Oral supplementation of healthy adults with 2'-O-fucosyllactose and lacto-N-neotetraose is well tolerated and shifts the intestinal microbiota

Emma Elison, Louise K Vigsnaes, Laura Rindom Krogsgaard, Julie Rasmussen, Nikolaj Sørensen, Bruce McConnell, Thierry Hennet, Morten O A Sommer, Peter Bytzer, Emma Elison, Louise K Vigsnaes, Laura Rindom Krogsgaard, Julie Rasmussen, Nikolaj Sørensen, Bruce McConnell, Thierry Hennet, Morten O A Sommer, Peter Bytzer

Abstract

The gut microbiota has been established as an important player influencing many aspects of human physiology. Breast milk, the first diet for an infant, contains human milk oligosaccharides (HMO) that shape the infant's gut microbiota by selectively stimulating the growth of specific bacteria, especially bifidobacteria. In addition to their bifidogenic activity, the ability of HMO to modulate immune function and the gut barrier makes them prime candidates to restore a beneficial microbiota in dysbiotic adults and provide health benefits. We conducted a parallel, double-blind, randomised, placebo-controlled, HMO-supplementation study in 100 healthy, adult volunteers, consuming chemically produced 2'-O-fucosyllactose (2'FL) and/or lacto-N-neotetraose (LNnT) at various daily doses and mixes or placebo for 2 weeks. All participants completed the study without premature discontinuation. Supplementation of 2'FL and LNnT at daily doses up to 20 g was shown to be safe and well tolerated, as assessed using the gastrointestinal symptoms rating scale. 16S rRNA sequencing analysis showed that HMO supplementation specifically modified the adult gut microbiota with the primary impact being substantial increases in relative abundance of Actinobacteria and Bifidobacterium in particular and a reduction in relative abundance of Firmicutes and Proteobacteria. This study provides the first set of data on safety, tolerance and impact of HMO on the adult gut microbiota. Collectively, the results from this study show that supplementing the diet with HMO is a valuable strategy to shape the human gut microbiota and specifically promote the growth of beneficial bifidobacteria.

Keywords: 2′FL 2′-O-fucosyllactose; GI gastrointestinal; GSRS gastrointestinal symptom rating scale; HMO human milk oligosaccharide; LNnT lacto-N-neotetraose; OTU operational taxonomic units; 2′-O-Fucosyllactose; Clinical study; Lacto-N-neotetraose; Safety; Tolerance.

Figures

Fig. 1
Fig. 1
Flow chart of the study. A total of 110 healthy, adult volunteers were screened for eligibility to participate in the study; 100 of them were randomised to one of the following intervention groups: 2′-O-fucosyllactose (2′FL), lacto-N-neotetraose (LNnT) or 2:1 mix of 2′FL:LNnT, each in three daily doses of 5, 10 or 20 g, or 2 g glucose as placebo. GSRS, gastrointestinal symptom rating scale.
Fig. 2
Fig. 2
Gastrointestinal symptom rating scale (GSRS) scores at the end of the intervention. Scores ranged from 1 (no discomfort) to 7 (very severe discomfort). (a) 2′-O-fucosyllactose (2′FL) supplementation groups and placebo group; (b) lacto-N-neotetraose (LNnT) supplementation groups and placebo group; (c) 2′FL:LNnT (2:1) mix supplementation groups and placebo group. , 20 g, , 10 g, , 5 g, , placebo. GSRS scores at the end of intervention for placebo and the intervention group were compared using a two-way ANOVA and Bonferroni’s multiple comparisons correction. * Significantly different between the intervention group and the placebo group (P<0·05).
Fig. 3
Fig. 3
Principal coordinates analysis plot of generalised UniFrac distances for all samples collected. (a) Before intervention and (b) after intervention. Phyla abundances are overlaid in blue. Samples are divided into intervention groups with the label at the centre of gravity for each group. Before intervention, there is no clear pattern. After intervention, the human milk oligosaccharide supplementation groups followed an axis of increasing Actinobacteria and decreasing Firmicutes for increasing doses.
Fig. 4
Fig. 4
Relative abundance of faecal bacteria at the phylum level. (a and b) Phyla level in the three 2′-O-fucosyllactose (2′FL) groups receiving 5, 10 or 20 g and placebo before and after intervention; (c and d) phyla level in the three lacto-N-neotetraose (LNnT) groups receiving 5, 10 or 20 g and placebo before and after intervention; (e and f) phyla level in the three mix groups receiving 5, 10 or 20 g of 2′FL:LNnT (2:1) and placebo before and after intervention. * Significantly different between before and after intervention (P<0·05). , Actinobacteria; , Bacteroidetes; , Firmicutes; , Proteobacteria; , others.
Fig. 5
Fig. 5
Change in sequence abundance of Actinobacteria (a) andBifidobacterium (b). The box represents the median and the 25th to 75th percentiles. The whiskers represent the smallest and largest changes observed. * Significantly different between the intervention group and the placebo group (P<0·05). 2′FL, 2′-O-fucosyllactose; LNnT, lacto-N-neotetraose; Mix, 2′FL:LNnT (2:1).
Fig. 6
Fig. 6
Change in sequence abundance of three operational taxonomic units (OTU) showing high similarity to the described Bifidobacterium species,B. adolescentis, B. longum and B. bifidum. The box represents the median and the 25th to 75th percentiles. The whiskers represent the smallest and largest changes observed. * Significantly different between the intervention group and the placebo group (P<0·05). 2′FL, 2′-O-fucosyllactose; LNnT, lacto-N-neotetraose; Mix, 2′FL:LNnT (2:1). ,s1_r64; , s1_r2031; , s1_r379.
Fig. 7
Fig. 7
Relative abundance of faecal bacteria at the genus level from before and after intervention. The eighteen genera, Bifidobacterium,Bacteroides, Barnesiella,Parabacteroides, Prevotella,Alistipes, Lactobacillus,Eubacterium, Blautia,Coprococcus, Dorea, Lachnospiracea incertae sedis, Roseburia,Faecalibacterium, Ruminococcus,Dialister, Escherichia/Shigella andAkkermansia, selected have been associated with obesity, irritable bowel syndrome or inflammatory bowel disease. (a) The mean of relative abundance of eighteen genera from the three intervention groups given 10 g of human milk oligosaccharide (HMO). (b) Relative abundance of eighteen genera from the placebo. Values are means, with their standard errors represented by vertical bars. Multiple t test was performed followed by a calculation of false discovery rate indicated as an adjusted P-value. * Significantly different between the groups (P<0·05). Lachn_inc_sedis,Lachnospiracea incertae sedis; , Before; , after.
Fig. 8
Fig. 8
SCFA in faeces. Concentrations are given as mm/g faeces of acetate (a), butyrate (b) and propionate (c) in samples from each intervention group and placebo before () and after () intervention. The box represents the median and the 25th to 75th percentiles. The whiskers represent the smallest and largest concentrations measured. 2′FL, 2′-O-fucosyllactose; LNnT, lacto-N-neotetraose; Mix, 2′FL:LNnT (2:1).

References

    1. Nicholson JK, Holmes E, Kinross J, et al. (2012) Host-gut microbiota metabolic interactions. Science 336, 1262–1267.
    1. Utzschneider KM, Kratz M, Damman CJ, et al. (2016) Mechanisms linking the gut microbiome and glucose metabolism. J Clin Endocrinol Metab 101, 1445–1454.
    1. Wu HJ & Wu E (2012) The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 3, 4–14.
    1. Belkaid Y & Hand TW (2014) Role of the microbiota in immunity and inflammation. Cell 157, 121–141.
    1. Shi HN & Walker A (2004) Bacterial colonization and the development of intestinal defences. Can J Gastroenterol 18, 493–500.
    1. Le Chatelier E, Nielsen T, Qin J, et al. (2013) Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541–546.
    1. Turnbaugh PJ, Hamady M, Yatsunenko T, et al. (2009) A core gut microbiome in obese and lean twins. Nature 457, 480–484.
    1. Qin N, Yang F, Li A, et al. (2014) Alterations of the human gut microbiome in liver cirrhosis. Nature 513, 59–64.
    1. Brown JM & Hazen SL (2015) The gut microbial endocrine organ: bacterially derived signals driving cardiometabolic diseases. Annu Rev Med 66, 343–359.
    1. Karlsson FH, Tremaroli V, Nookaew I, et al. (2013) Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 498, 99–103.
    1. Qin J, Li Y, Cai Z, et al. (2012) A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490, 55–60.
    1. Palm NW, de Zoete MR, Cullen TW, et al. (2014) Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 158, 1000–1010.
    1. Zackular JP, Rogers MA, Ruffin MTt, et al. (2014) The human gut microbiome as a screening tool for colorectal cancer. Cancer Prev Res (Phila) 7, 1112–1121.
    1. Fujimura KE, Slusher NA, Cabana MD, et al. (2010) Role of the gut microbiota in defining human health. Expert Rev Anti Infect Ther 8, 435–454.
    1. van Nood E, Vrieze A, Nieuwdorp M, et al. (2013) Duodenal infusion of donor feces for recurrent Clostridium difficile . N Engl J Med 368, 407–415.
    1. Ballard O & Morrow AL (2013) Human milk composition: nutrients and bioactive factors. Pediatr Clin North Am 60, 49–74.
    1. Bezirtzoglou E, Tsiotsias A & Welling GW (2011) Microbiota profile in feces of breast- and formula-fed newborns by using fluorescence in situ hybridization (FISH). Anaerobe 17, 478–482.
    1. Gabrielli O, Zampini L, Galeazzi T, et al. (2011) Preterm milk oligosaccharides during the first month of lactation. Pediatrics 128, e1520–e1531.
    1. ten Bruggencate SJ, Bovee-Oudenhoven IM, Feitsma AL, et al. (2014) Functional role and mechanisms of sialyllactose and other sialylated milk oligosaccharides. Nutr Rev 72, 377–389.
    1. Rudloff S, Obermeier S, Borsch C, et al. (2006) Incorporation of orally applied (13)C-galactose into milk lactose and oligosaccharides. Glycobiology 16, 477–487.
    1. Garrido D, Dallas DC & Mills DA (2013) Consumption of human milk glycoconjugates by infant-associated bifidobacteria: mechanisms and implications. Microbiology 159, 649–664.
    1. Ruiz-Moyano S, Totten SM, Garrido DA, et al. (2013) Variation in consumption of human milk oligosaccharides by infant gut-associated strains of Bifidobacterium breve . Appl Environ Microbiol 79, 6040–6049.
    1. Yu ZT, Chen C, Kling DE, et al. (2013) The principal fucosylated oligosaccharides of human milk exhibit prebiotic properties on cultured infant microbiota. Glycobiology 23, 169–177.
    1. Yu ZT, Chen C & Newburg DS (2013) Utilization of major fucosylated and sialylated human milk oligosaccharides by isolated human gut microbes. Glycobiology 23, 1281–1292.
    1. Marcobal A, Barboza M, Froehlich JW, et al. (2010) Consumption of human milk oligosaccharides by gut-related microbes. J Agric Food Chem 58, 5334–5340.
    1. Marcobal A & Sonnenburg JL (2012) Human milk oligosaccharide consumption by intestinal microbiota. Clin Microbiol Infect 18, Suppl. 4, 12–15.
    1. Ward RE, Ninonuevo M, Mills DA, et al. (2007) In vitro fermentability of human milk oligosaccharides by several strains of bifidobacteria. Mol Nutr Food Res 51, 1398–1405.
    1. Bode L, Kunz C, Muhly-Reinholz M, et al. (2004) Inhibition of monocyte, lymphocyte, and neutrophil adhesion to endothelial cells by human milk oligosaccharides. Thromb Haemost 92, 1402–1410.
    1. Bode L, Rudloff S, Kunz C, et al. (2004) Human milk oligosaccharides reduce platelet-neutrophil complex formation leading to a decrease in neutrophil beta 2 integrin expression. J Leukoc Biol 76, 820–826.
    1. Comstock SS, Wang M, Hester SN, et al. (2014) Select human milk oligosaccharides directly modulate peripheral blood mononuclear cells isolated from 10-d-old pigs. Br J Nutr 111, 819–828.
    1. Newburg DS, Tanritanir AC & Chakrabarti S (2016) Lactodifucotetraose, a human milk oligosaccharide, attenuates platelet function and inflammatory cytokine release. J Thromb Thrombolysis 42, 46–55.
    1. Coppa GV, Zampini L, Galeazzi T, et al. (2006) Human milk oligosaccharides inhibit the adhesion to Caco-2 cells of diarrheal pathogens: Escherichia coli, Vibrio cholerae, and Salmonella fyris . Pediatr Res 59, 377–382.
    1. Hester SN, Chen X, Li M, et al. (2013) Human milk oligosaccharides inhibit rotavirus infectivity in vitro and in acutely infected piglets. Br J Nutr 110, 1233–1242.
    1. Morrow AL, Ruiz-Palacios GM, Jiang X, et al. (2005) Human-milk glycans that inhibit pathogen binding protect breast-feeding infants against infectious diarrhea. J Nutr 135, 1304–1307.
    1. Newburg DS, Ruiz-Palacios GM & Morrow AL (2005) Human milk glycans protect infants against enteric pathogens. Annu Rev Nutr 25, 37–58.
    1. Newburg DS, Ruiz-Palacios GM, Altaye M, et al. (2004) Innate protection conferred by fucosylated oligosaccharides of human milk against diarrhea in breastfed infants. Glycobiology 14, 253–263.
    1. Turroni F, Peano C, Pass DA, et al. (2012) Diversity of bifidobacteria within the infant gut microbiota. PLOS ONE 7, e36957.
    1. Sela DA & Mills DA (2010) Nursing our microbiota: molecular linkages between bifidobacteria and milk oligosaccharides. Trends Microbiol 18, 298–307.
    1. Kerckhoffs AP, Samsom M, van der Rest ME, et al. (2009) Lower bifidobacteria counts in both duodenal mucosa-associated and fecal microbiota in irritable bowel syndrome patients. World J Gastroenterol 15, 2887–2892.
    1. Parkes GC, Rayment NB, Hudspith BN, et al. (2012) Distinct microbial populations exist in the mucosa-associated microbiota of sub-groups of irritable bowel syndrome. Neurogastroenterol Motil 24, 31–39.
    1. Schwiertz A, Taras D, Schafer K, et al. (2010) Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 18, 190–195.
    1. Le KA, Li Y, Xu X, et al. (2012) Alterations in fecal Lactobacillus and Bifidobacterium species in type 2 diabetic patients in Southern China population. Front Physiol 3, 496.
    1. Ouwehand AC, Salminen S & Isolauri E (2002) Probiotics: an overview of beneficial effects. Antonie van Leeuwenhoek 82, 279–289.
    1. Coulet M, Phothirath P, Allais L, et al. (2014) Pre-clinical safety evaluation of the synthetic human milk, nature-identical, oligosaccharide 2’-O-Fucosyllactose (2’FL). Regul Toxicol Pharmacol 68, 59–69.
    1. Coulet M, Phothirath P, Constable A, et al. (2013) Pre-clinical safety assessment of the synthetic human milk, nature-identical, oligosaccharide Lacto-N-neotetraose (LNnT). Food Chem Toxicol 62, 528–537.
    1. EFSA Panel on Dietetic Products Nutrition and Allergies (2015) Safety of 2’-O-fucosyllactose as a novel food ingredient pursuant to Regulation (EC) No 258/97. EFSA J 13, 4184.
    1. EFSA Panel on Dietetic Products Nutrition and Allergies (2015) Safety of lacto-N-neotetraose as a novel food ingredient pursuant to Regulation (EC) No 258/97. EFSA J 13, 4183.
    1. Dimenas E, Glise H, Hallerback B, et al. (1995) Well-being and gastrointestinal symptoms among patients referred to endoscopy owing to suspected duodenal ulcer. Scand J Gastroenterol 30, 1046–1052.
    1. Lewis SJ & Heaton KW (1997) Stool form scale as a useful guide to intestinal transit time. Scand J Gastroenterol 32, 920–924.
    1. Klindworth A, Pruesse E, Schweer T, et al. (2013) Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res 41, e1.
    1. Edgar RC (2013) UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods 10, 996–998.
    1. Edgar RC, Haas BJ, Clemente JC, et al. (2011) UCHIME improves sensitivity and speed of chimera detection. Bioinformatics 27, 2194–2200.
    1. Cole JR, Wang Q, Fish JA, et al. (2014) Ribosomal Database Project: data and tools for high throughput rRNA analysis. Nucleic Acids Res 42, D633–D642.
    1. Chen J, Bittinger K, Charlson ES, et al. (2012) Associating microbiome composition with environmental covariates using generalized UniFrac distances. Bioinformatics 28, 2106–2113.
    1. Casen C, Vebo HC, Sekelja M, et al. (2015) Deviations in human gut microbiota: a novel diagnostic test for determining dysbiosis in patients with IBS or IBD. Aliment Pharmacol Ther 42, 71–83.
    1. Jeffery IB, O’Toole PW, Ohman L, et al. (2012) An irritable bowel syndrome subtype defined by species-specific alterations in faecal microbiota. Gut 61, 997–1006.
    1. Walters WA, Xu Z & Knight R (2014) Meta-analyses of human gut microbes associated with obesity and IBD. FEBS Lett 588, 4223–4233.
    1. Ten Bruggencate SJ, Bovee-Oudenhoven IM, Lettink-Wissink ML, et al. (2006) Dietary fructooligosaccharides affect intestinal barrier function in healthy men. J Nutr 136, 70–74.
    1. Dimenas E, Carlsson G, Glise H, et al. (1996) Relevance of norm values as part of the documentation of quality of life instruments for use in upper gastrointestinal disease. Scand J Gastroenterol 221, 8–13.
    1. Costabile A, Kolida S, Klinder A, et al. (2010) A double-blind, placebo-controlled, cross-over study to establish the bifidogenic effect of a very-long-chain inulin extracted from globe artichoke (Cynara scolymus) in healthy human subjects. Br J Nutr 104, 1007–1017.
    1. Davis LM, Martinez I, Walter J, et al. (2010) A dose dependent impact of prebiotic galactooligosaccharides on the intestinal microbiota of healthy adults. Int J Food Microbiol 144, 285–292.
    1. Vulevic J, Drakoularakou A, Yaqoob P, et al. (2008) Modulation of the fecal microflora profile and immune function by a novel trans-galactooligosaccharide mixture (B-GOS) in healthy elderly volunteers. Am J Clin Nutr 88, 1438–1446.
    1. Davis LM, Martinez I, Walter J, et al. (2011) Barcoded pyrosequencing reveals that consumption of galactooligosaccharides results in a highly specific bifidogenic response in humans. PLoS ONE 6, e25200.
    1. Finegold SM, Li Z, Summanen PH, et al. (2014) Xylooligosaccharide increases bifidobacteria but not lactobacilli in human gut microbiota. Food Funct 5, 436–445.
    1. Duranti S, Turroni F, Lugli GA, et al. (2014) Genomic characterization and transcriptional studies of the starch-utilizing strain Bifidobacterium adolescentis 22L. Appl Environ Microbiol 80, 6080–6090.
    1. Duncan SH, Lobley GE, Holtrop G, et al. (2008) Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes (Lond) 32, 1720–1724.
    1. Mangin I, Leveque C, Magne F, et al. (2012) Long-term changes in human colonic Bifidobacterium populations induced by a 5-day oral amoxicillin-clavulanic acid treatment. PLOS ONE 7, e50257.
    1. Sokol H, Seksik P, Furet JP, et al. (2009) Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm Bowel Dis 15, 1183–1189.

Source: PubMed

3
Iratkozz fel