.4)., (2009).

Source: PubMed

Подписаться