TY - JOUR AU - Mansell, Thomas J AB - Abstract The human gut is an ecosystem comprising trillions of microbes interacting with the host. The composition of the microbiota and their interactions play roles in different biological processes and in the development of human diseases. Close relationships between dietary modifications, microbiota composition and health status have been established. This review focuses on prebiotics, or compounds which selectively encourage the growth of beneficial bacteria, their mechanisms of action and benefits to human hosts. We also review advances in synthesis technology for human milk oligosaccharides, part of one of the most well-characterized prebiotic–probiotic relationships. Current and future research in this area points to greater use of prebiotics as tools to manipulate the microbial and metabolic diversity of the gut for the benefit of human health. Introduction The human gastrointestinal tract hosts an enormous population of microorganisms, bacteria, archaea, and eukarya [5], roughly 40 trillion cells [132, 133]. Although the gut microbiota has been explored for centuries, only recently have correlations been made between the population of the gut and human diseases [21], including Crohn’s disease [58], inflammatory bowel disease [77], cardiovascular disease [75, 139] and cancer [45, 144]. In addition to host genetics, the composition of the gut microbiome is influenced to a large extent by environmental factors including diet [14, 35, 54, 86, 96, 149, 167], especially in early life [175]. Given the connection between gut population dynamics and disease states, strategies to alter microbiome composition have been developed including fecal transplants [46, 68], introduction of consortia or single beneficial microorganisms (also known as probiotics) [30, 148] and purveyal of substrates to support growth of commensal microorganisms [9]. While it may not be surprising that the overall diet can affect the composition of the microbiome, certain compounds, known as prebiotics, have been found to uniquely influence the growth of particular beneficial microbes [53]. Prebiotic enrichment is not a new concept. The term “bifidus factor” was coined in the 1950s to account for the enrichment of beneficial Lactobacillus bifidus in infants [57]. Recently, a consensus definition was recently proposed by the International Scientific Association of Probiotics and Prebiotics (ISAPP) as “a substrate that is selectively utilized by host microorganisms conferring a health benefit” [50]. The mechanisms of the benefit of prebiotics are manifold, but are essentially either direct (e.g., carbohydrate structures similar to host glycans can block adhesion to host cells [78]) or downstream, encouraging the proliferation of bacteria whose metabolites directly affect the gut environment or host gene expression (e.g., the effect of short-chain fatty acids on both gut pH and modulation of immune function [156]). In this review, we describe the most common prebiotics, their role in manipulating the microbiome and their downstream effects on human health, as well as strategies for chemical and biotechnological synthesis of these important compounds. Carbohydrate prebiotics The accessibility of a carbon source is a major contributor to a microbe’s ability to produce biomass [108, 164]. It follows, then, that microbial populations might change in response to changing carbon sources based on each species’ ability to use those sources. Species from the two major bacterial phyla, Bacteroidetes and Firmicutes, have been identified that break down polysaccharides and complex oligosaccharides [126, 127] including Bacteroides thetaiotaomicron [90], Bacteroides cellulolyticus [91] and Roseburia intestinalis [39]. Other mechanisms besides carbohydrate utilization, like cross-feeding [150], allow homogeneous bacterial populations to give rise to more diverse populations where metabolites from one strain provide a niche for the other [7, 147]. Human milk oligosaccharides (HMOs) Evidence from as early as the 1930s showed lower risk of morbidity or mortality due to enteric disease in breast-fed infants [55]. In addition to lipids and sugars such as lactose that directly provide nutrition to infants, human breast milk is rich in biologically active compounds that exhibit antimicrobial and immunomodulatory activities [3]. It also contains human milk oligosaccharides (HMOs), carbohydrates which are not digestible by the infant but instead influence the gut microbiome, which will be the main focus of this review. The symbiotic relationship between human milk and beneficial bacteria is the result of 200 million years of co-evolution [174] and represents nature’s strategy for establishing a healthy human infant microbiome. With the growing evidence of the beneficial functions that HMOs offer to infants, various methods of synthesis have been devised. Before the advent of current synthesis technologies, alternative complex oligosaccharides such as fructans and galactans which can be sourced more readily and economically were derived from relatively abundant resources. The structures of prebiotics discussed in this review have been illustrated in Fig. 1. Beneficial outcomes from consumption of prebiotics are listed in Table 1. Fig. 1 Open in new tabDownload slide Structural diversity of carbohydrate prebiotics. a Fructans: inulin is composed of fructose units linked to each other by β-(2,1) bonds, found in many plants. b Galactans: GOS are composed of β-(1,6)-linked galactose residues with a terminal glucose or galactose at the reducing end that are produced by enzymatic transgalactosylation of glucose, galactose, or lactose. c Xylooligosaccharides, XOS are produced from xylan-rich lignocellulosic materials, consisting of xylose residues linked via β-(1,4) linkages. Side groups such as α‐d‐glucopyranosyl uronic acid, acetyl groups, or arabinofuranosyl residues leads to branched XOS. d Mannan-oligosaccharides, MOS are composed of β-(1,4)-linked mannose residues, derived from many plants and yeast cell walls. e HMO blueprint and simple HMOs, all composed of a lactose core at the reducing end. Lactose can be fucosylated or sialylated terminally or elongated by lacto-N-biose (β-(1,3)-linked, type I) or N-acetyllactosamine (β-(1,4)-linked, type II) structures. The structures pictured are (i) basic structure of HMOs, (ii) 2′-fucosyllactose, (iii) 3-fucosyllactose, (iv) 6′-sialyllactose, (v) lacto-N-tetraose, and (vi) lacto-N-neotetraose Prebiotics and their beneficial outcomes Prebiotic oligosaccharides . Benefit to host . Proposed responsible microorganism . References . HMO Anti-inflammatory effect, regulation of permeability of host cells, protection from diarrhea, necrotizing enterocolitis and respiratory tract infections, diabetes, prevent binding of pathogens like rotavirus and Campylobacter jejuni to host cells Bifidobacterium longum subsp. infantis, Bacteroides [12, 27, 99, 162, 165, 170, 171] FOS Anti-inflammatory effect, improved IBS symptoms, lower blood glucose level, anti-cancer, diarrhea treatment, reduces atopic dermatitis Bifidobacteria, Faecalibacterium prausnitzii, Bacteroidetes [83, 95, 105, 112, 171] Inulin Improved bowel function, increased bifidobacteria population, lower serum lipopolysaccharide level, immunomodulatory action, increased mineral absorption, cardiovascular effects, anti-cancer Bifidobacteria, Bacteroidetes [33, 36, 44, 73, 114, 146, 154, 157] GOS Immunomodulatory action, pathogen adherence inhibition, improved IBS symptoms, reduces atopic dermatitis Bifidobacteria, lactobacilli [44, 95, 116, 135] MOS Lower IBS symptoms, increased expression of proinflammatory mediators, lower proinflammatory cytokines, prevent binding of pathogens to host cells Lactobacilli [43, 102] XOS Immunomodulatory action, increased plasma HDL concentration, anti-inflammatory effect, reduction in triglycerides, lower dose requirement for clinical efficacy, anti-cancer Bifidobacteria, lactobacilli [59, 66, 117, 128, 163] Prebiotic oligosaccharides . Benefit to host . Proposed responsible microorganism . References . HMO Anti-inflammatory effect, regulation of permeability of host cells, protection from diarrhea, necrotizing enterocolitis and respiratory tract infections, diabetes, prevent binding of pathogens like rotavirus and Campylobacter jejuni to host cells Bifidobacterium longum subsp. infantis, Bacteroides [12, 27, 99, 162, 165, 170, 171] FOS Anti-inflammatory effect, improved IBS symptoms, lower blood glucose level, anti-cancer, diarrhea treatment, reduces atopic dermatitis Bifidobacteria, Faecalibacterium prausnitzii, Bacteroidetes [83, 95, 105, 112, 171] Inulin Improved bowel function, increased bifidobacteria population, lower serum lipopolysaccharide level, immunomodulatory action, increased mineral absorption, cardiovascular effects, anti-cancer Bifidobacteria, Bacteroidetes [33, 36, 44, 73, 114, 146, 154, 157] GOS Immunomodulatory action, pathogen adherence inhibition, improved IBS symptoms, reduces atopic dermatitis Bifidobacteria, lactobacilli [44, 95, 116, 135] MOS Lower IBS symptoms, increased expression of proinflammatory mediators, lower proinflammatory cytokines, prevent binding of pathogens to host cells Lactobacilli [43, 102] XOS Immunomodulatory action, increased plasma HDL concentration, anti-inflammatory effect, reduction in triglycerides, lower dose requirement for clinical efficacy, anti-cancer Bifidobacteria, lactobacilli [59, 66, 117, 128, 163] Open in new tab Prebiotics and their beneficial outcomes Prebiotic oligosaccharides . Benefit to host . Proposed responsible microorganism . References . HMO Anti-inflammatory effect, regulation of permeability of host cells, protection from diarrhea, necrotizing enterocolitis and respiratory tract infections, diabetes, prevent binding of pathogens like rotavirus and Campylobacter jejuni to host cells Bifidobacterium longum subsp. infantis, Bacteroides [12, 27, 99, 162, 165, 170, 171] FOS Anti-inflammatory effect, improved IBS symptoms, lower blood glucose level, anti-cancer, diarrhea treatment, reduces atopic dermatitis Bifidobacteria, Faecalibacterium prausnitzii, Bacteroidetes [83, 95, 105, 112, 171] Inulin Improved bowel function, increased bifidobacteria population, lower serum lipopolysaccharide level, immunomodulatory action, increased mineral absorption, cardiovascular effects, anti-cancer Bifidobacteria, Bacteroidetes [33, 36, 44, 73, 114, 146, 154, 157] GOS Immunomodulatory action, pathogen adherence inhibition, improved IBS symptoms, reduces atopic dermatitis Bifidobacteria, lactobacilli [44, 95, 116, 135] MOS Lower IBS symptoms, increased expression of proinflammatory mediators, lower proinflammatory cytokines, prevent binding of pathogens to host cells Lactobacilli [43, 102] XOS Immunomodulatory action, increased plasma HDL concentration, anti-inflammatory effect, reduction in triglycerides, lower dose requirement for clinical efficacy, anti-cancer Bifidobacteria, lactobacilli [59, 66, 117, 128, 163] Prebiotic oligosaccharides . Benefit to host . Proposed responsible microorganism . References . HMO Anti-inflammatory effect, regulation of permeability of host cells, protection from diarrhea, necrotizing enterocolitis and respiratory tract infections, diabetes, prevent binding of pathogens like rotavirus and Campylobacter jejuni to host cells Bifidobacterium longum subsp. infantis, Bacteroides [12, 27, 99, 162, 165, 170, 171] FOS Anti-inflammatory effect, improved IBS symptoms, lower blood glucose level, anti-cancer, diarrhea treatment, reduces atopic dermatitis Bifidobacteria, Faecalibacterium prausnitzii, Bacteroidetes [83, 95, 105, 112, 171] Inulin Improved bowel function, increased bifidobacteria population, lower serum lipopolysaccharide level, immunomodulatory action, increased mineral absorption, cardiovascular effects, anti-cancer Bifidobacteria, Bacteroidetes [33, 36, 44, 73, 114, 146, 154, 157] GOS Immunomodulatory action, pathogen adherence inhibition, improved IBS symptoms, reduces atopic dermatitis Bifidobacteria, lactobacilli [44, 95, 116, 135] MOS Lower IBS symptoms, increased expression of proinflammatory mediators, lower proinflammatory cytokines, prevent binding of pathogens to host cells Lactobacilli [43, 102] XOS Immunomodulatory action, increased plasma HDL concentration, anti-inflammatory effect, reduction in triglycerides, lower dose requirement for clinical efficacy, anti-cancer Bifidobacteria, lactobacilli [59, 66, 117, 128, 163] Open in new tab HMOs are the third most abundant component in human milk by dry weight, after lactose and lipids, and constitute a family of structurally diverse unconjugated glycans with a core lactose residue at the reducing end, a backbone of alternating N-acetyl glucosamine and galactose, and/or other moieties including fucose (α-1,2, α-1,3, or α-1,4 linked) or sialic acid (α-2,3 or α-2,6 linked) [12] (Fig. 1e). Human breast milk contains about 5–15 g/L of free oligosaccharides [12, 78]. Genome sequencing of bifidobacteria such as Bifidobacterium longum subsp. infantis and other isolates revealed gene clusters encoding glycosyl hydrolases and transporters [174] that allow them to consume HMOs as a primary carbon source [152]. B. longum subsp. infantis, B. longum subsp. longum, B. breve and B. pseudocatenulatum, internalize HMOs via ATP-binding cassette (ABC) transporters and hydrolyze these oligosaccharides using intracellular glycosyl hydrolases [88]. This strategy protects the infant by giving an ecological advantage to these bifidobacteria, preventing potential pathogens from colonizing the gut [23]. Other species including B. bifidum and Bacteroides species secrete extracellular hydrolases that break down oligosaccharides before the sugars are internalized. Sugars are fermented to acetate, which can be converted to butyrate by obligate anaerobes [93] and is found in higher concentrations in infants with healthy guts [153]. There are a few reported mechanisms by which the proliferation of bifidobacteria helps in protecting infants from diseases like diarrhea and necrotizing enterocolitis. Their unique mechanism of metabolizing HMOs is also accompanied by improved binding to the intestinal mucosal layer [27]. A recent metagenomic study, The Environmental Determinants of Diabetes in the Young (TEDDY), showed an overrepresentation of bifidobacteria and the presence of HMO utilization genes in breast-fed infants [155]. These data also supported the protective effects of short-chain fatty acids (SCFAs) in early-onset Type 1 diabetes (T1D), as was seen in mice [89]. A major challenge in developing and utilizing HMOs for therapeutic purposes is the limited supply of human milk and difficulty in isolation from other milk sources [13]. Thus, chemical and biotechnological synthesis of HMOs has become a field of great interest, as outlined below. Chemical synthesis The assembly of complex carbohydrates is challenging due to the combined demands of elaborate procedures for glycosyl donor and acceptor preparation and the requirements of regio- and stereoselectivity in glycosylation. They involve the preparation of selectively protected monosaccharide units, one with a strategically positioned free hydroxyl group (a nucleophilic acceptor) and one bearing a labile leaving group at the anomeric carbon that acts as a glycosyl donor in the ensuing glycosylation reaction. The remaining hydroxyl groups need to be protected (1) to tune the overall electronic properties of the donors and acceptors so that the donor–acceptor pair can be matched and (2) for further deprotection and glycosylation or functional-group modifications [161]. Chemoselective, orthogonal and iterative glycosylation strategies, which exploit differential reactivities of anomeric leaving groups, allow several selected glycosyl donors to react in a specific order resulting in a single oligosaccharide product [158]. Plante et al. devised a solid-phase synthesis method that allows automated synthesis by framing the repetitive glycosylation and deprotection into a coupling cycle [111]. By exposing the nucleophilic acceptor hydroxyl group on the solid support to the glycosylating agent in solution, support-bound oligosaccharides can be produced. Solid-phase synthesis eases purification but the complexity of protection–deprotection remains the same and is accompanied by steric hindrance [131]. One-pot synthesis methods have been developed that involve multistep reactions performed in a single setup, thus eliminating the need to isolate intermediates and shortening the experimental time frame. Wang et al. developed a highly regioselective, combinatorial, orthogonal and one-pot protection method that can aid in glycan synthesis using a single catalyst, which gives selectively one isomer at each step [161]. The complex and dynamic nature of the glycans makes their synthesis via traditional chemical methods challenging [109] and the numerous selective protection and deprotection steps makes it tedious. This is exacerbated by the formation of a mixture of anomers which would involve an additional separation step [34]. Enzymatic/chemoenzymatic synthesis Biocatalytic approaches avoid some of the challenges faced in chemical synthesis. The use of enzymes as catalysts means regioselectivity and stereospecificity are better controlled and there are fewer steps involved, making the process cost effective and less time consuming [34, 109]. Two different enzymatic approaches can be taken: (1) glycosyltransferase-catalyzed synthesis, which requires sugar nucleotides or sugar-1-phosphates as donors, and (2) glycosidase-catalyzed synthesis. These enzymatic methods for synthesis involve either isolation or recombinant production of the enzymes [62]. The substrates for the glycosyltransferases are nucleotide sugars (e.g., UDP-glucose) and require an acceptor for the transfer of the glycosyl residue. The glycosidase-catalyzed pathway involves the transfer of an enzyme-bound glycosyl residue to an acceptor (water, in the absence of an organic compound). Enzymatic solid-phase synthesis combines advantages of enzymatic synthesis with the ease of purification that comes with solid-phase methods [130]. One-pot synthesis methods require protection of groups only during building block synthesis which is in the order of increasing reactivity [173]. To build more complex HMO structures of varying lengths, linkages or branching, a chemoenzymatic strategy is more desirable, in which a chemically synthesized acceptor is further modified via enzymatic steps. A one-pot multienzyme approach was used to synthesize lacto-N-neotetraose in two steps via azido alkyl-lactoside using glycosyltransferases and this strategy was reiterated to synthesize sialyl- and fucosyl-derivatives [25]. Another approach is to synthesize an acceptor with a cleavable anomeric linker to allow for purification of reaction intermediates [115]. After the enzymatic extension is complete, the linker can be hydrolyzed to give oligosaccharides with a free reducing end. A solid-phase strategy was used to synthesize sialylated HMOs, which is generally challenging due to steric hindrance and reduced reactivity of the position. Fair et al. overcame this limitation using automated solid-phase assembly of the glycan backbone followed by enzymatic sialylation with an α-(2,3)-sialyltransferase, and obtained sialylated HMOs with high regio- and stereoselectivity [41]. Microbial synthesis New strategies for oligosaccharide production have been developed using metabolically engineered microbes. This approach has brought down the cost of synthesis of glycans significantly since they do not require the overproduction and isolation of the glycosyltransferases and expensive precursors and have high production titers [22]. Lactose has commonly been used as the sugar acceptor because of its abundance and its presence in all HMOs. Figure 2 exemplifies engineered metabolic pathways for synthesis of common HMOs. Fig. 2 Open in new tabDownload slide Engineered metabolic pathways for the production of fucosyllactose, sialyllactose and lacto-N-triose. Lactose, l-fucose and sialic acid are transported into the cell by specific permeases LacY, FucP and NanT. Glycerol serves as the carbon/energy source. Lactose serves as the substrate acceptor and in many cases the lacZ is deleted to prevent lactose from being metabolized. Molecules and enzymes have been abbreviated as: Glc d-glucose, Gal d-galactose, l-Fuc l-fucose, Neu5Ac sialic acid, Lac lactose, GDP-4k-6d-Man GDP-4-keto-6-deoxymannose, GDP-Man GDP-d-mannose, ManNAc N-acetylmannosamine, UDP-GlcNAc uridine diphosphate N-acetylglucosamine, 2′-FL 2′-fucosyllactose, 6′-SL 6′-sialyllactose, LNT2 lacto-N-triose II, LacY lactose permease, LacZ β-galactosidase, FucP fucose permease, WcaG GDP-fucose synthase, GlpF glycerol MIP channel, ManB phosphomannomutase, ManC mannose-1-phosphate guanylyltransferase, Gmd GDP-mannose 4,6-dehydratase, FutC α-1,2-fucosyltransferase, ST6Gal β-galactoside α-2,6-sialyltransferase, LgtA N-acetylglucosaminyltransferase, GlmS glutamine-fructose-6-phosphate aminotransferase, GlmM phosphoglucosamine mutase, GlmU N-acetylglucosamine 1-phosphate uridyltransferase, NeuC UDP-N-acetylglucosamine 2-epimerase, NeuB N-acetyl neuraminic acid synthetase, NeuA N-acylneuraminate cytidylyltransferase, NanT sialic acid transporter Several workhorse microbes have been used as hosts, mainly dependent on the commercial availability of the expression vector and the ease of manipulation. Escherichia coli has been the host of choice but the use of species that are naturally evolved to rapidly regenerate sugar nucleotides reduces a number of steps taken to genetically modify certain pathways. Saccharomyces cerevisiae, carrying plasmids expressing the human β-1,4-galactosyltransferase gene, was used to successfully produce LacNAc from GlcNAc and UDP-Gal [61]. Ruffing et al. successfully engineered Agrobacterium spp. to synthesize galactose-core oligosaccharides by exploiting the UDP-galactose regeneration system [124]. E. coli and Corynebacterium ammoniagenes strains have been used to synthesize N-acetyllactosamine, 3′-sialyllactose and Lewisx blood group antigen structures. E. coli strains overexpressing CMP-NeuAc synthetase and CTP synthetase genes were used to produce cytidine 5′ monophospho-N-acetylneuraminic acid (CMP-NeuAc). The production of this nucleotide sugar donor coupled with the overexpression of the α-(2,3)-sialyltransferase gene of Neisseria gonorrhoeae and coupling with C. ammoniagenes that forms UTP from orotic acid resulted in high titer production of 3′-sialyllactose [40]. 2′-Fucosyllactose (2′-FL) is the most abundant oligosaccharide in human milk [24] and has been shown to offer protection against pathogens including Campylobacter jejuni [125]. Scientists have developed different biosynthetic pathways in E. coli to produce 2′-FL [6, 81]. Baumgärtner et al. chromosomally integrated recombinant α-(1,2)-fucosyltransferase from Helicobacter pylori and genes from the GDP-l-fucose de novo pathway, which transforms mannose-6-phosphate to GDP-l-fucose, into E. coli [6]. With the addition of the salvage pathway, the intracellular GDP-l-fucose concentration was controlled via FucP using extracellularly added l-fucose. This resulted in a tenfold greater yield and a final 2′-FL concentration of 20 g/L. Earlier, another group produced 2′-FL using two expression plasmids where glycosylation was achieved by the Neisseria meningitidis lgtAB genes [37]. Recently, Yu et al. demonstrated production of 2′-FL in engineered S. cerevisiae via the salvage pathway to produce 2′-FL using l-fucose and lactose [169]. Although 3-FL can be synthesized by a similar approach using α-1,3-fucosyltransferase, its poor expression results in very low titers. Yu et al. engineered it by removing the membrane-binding region and elongating the heptad repeats to achieve a 10–20-fold increase in 3-FL titer [168]. Priem et al. expressed the β-1,3 N-acetylglucosaminyltransferase lgtA gene and the β-1,4 galactosyltransferase lgtB gene of N. meningitidis in E. coli and by sequential reactions produced lacto-N-triose (GlcNAcβ-1,3Galβ-1,4Glc) and lacto-N-neotetraose (Galβ-1,4GlcNAcβ-1,3Galβ-1,4Glc). They also produced 3′-sialyllactose by inactivating the nanA gene which encodes an aldolase and by exogenous addition of sialic acid [113]. More recently, Guo et al. demonstrated synthesis of 6′-sialyllactose, using a unique trans-sialidase to transfer a sialyl residue to lactose by transglycosylation, with high transglycosylation activity and strict regioselectivity [56]. In glycosidase-catalyzed synthesis, there is typically competition between transglycosylation and hydrolysis, which can be improved by employing higher acceptor/donor ratios that increase the probability of nucleophilic attack by the acceptor on the enzyme to form a glycosyl–enzyme intermediate. Zeuner et al. engineered an α-1,3/4-l-fucosidase to completely remove its hydrolytic activity and improved the transfucosylation activity to achieve threefold higher yields in their fucosylated product [172]. Excellent reviews covering the recent developments in synthesizing galactose-containing oligosaccharides [26] and regarding commercialization and regulatory framework [69] can be found at the aforementioned references. Fructans: inulin and fructooligosaccharide (FOS) Inulin, a polysaccharide abundant in commercial plant sources such as chicory and agave, is a polydisperse β-(2,1) fructan with the fructose units linked to each other by β-(2,1) bonds, with a degree of polymerization (DP) ranging from 2 to 60 [110] (Fig. 1a). Each fructose chain is capped by a terminal α-(1,2)-linked glucose molecule. Fructooligosaccharide (FOS) is composed of shorter chain oligomers, with a DP between 2 and 10, which can be produced by hydrolysis of inulin using endoinulinase. The prebiotic property of inulin is derived from its β-(2,1) linkages that remain indigestible. Their selective fermentation by bifidobacteria that produce an intracellular inulinase to hydrolyse the β-(2,1) fructose linkages, allows for the modulation of the composition of the gut microbiota [119]. Fructans are widely used as additives in food for their health boosting properties, especially in conjunction with probiotics, for a synergistic “synbiotic” effect [100]. Their physiological effects are dictated by their chain length, as shown by a study where FOS was fermented more rapidly and produced more butyrate compared to inulin [138]. In a simulated microbial environment seeded with fecal samples, supplementation of inulin resulted in increased Bacterioides uniformis or Bacterioides caccae [29]. Galactans: galactooligosaccharides (GOS) GOS are composed of galactose residues with a terminal glucose or galactose at the reducing end (Fig. 1b). Commercially, GOS are produced by enzymatic transgalactosylation of glucose, galactose, or lactose attached via β-(1, 2, 3, 4, or 6) linkages, with a DP of 2–8. GOS utilization is dependent on the linkage specificity of the β-galactosidases. Studies have shown a dose-dependent increase in fecal bifidobacteria in infants and increased SCFA levels upon fermentation [8, 42, 134]. GOS has been widely used in infant formula and other confectioneries and beverages [52, 151] and together with commercially available probiotic bifidobacteria and lactobacilli [143]. Xylooligosaccharides (XOS) XOS are produced from xylan-rich lignocellulosic materials, consisting of xylose residues linked via β-(1,4) linkages with a DP between 2 and 10 (Fig. 1c). Depending on the source, XOS are usually accompanied by other side groups such as α‐d‐glucopyranosyl uronic acid, acetyl groups, or arabinofuranosyl residues, whose presence results in branched structures [1]. Studies showed the selective ability of certain probiotic strains, Bifidobacterium adolescentis and Lactobacillus plantarum to utilize XOS owing to the presence of xylanolytic enzyme systems [76, 103]. Compared to other prebiotic substrates, including FOS, GOS and MOS, XOS produced the largest amount of SCFA upon degradation by fecal microbiota of pigs [136]. There is direct experimental evidence for the prebiotic efficacy of XOS in humans, including increased proportion of bifidobacteria [65] and SCFA levels [74]. Mannan-oligosaccharides (MOS) MOS are oligomers composed of mannose residues, linked together by β-1,4 glycosidic bonds, with a DP in the range 2–10 (Fig. 1d). Derived from yeast cell walls, MOS has been widely used to improve growth and health performance of livestock and aquaculture [94]. Supplementation of MOS to chicken feed reduced Clostridium perfringens and E. coli, and increased the relative population of Lactobacillus spp. [71]. Bacterial genera such as Bacteroides and Bacillus produce mannanases that can cleave β-1,4 mannopyranoside in mannan products. MOS has been shown to bind to mannose-specific lectins expressed in bacteria with fimbriae and hence, reduces bacterial adhesion to mannose-containing glycoprotein receptors on intestinal epithelial cells [102]. Mechanisms of action of prebiotics While prebiotics can manipulate the bacterial population, they can also benefit the host via gut environment through direct interactions with potential pathogens and/or the immune system. Figure 3 summarizes the proposed mechanisms of prebiotic action. Fig. 3 Open in new tabDownload slide Schematic diagram illustrating mechanisms by which prebiotics influence the gut microbiota. The effect of prebiotics is shown below the dashed line. (1) Selective colonization of probiotic bacteria (bifidobacteria and lactobacilli) by competition with pathogenic strains for prebiotics. (2) Production of short-chain fatty acids (SCFA), acetate, propionate and butyrate, resulting in reduction of pH. (3) Exclusion of pathogen binding to epithelial cells. (4) Enhanced intestinal barrier function by increased mucus. (5) Modulation of immune response Prebiotics are capable of interacting with the carbohydrate receptors on immune cells. Receptors on phagocytes and natural killer (NK) cells can be activated by binding of β-glucans which triggers neutrophil phagocytosis [122]. Glucans are a heterogeneous group of glucose polymers, composed of a backbone of β-(1,3)-linked β-d-glucopyranosyl units with β-(1,6)-linked side chains. Dectin-1, a NK-cell-receptor-like C-type lectin [16], was identified as a macrophage receptor for β-glucans, eliciting antitumor and antimicrobial activity [17]. Inulin-type fructans are also receptors for gut dendritic cells (DCs), via Toll-like receptors, C-type lectin receptors, and galectins, that induce anti-inflammatory cytokines [157]. The activity of HMOs against bacterial and viral infections can be explained by their absorption from the GI tract into the system and serving as receptors for bacterial or viral adhesion molecules [101]. Another mechanism by which prebiotics promote health is by promoting the production of metabolites, e.g., folate, indoles, secondary bile acids, trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFAs), through fermentation of the prebiotic compounds [21]. The gut microbiota are endowed with plethora of glycoside hydrolases and polysaccharide lyases that are capable of breaking down the indigestible carbohydrates that traverse intact all the way to the distal gut [97]. It is the fermentation of these carbohydrates that produces SCFAs. These SCFAs can be transported across the intestine and into the systemic compartments, affecting the immune system. The SCFAs, acetate, propionate and butyrate in particular, showcase antimicrobial activity, lead to the lowering of pH in the gut, and heavily impacting the host metabolism and immunity, including from food and self-antigens [10, 49, 53, 140]. SCFAs also inhibit histone-deacetylase (HDAC) activity that affects gene transcription [137]. SCFAs can also bind to G-coupled protein receptors on leukocytes. Butyrate serves not only as a carbon source for colonic cells and hence helps to maintain a healthy gut barrier but also aids in maintaining the anaerobic conditions inside the gut [19, 20]. One study reported downregulation of inflammatory cytokines upon treatment with propionate but no effect with XOS indicating the indirect SCFA-mediated anti-inflammatory effect [59]. Another outcome of prebiotic treatment is the selective increase/decrease in specific intestinal bacteria that in turn, can induce health benefits. Early studies showed the growth advantage of bifidobacteria over E. coli and Clostridium spp. that cannot utilize XOS, leading to the predominance of bifidobacterial population [103]. Treatment with FOS was also associated with higher intestinal bifidobacteria counts in another study [66]. These bifidobacteria can digest XOS and FOS to produce lactate and SCFAs. One study showed that the gut microbial composition is a key player in energy homeostasis and hence dysbiosis can contribute towards obesity [84]. It was hypothesized that the Firmicutes-enriched cecal microbiota (relative to Bacteroidetes-enriched microbiota) promoted obesity by limiting energy uptake/storage. Role of prebiotics in human health The gut microbiota has been considered as functioning like an organ, constituting multitudes of cells and performing various functions in the human body. Prebiotics have a profound effect in modulating the gut microbiota, and hence a concomitant effect on human health. With increasing number of pre-clinical studies underway supporting the benefits of prebiotics, they are becoming an attractive therapeutic target for the prevention and treatment of human diseases. Immunomodulation Prebiotics may affect the immune system, either directly or indirectly via metabolites produced through fermentation or probiotic-mediated modulation of gene expression. β-Glucans interact with cell surface receptors to induce macrophages and NK cells which can also lead to inhibition of tumor growth in its promotion stage [2]. HMOs have shown to directly affect the development of the neonatal immune system. They stimulate production of cytokines and proliferation of immune cellular population [32]. In elderly adults, GOS was shown to increase phagocytosis, NK cell activity and production of anti-inflammatory cytokines and to decrease the production of proinflammatory cytokines [159]. HMOs containing sialic acid moieties were shown to inhibit the binding of rotavirus to host cells, directly affecting immune activity in rotavirus-infected neonates [31]. Several studies showed that mice exposed to inulin enriched with oligofructose, raffinose or FOS had enhanced production of cytokines [64, 98, 121]. HMOs also exhibit anti-adhesive properties against pathogenic bacteria and toxins, preventing attachment to epithelial cells [87]. Irritable bowel syndrome (IBS) Irritable bowel syndrome (IBS) is a common functional disorder characterized by abdominal discomfort and pain, bloating and an alteration in bowel habit [142]. Studies have shown that soluble fibers in partially hydrolyzed guar gum can alleviate abdominal pain and improve bowel habits [60]. Another study showed a decrease in digestive discomfort in patients given FOS [105]. GOS was also shown to significantly improve IBS symptoms [135]. Inflammatory bowel disease (IBD) The two main types of inflammatory bowel disease are ulcerative colitis and Crohn’s disease. Crohn’s disease usually affects the intestine and causes inflammation that affects the body’s ability to digest food, absorb nutrients and eliminate waste. Ulcerative colitis mainly affects the lining of the large intestine and rectum and chronic colitis can lead to colon cancer. IBD is associated with increased levels of the proinflammatory cytokines interleukin-1 (IL-1), IL-6, IL-8, and tumor necrosis factor α (TNF-α) [120]. Treatment with inulin and oligofructose was shown to reduce inflammation in rat models with colitis [63]. Reduced severity of DSS-induced colitis was also observed by another study on treatment with a combination of inulin/oligofructose and B. infantis [104]. Consumption of MOS also attenuated colitis symptoms in mice with DSS-induced colitis [43], presumably by altering the microbiota. Mental health and neurological disease The gut-brain axis has emerged as an exciting concept in microbial endocrinology. This crosstalk between the gut and brain is mediated by the vagus nerve [15]. There is evidence that shows the ability of the gut microbiota to communicate with the brain and modulate behavior by activating neural pathways and central nervous system (CNS) signaling systems [47]. Treatment with XOS was shown to reduce effects from pro-inflammatory cytokines in the brain [48]. Long-term consumption of XOS diminished activation of microglia and restored cognitive function in obese rats [28]. Chronic supplementation of FOS/GOS exhibited both antidepressant and anxiolytic effects and also reduced stress-induced corticosterone release by modifying gene expression in the hippocampus and hypothalamus in mice [18]. Similarly, significant decrease in anxiety was observed in animals treated with MOS [43]. Cardiovascular disease Cardiovascular diseases are associated with high levels of cholesterol and/or triglycerides [72]. Studies in rats with prebiotic fiber demonstrated a significant decrease in serum cholesterol levels [107]. Consumption of XOS by rats also showed a decline in the level of triglycerides [66]. Other studies have reported the reduction in cholesterol on consumption of FOS in diabetic subjects [166] and improvement in lipid profiles with inulin [118]. Propionate, produced by prebiotic fermentation, also lowers serum cholesterol levels and inhibits expression of the genes associated with intestinal cholesterol biosynthesis [4]. Cancer Numerous studies have identified prebiotics exhibiting anti-cancer properties [11, 51, 141]. Non-digestible polyphenols have the ability to alter the gut microbiota population, favoring strains capable of breaking down polyphenols, e.g., Bifidobacterium spp. and Lactobacillus spp., and producing simple phenolic metabolites. These metabolites can modulate enzyme and gene expression, e.g., phenolic residues like coumaric and caffeic acid produced by degradation of polyphenolics suppressed the histone-deacetylase activity in human colon cancer cells [160]. Butyrate was also reported to suppress cancer by inducing apoptosis in cancer cells and limiting histone-deacetylase activity [106]. Microbial metabolites of quercetin and caffeic acid lowered the DNA damage in human adenoma cells by regulating enzymes involved in detoxification and inflammation [92]. XOS and FOS were shown to inhibit colonic aberrant crypt foci in rats by lowering cecal pH and serum triglyceride levels [123]. It has also been proposed that absorption of micronutrients helped alleviate cancer risks. Consumption of inulin, FOS, and GOS was shown to increase the solubility of minerals and hence enhance their absorption [129]. Conclusion Significant progress has been made over the past few decades in recognizing the importance of gut microbiota to overall health. Going forward, armed with new tools and innovative approaches, opportunities still exist to allow researchers to further elucidate the mechanisms involved. Realization of the properties and benefits of prebiotics offer a new dimension for the development of functional foods and new treatment strategies for therapeutic targeting of the gut microbiota. However, further investigations are needed to elucidate the mechanisms involved in the treatment of diseases and confirmed with experimental models and clinical trials before prebiotics can be used as a potential therapeutic. In this review, we focused only on carbohydrate-based prebiotics but more recent studies have shown the prebiotic effects of other molecules including phytochemicals. Phytochemicals are bioactive non-nutrient plant compounds shown to exhibit selective stimulation of the gut microbiota [145]. Some polyphenols, for example, are not absorbed and reach the colon where they undergo metabolic transformations through the gut microbiota: fission of cyclic rings, deglycosylation, ester hydrolysis, and these metabolites are absorbed by the body [82]. These phytochemicals have been reported to lower risks of major chronic diseases, including hypertension, diabetes, neurodegenerative diseases and cancer [85]. Further studies need to be carried out to better understand their mechanisms and elucidate their benefits to host health. Engineering strategies to develop tools for the enrichment of the gut microbiota are evolving and custom-designed prebiotics is another area that needs further exploration, to better understand their effects on immunomodulation and interactions with pathogenic microbes. With current large-scale synthesis routes, elaborate biological investigations can be carried out. In addition, there is still a need to understand the effects all components of the microbiota including the spectrum of fungi, archaea, bacteriophages and viruses. The approach of coupling prebiotics and probiotics (as synbiotics), still has untapped capabilities toward influencing and improving human health. Another exciting development in this area is the creation of engineered bacteria capable of producing metabolites, peptides, or proteins that benefit the host, i.e., live biotherapeutics [38, 67, 79, 80]. As development of these engineered probiotic organisms continues, it follows that strategies for controlling their population dynamics will be essential for optimizing their efficacy. Two recent studies have approached this topic by creating orthogonal niches for bacteria via diet-based intervention [70, 133]. Essentially, prebiotics that can only be consumed by the species of interest can uniquely and reversibly upregulate that species in the gut. As we understand more fully the biology of prebiotics and their relationship to the dynamics of the gut microbiome and metabolome and workings of the human body, we will be better positioned to take more sophisticated approaches to shaping the microbiota for better health. Acknowledgements This work was supported by the Iowa State University Startup Funds. F.E. was supported in part by the Manley Hoppe Professorship and T.J.M. by the Karen and Denny Vaughn Faculty Fellowship. References 1. Aachary AA , Prapulla SG Xylooligosaccharides (XOS) as an emerging prebiotic: microbial synthesis, utilization, structural characterization, bioactive properties, and applications Compr Rev Food Sci Food Saf 2011 10 2 16 Google Scholar OpenURL Placeholder Text WorldCat 2. Akramiene D , Kondrotas A, Didziapetriene J, Kevelaitis E Effects of beta-glucans on the immune system Medicina 2007 43 597 606 Google Scholar OpenURL Placeholder Text WorldCat 3. Andreas NJ , Kampmann B, Mehring Le-Doare K Human breast milk: a review on its composition and bioactivity Early Hum Dev 2015 91 629 635 Google Scholar OpenURL Placeholder Text WorldCat 4. Arora T , Sharma R, Frost G Propionate. Anti-obesity and satiety enhancing factor? Appetite 2011 56 511 515 Google Scholar OpenURL Placeholder Text WorldCat 5. Bäckhed F , Ley RE, Sonnenburg JL, Peterson DA, Gordon JI Host-bacterial mutualism in the human intestine Science 2005 307 1915 1920 Google Scholar OpenURL Placeholder Text WorldCat 6. Baumgärtner F , Seitz L, Sprenger GA, Albermann C Construction of Escherichia coli strains with chromosomally integrated expression cassettes for the synthesis of 2′-fucosyllactose Microb Cell Fact 2013 12 1 13 Google Scholar OpenURL Placeholder Text WorldCat 7. Belenguer A , Duncan SH, Calder AG, Holtrop G, Louis P, Lobley GE, Flint HJ Two routes of metabolic cross-feeding between Bifidobacterium adolescentis and butyrate-producing anaerobes from the human gut Appl Environ Microbiol 2006 72 3593 3599 1472403 Google Scholar OpenURL Placeholder Text WorldCat 8. Ben X-M , Zhou X-Y, Zhao W-H, Yu W-L, Pan W, Zhang W-L, Wu S-M, Van Beusekom CM, Schaafsma A Supplementation of milk formula with galacto-oligosaccharides improves intestinal micro-flora and fermentation in term infants Chin Med J 2004 117 927 931 Google Scholar OpenURL Placeholder Text WorldCat 9. Bindels LB , Delzenne NM, Cani PD, Walter J Towards a more comprehensive concept for prebiotics Nat Rev Gastroenterol Hepatol 2015 12 303 310 Google Scholar OpenURL Placeholder Text WorldCat 10. Bindels LB , Dewulf EM, Delzenne NM GPR43/FFA2: physiopathological relevance and therapeutic prospects Trends Pharmacol Sci 2013 34 226 232 Google Scholar OpenURL Placeholder Text WorldCat 11. Birt DF , Boylston T, Hendrich S, Jane J-L, Hollis J, Li L, McClelland J, Moore S, Phillips GJ, Rowling M, Schalinske K, Scott MP, Whitley EM Resistant starch: promise for improving human health Adv Nutr 2013 4 587 601 3823506 Google Scholar OpenURL Placeholder Text WorldCat 12. Bode L Human milk oligosaccharides: every baby needs a sugar mama Glycobiology 2012 22 1147 1162 3406618 Google Scholar OpenURL Placeholder Text WorldCat 13. Bode L , Contractor N, Barile D, Pohl N, Prudden AR, Boons G-J, Jin Y-S, Jennewein S Overcoming the limited availability of human milk oligosaccharides: challenges and opportunities for research and application Nutr Rev 2016 74 635 644 6281035 Google Scholar OpenURL Placeholder Text WorldCat 14. Bokulich NA , Chung J, Battaglia T, Henderson N, Jay M, Li H, Lieber AD, Wu F, Perez-Perez GI, Chen Y, Schweizer W, Zheng X, Contreras M, Dominguez-Bello MG, Blaser MJ Antibiotics, birth mode, and diet shape microbiome maturation during early life Sci Transl Med 2016 8 343ra82 5308924 Google Scholar OpenURL Placeholder Text WorldCat 15. Bravo JA , Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, Bienenstock J, Cryan JF Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve Proc Natl Acad Sci USA 2011 108 16050 16055 Google Scholar OpenURL Placeholder Text WorldCat 16. Brown GD Dectin-1: a signalling non-TLR pattern-recognition receptor Nat Rev Immunol 2006 6 33 43 Google Scholar OpenURL Placeholder Text WorldCat 17. Brown GD , Gordon S Immune recognition. A new receptor for beta-glucans Nature 2001 413 36 37 Google Scholar OpenURL Placeholder Text WorldCat 18. Burokas A , Arboleya S, Moloney RD, Peterson VL, Murphy K, Clarke G, Stanton C, Dinan TG, Cryan JF Targeting the microbiota-gut-brain axis: prebiotics have anxiolytic and antidepressant-like effects and reverse the impact of chronic stress in mice Biol Psychiatry 2017 82 472 487 Google Scholar OpenURL Placeholder Text WorldCat 19. Byndloss MX , Olsan EE, Rivera-Chávez F, Tiffany CR, Cevallos SA, Lokken KL, Torres TP, Byndloss AJ, Faber F, Gao Y, Litvak Y, Lopez CA, Xu G, Napoli E, Giulivi C, Tsolis RM, Revzin A, Lebrilla CB, Bäumler AJ Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion Science 2017 357 570 575 5642957 Google Scholar OpenURL Placeholder Text WorldCat 20. Cani PD Gut cell metabolism shapes the microbiome Science 2017 357 548 549 Google Scholar OpenURL Placeholder Text WorldCat 21. Cani PD Human gut microbiome: hopes, threats and promises Gut 2018 67 1716 1725 6109275 Google Scholar OpenURL Placeholder Text WorldCat 22. Chang HN , Lee SY, Nielsen J, Stephanopoulos G Emerging areas in bioengineering 2018 Chichester Wiley Google Scholar Google Preview OpenURL Placeholder Text WorldCat COPAC 23. Charbonneau MR , Blanton LV, DiGiulio DB, Relman DA, Lebrilla CB, Mills DA, Gordon JI A microbial perspective of human developmental biology Nature 2016 535 48 55 5358965 Google Scholar OpenURL Placeholder Text WorldCat 24. Chaturvedi P , Warren CD, Altaye M, Morrow AL, Ruiz-Palacios G, Pickering LK, Newburg DS Fucosylated human milk oligosaccharides vary between individuals and over the course of lactation Glycobiology 2001 11 365 372 Google Scholar OpenURL Placeholder Text WorldCat 25. Chen C , Zhang Y, Xue M, Liu X-W, Li Y, Chen X, Wang PG, Wang F, Cao H Sequential one-pot multienzyme (OPME) synthesis of lacto-N-neotetraose and its sialyl and fucosyl derivatives Chem Commun 2015 51 7689 7692 Google Scholar OpenURL Placeholder Text WorldCat 26. Chen R Enzyme and microbial technology for synthesis of bioactive oligosaccharides: an update Appl Microbiol Biotechnol 2018 102 3017 3026 Google Scholar OpenURL Placeholder Text WorldCat 27. Chichlowski M , De Lartigue G, German JB, Raybould HE, Mills DA Bifidobacteria isolated from infants and cultured on human milk oligosaccharides affect intestinal epithelial function J Pediatr Gastroenterol Nutr 2012 55 321 327 3381975 Google Scholar OpenURL Placeholder Text WorldCat 28. Chunchai T , Thunapong W, Yasom S, Wanchai K, Eaimworawuthikul S, Metzler G, Lungkaphin A, Pongchaidecha A, Sirilun S, Chaiyasut C, Pratchayasakul W, Thiennimitr P, Chattipakorn N, Chattipakorn SC Decreased microglial activation through gut-brain axis by prebiotics, probiotics, or synbiotics effectively restored cognitive function in obese-insulin resistant rats J Neuroinflamm 2018 15 11 Google Scholar OpenURL Placeholder Text WorldCat 29. Chung WSF , Walker AW, Louis P, Parkhill J, Vermeiren J, Bosscher D, Duncan SH, Flint HJ Modulation of the human gut microbiota by dietary fibres occurs at the species level BMC Biol 2016 14 3 4709873 Google Scholar OpenURL Placeholder Text WorldCat 30. Collins MD , Gibson GR Probiotics, prebiotics, and synbiotics: approaches for modulating the microbial ecology of the gut Am J Clin Nutr 1999 69 1052S 1057S Google Scholar OpenURL Placeholder Text WorldCat 31. Comstock SS , Li M, Wang M, Monaco MH, Kuhlenschmidt TB, Kuhlenschmidt MS, Donovan SM Dietary human milk oligosaccharides but not prebiotic oligosaccharides increase circulating natural killer cell and mesenteric lymph node memory t cell populations in noninfected and rotavirus-infected neonatal piglets J Nutr 2017 147 1041 1047 5443461 Google Scholar OpenURL Placeholder Text WorldCat 32. Comstock SS , Wang M, Hester SN, Li M, Donovan SM Select human milk oligosaccharides directly modulate peripheral blood mononuclear cells isolated from 10-d-old pigs Br J Nutr 2014 111 819 828 Google Scholar OpenURL Placeholder Text WorldCat 33. Costabile A , Kolida S, Klinder A, Gietl E, Bäuerlein M, Frohberg C, Landschütze V, Gibson GR 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 2010 104 1007 1017 Google Scholar OpenURL Placeholder Text WorldCat 34. Crout DH , Vic G Glycosidases and glycosyl transferases in glycoside and oligosaccharide synthesis Curr Opin Chem Biol 1998 2 98 111 Google Scholar OpenURL Placeholder Text WorldCat 35. David LA , Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ Diet rapidly and reproducibly alters the human gut microbiome Nature 2014 505 559 563 Google Scholar OpenURL Placeholder Text WorldCat 36. Dewulf EM , Cani PD, Claus SP, Fuentes S, Puylaert PGB, Neyrinck AM, Bindels LB, de Vos WM, Gibson GR, Thissen J-P, Delzenne NM Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women Gut 2013 62 1112 1121 Google Scholar OpenURL Placeholder Text WorldCat 37. Drouillard S , Driguez H, Samain E Large-scale synthesis of H-antigen oligosaccharides by expressing Helicobacter pylori α1,2-fucosyltransferase in metabolically engineered Escherichia coli cells Angew Chem Int Ed 2006 45 1778 1780 Google Scholar OpenURL Placeholder Text WorldCat 38. Durrer KE , Allen MS, Hunt von Herbing I Genetically engineered probiotic for the treatment of phenylketonuria (PKU); assessment of a novel treatment in vitro and in the PAHenu2 mouse model of PKU PLoS One 2017 12 e0176286 5435137 Google Scholar OpenURL Placeholder Text WorldCat 39. El Kaoutari A , Armougom F, Gordon JI, Raoult D, Henrissat B The abundance and variety of carbohydrate-active enzymes in the human gut microbiota Nat Rev Microbiol 2013 11 497 504 Google Scholar OpenURL Placeholder Text WorldCat 40. Endo T , Koizumi S, Tabata K, Ozaki A Large-scale production of CMP-NeuAc and sialylated oligosaccharides through bacterial coupling Appl Microbiol Biotechnol 2000 53 257 261 Google Scholar OpenURL Placeholder Text WorldCat 41. Fair RJ , Hahm HS, Seeberger PH Combination of automated solid-phase and enzymatic oligosaccharide synthesis provides access to α(2,3)-sialylated glycans Chem Commun 2015 51 6183 6185 Google Scholar OpenURL Placeholder Text WorldCat 42. Fanaro S , Marten B, Bagna R, Vigi V, Fabris C, Peña-Quintana L, Argüelles F, Scholz-Ahrens KE, Sawatzki G, Zelenka R, Schrezenmeir J, de Vrese M, Bertino E Galacto-oligosaccharides are bifidogenic and safe at weaning: a double-blind randomized multicenter study J Pediatr Gastroenterol Nutr 2009 48 82 88 Google Scholar OpenURL Placeholder Text WorldCat 43. Ferenczi S , Szegi K, Winkler Z, Barna T, Kovács KJ Oligomannan prebiotic attenuates immunological, clinical and behavioral symptoms in mouse model of inflammatory bowel disease Sci Rep 2016 6 34132 5034233 Google Scholar OpenURL Placeholder Text WorldCat 44. Fernandes R , do Rosario VA, Mocellin MC, Kuntz MGF, Trindade EBSM Effects of inulin-type fructans, galacto-oligosaccharides and related synbiotics on inflammatory markers in adult patients with overweight or obesity: a systematic review Clin Nutr 2017 36 1197 1206 Google Scholar OpenURL Placeholder Text WorldCat 45. Ferreira RM , Pereira-Marques J, Pinto-Ribeiro I, Costa JL, Carneiro F, Machado JC, Figueiredo C Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota Gut 2018 67 226 236 Google Scholar OpenURL Placeholder Text WorldCat 46. Floch MH Fecal bacteriotherapy, fecal transplant, and the microbiome J Clin Gastroenterol 2010 44 529 530 Google Scholar OpenURL Placeholder Text WorldCat 47. Foster JA , McVey Neufeld K-A Gut-brain axis: how the microbiome influences anxiety and depression Trends Neurosci 2013 36 305 312 Google Scholar OpenURL Placeholder Text WorldCat 48. Freeman LR , Haley-Zitlin V, Rosenberger DS, Granholm A-C Damaging effects of a high-fat diet to the brain and cognition: a review of proposed mechanisms Nutr Neurosci 2014 17 241 251 Google Scholar OpenURL Placeholder Text WorldCat 49. Furusawa Y , Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, Takahashi M, Fukuda NN, Murakami S, Miyauchi E, Hino S, Atarashi K, Onawa S, Fujimura Y, Lockett T, Clarke JM, Topping DL, Tomita M, Hori S, Ohara O, Morita T, Koseki H, Kikuchi J, Honda K, Hase K, Ohno H Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells Nature 2013 504 446 450 Google Scholar OpenURL Placeholder Text WorldCat 50. Gibson GR , Hutkins R, Sanders ME, Prescott SL, Reimer RA, Salminen SJ, Scott K, Stanton C, Swanson KS, Cani PD, Verbeke K, Reid G Expert consensus document: the International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics Nat Rev Gastroenterol Hepatol 2017 14 491 502 Google Scholar OpenURL Placeholder Text WorldCat 51. Gibson GR , Probert HM, Loo JV, Rastall RA, Roberfroid MB Dietary modulation of the human colonic microbiota: updating the concept of prebiotics Nutr Res Rev 2004 17 259 275 Google Scholar OpenURL Placeholder Text WorldCat 52. Gibson GR , Rastall RA Prebiotics: development & application 2006 Chichester Wiley Google Scholar Google Preview OpenURL Placeholder Text WorldCat COPAC 53. Gibson GR , Roberfroid MB Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics J Nutr 1995 125 1401 1412 Google Scholar OpenURL Placeholder Text WorldCat 54. Gill SR , Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, Gordon JI, Relman DA, Fraser-Liggett CM, Nelson KE Metagenomic analysis of the human distal gut microbiome Science 2006 312 1355 1359 3027896 Google Scholar OpenURL Placeholder Text WorldCat 55. Grulee CG , Sanford HN, Herron PH Breast and artificial feeding: influence on morbidity and mortality of twenty thousand infants JAMA 1934 103 735 739 Google Scholar OpenURL Placeholder Text WorldCat 56. Guo L , Chen X, Xu L, Xiao M, Lu L Enzymatic synthesis of 6′-sialyllactose, a dominant sialylated human milk oligosaccharide, by a novel exo-α-sialidase from Bacteroides fragilis NCTC9343 Appl Environ Microbiol 2018 10.1128/aem.00071-18 6275343 Google Scholar OpenURL Placeholder Text WorldCat Crossref 57. Gyorgy P , Norris RF, Rose CS Bifidus factor. I. A variant of Lactobacillus bifidus requiring a special growth factor Arch Biochem Biophys 1954 48 193 201 Google Scholar OpenURL Placeholder Text WorldCat 58. Haberman Y , Tickle TL, Dexheimer PJ, Kim M-O, Tang D, Karns R, Baldassano RN, Noe JD, Rosh J, Markowitz J, Heyman MB, Griffiths AM, Crandall WV, Mack DR, Baker SS, Huttenhower C, Keljo DJ, Hyams JS, Kugathasan S, Walters TD, Aronow B, Xavier RJ, Gevers D, Denson LA Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature J Clin Invest 2014 124 3617 3633 4109533 Google Scholar OpenURL Placeholder Text WorldCat 59. Hansen CHF , Frøkiær H, Christensen AG, Bergström A, Licht TR, Hansen AK, Metzdorff SB Dietary xylooligosaccharide downregulates IFN-γ and the low-grade inflammatory cytokine IL-1β systemically in mice J Nutr 2013 143 533 540 Google Scholar OpenURL Placeholder Text WorldCat 60. Hardy H , Harris J, Lyon E, Beal J, Foey AD Probiotics, prebiotics and immunomodulation of gut mucosal defences: homeostasis and immunopathology Nutrients 2013 5 1869 1912 3725482 Google Scholar OpenURL Placeholder Text WorldCat 61. Herrmann GF , Elling L, Krezdorn CH, Kleene R, Berger EG, Wandrey C Use of transformed whole yeast cells expressing β-1,4-galactosyltransferase for the synthesis of N-acetyllactosamine Bioorg Med Chem Lett 1995 5 673 676 Google Scholar OpenURL Placeholder Text WorldCat 62. Herrmann GF , Wang P, Shen G-J, Wong C-H Recombinant whole cells as catalysts for the enzymatic synthesis of oligosaccharides and glycopeptides Angew Chem Int Ed Engl 1994 33 1241 1242 Google Scholar OpenURL Placeholder Text WorldCat 63. Hoentjen F , Welling GW, Harmsen HJM, Zhang X, Snart J, Tannock GW, Lien K, Churchill TA, Lupicki M, Dieleman LA Reduction of colitis by prebiotics in HLA-B27 transgenic rats is associated with microflora changes and immunomodulation Inflamm Bowel Dis 2005 11 977 985 Google Scholar OpenURL Placeholder Text WorldCat 64. Hosono A , Ozawa A, Kato R, Ohnishi Y, Nakanishi Y, Kimura T, Nakamura R Dietary fructooligosaccharides induce immunoregulation of intestinal IgA secretion by murine Peyer’s patch cells Biosci Biotechnol Biochem 2003 67 758 764 Google Scholar OpenURL Placeholder Text WorldCat 65. Howard MD , Gordon DT, Garleb KA, Kerley MS Dietary fructooligosaccharide, xylooligosaccharide and gum arabic have variable effects on cecal and colonic microbiota and epithelial cell proliferation in mice and rats J Nutr 1995 125 2604 2609 Google Scholar OpenURL Placeholder Text WorldCat 66. Hsu C-K , Liao J-W, Chung Y-C, Hsieh C-P, Chan Y-C Xylooligosaccharides and fructooligosaccharides affect the intestinal microbiota and precancerous colonic lesion development in rats J Nutr 2004 134 1523 1528 Google Scholar OpenURL Placeholder Text WorldCat 67. Isabella VM , Ha BN, Castillo MJ, Lubkowicz DJ, Rowe SE, Millet YA, Anderson CL, Li N, Fisher AB, West KA, Reeder PJ, Momin MM, Bergeron CG, Guilmain SE, Miller PF, Kurtz CB, Falb D Development of a synthetic live bacterial therapeutic for the human metabolic disease phenylketonuria Nat Biotechnol 2018 36 857 864 Google Scholar OpenURL Placeholder Text WorldCat 68. Kassam Z , Hundal R, Marshall JK, Lee CH Fecal transplant via retention enema for refractory or recurrent Clostridium difficile infection Arch Intern Med 2012 172 191 193 Google Scholar OpenURL Placeholder Text WorldCat 69. Bych K , Miks MH, Johanson T, Hederos MJ, Vigsnaes LK, Becker P Production of HMOs using microbial hosts—from cell engineering to large scale production Curr Opin Biotechnol 2018 56 130 137 Google Scholar OpenURL Placeholder Text WorldCat 70. Kearney SM , Gibbons SM, Erdman SE, Alm EJ Orthogonal dietary niche enables reversible engraftment of a gut bacterial commensal Cell Rep 2018 24 1842 1851 6724203 Google Scholar OpenURL Placeholder Text WorldCat 71. Kim G-B , Seo YM, Kim CH, Paik IK Effect of dietary prebiotic supplementation on the performance, intestinal microflora, and immune response of broilers Poult Sci 2011 90 75 82 Google Scholar OpenURL Placeholder Text WorldCat 72. King A Prevention. Cost-effectiveness of cardiovascular disease prevention and management in the developing world Nat Rev Cardiol 2012 9 258 Google Scholar OpenURL Placeholder Text WorldCat 73. Klinder A , Forster A, Caderni G, Femia AP, Pool-Zobel BL Fecal water genotoxicity is predictive of tumor-preventive activities by inulin-like oligofructoses, probiotics (Lactobacillus rhamnosus and Bifidobacterium lactis), and their synbiotic combination Nutr Cancer 2004 49 144 155 Google Scholar OpenURL Placeholder Text WorldCat 74. Kobayashi T , Okazaki M, Fujikawa S, Koga K Effect of xylooligosaccharides on feces of men J Jpn Soc Biosci Biotech Agrochem 1991 65 1651 1653 Google Scholar OpenURL Placeholder Text WorldCat 75. Koeth RA , Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WHW, Bushman FD, Lusis AJ, Hazen SL Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis Nat Med 2013 19 576 585 3650111 Google Scholar OpenURL Placeholder Text WorldCat 76. Kontula P , von Wright A, Mattila-Sandholm T Oat bran β-gluco- and xylo-oligosaccharides as fermentative substrates for lactic acid bacteria Int J Food Microbiol 1998 45 163 169 Google Scholar OpenURL Placeholder Text WorldCat 77. Kostic AD , Xavier RJ, Gevers D The microbiome in inflammatory bowel disease: current status and the future ahead Gastroenterology 2014 146 1489 1499 4034132 Google Scholar OpenURL Placeholder Text WorldCat 78. Kunz C , Rudloff S, Baier W, Klein N, Strobel S Oligosaccharides in human milk: structural, functional, and metabolic aspects Annu Rev Nutr 2000 20 699 722 Google Scholar OpenURL Placeholder Text WorldCat 79. Kurtz CB , Millet YA, Puurunen MK, Perreault M, Charbonneau MR, Isabella VM, Kotula JW, Antipov E, Dagon Y, Denney WS, Wagner DA, West KA, Degar AJ, Brennan AM, Miller PF An engineered E. coli Nissle improves hyperammonemia and survival in mice and shows dose-dependent exposure in healthy humans Sci Transl Med 2019 10.1126/scitranslmed.aau7975 Google Scholar OpenURL Placeholder Text WorldCat Crossref 80. Kurtz C , Denney WS, Blankstein L, Guilmain SE, Machinani S, Kotula J, Saha S, Miller P, Brennan AM Translational development of microbiome-based therapeutics: kinetics of E. coli Nissle and engineered strains in humans and nonhuman primates Clin Transl Sci 2018 11 200 207 Google Scholar OpenURL Placeholder Text WorldCat 81. Lee W-H , Pathanibul P, Quarterman J, Jo J-H, Han NS, Miller MJ, Jin Y-S, Seo J-H Whole cell biosynthesis of a functional oligosaccharide, 2′-fucosyllactose, using engineered Escherichia coli Microb Cell Fact 2012 11 1 9 Google Scholar OpenURL Placeholder Text WorldCat 82. Lewandowska U , Szewczyk K, Hrabec E, Janecka A, Gorlach S Overview of metabolism and bioavailability enhancement of polyphenols J Agric Food Chem 2013 61 12183 12199 Google Scholar OpenURL Placeholder Text WorldCat 83. Lewis S , Burmeister S, Brazier J Effect of the Prebiotic oligofructose on relapse of Clostridium difficile-associated diarrhea: a randomized, controlled study Clin Gastroenterol Hepatol 2005 3 442 448 Google Scholar OpenURL Placeholder Text WorldCat 84. Ley RE , Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI Obesity alters gut microbial ecology Proc Natl Acad Sci USA 2005 102 11070 11075 Google Scholar OpenURL Placeholder Text WorldCat 85. Liu RH Potential synergy of phytochemicals in cancer prevention: mechanism of action J Nutr 2004 134 3479S 3485S Google Scholar OpenURL Placeholder Text WorldCat 86. Makki K , Deehan EC, Walter J, Bäckhed F The impact of dietary fiber on gut microbiota in host health and disease Cell Host Microbe 2018 23 705 715 Google Scholar OpenURL Placeholder Text WorldCat 87. Manthey CF , Autran CA, Eckmann L, Bode L Human milk oligosaccharides protect against enteropathogenic Escherichia coli attachment in vitro and EPEC colonization in suckling mice J Pediatr Gastroenterol Nutr 2014 58 165 168 Google Scholar OpenURL Placeholder Text WorldCat 88. Marcobal A , Barboza M, Sonnenburg ED, Pudlo N, Martens EC, Desai P, Lebrilla CB, Weimer BC, Mills DA, German JB, Sonnenburg JL Bacteroides in the infant gut consume milk oligosaccharides via mucus-utilization pathways Cell Host Microbe 2011 10 507 514 3227561 Google Scholar OpenURL Placeholder Text WorldCat 89. Mariño E , Richards JL, McLeod KH, Stanley D, Yap YA, Knight J, McKenzie C, Kranich J, Oliveira AC, Rossello FJ, Krishnamurthy B, Nefzger CM, Macia L, Thorburn A, Baxter AG, Morahan G, Wong LH, Polo JM, Moore RJ, Lockett TJ, Clarke JM, Topping DL, Harrison LC, Mackay CR Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes Nat Immunol 2017 18 552 562 Google Scholar OpenURL Placeholder Text WorldCat 90. Martens EC , Koropatkin NM, Smith TJ, Gordon JI Complex glycan catabolism by the human gut microbiota: the Bacteroidetes Sus-like paradigm J Biol Chem 2009 284 24673 24677 2757170 Google Scholar OpenURL Placeholder Text WorldCat 91. McNulty NP , Wu M, Erickson AR, Pan C, Erickson BK, Martens EC, Pudlo NA, Muegge BD, Henrissat B, Hettich RL, Gordon JI Effects of diet on resource utilization by a model human gut microbiota containing Bacteroides cellulosilyticus WH2, a symbiont with an extensive glycobiome PLoS Biol 2013 11 e1001637 3747994 Google Scholar OpenURL Placeholder Text WorldCat 92. Miene C , Weise A, Glei M Impact of polyphenol metabolites produced by colonic microbiota on expression of COX-2 and GSTT2 in human colon cells (LT97) Nutr Cancer 2011 63 653 662 Google Scholar OpenURL Placeholder Text WorldCat 93. Miller TL , Wolin MJ Pathways of acetate, propionate, and butyrate formation by the human fecal microbial flora Appl Environ Microbiol 1996 62 1589 1592 167932 Google Scholar OpenURL Placeholder Text WorldCat 94. Momeni-Moghaddam P , Keyvanshokooh S, Ziaei-Nejad S, Parviz Salati A, Pasha-Zanoosi H Effects of mannan oligosaccharide supplementation on growth, some immune responses and gut lactic acid bacteria of common carp (Cyprinus Carpio) fingerlings Vet Res Forum 2015 6 239 244 4611979 Google Scholar OpenURL Placeholder Text WorldCat 95. Moro G , Arslanoglu S, Stahl B, Jelinek J, Wahn U, Boehm G A mixture of prebiotic oligosaccharides reduces the incidence of atopic dermatitis during the first six months of age Arch Dis Child 2006 91 814 819 2066015 Google Scholar OpenURL Placeholder Text WorldCat 96. Muegge BD , Kuczynski J, Knights D, Clemente JC, González A, Fontana L, Henrissat B, Knight R, Gordon JI Diet drives convergence in gut microbiome functions across mammalian phylogeny and within humans Science 2011 332 970 974 3303602 Google Scholar OpenURL Placeholder Text WorldCat 97. Musso G , Gambino R, Cassader M Gut microbiota as a regulator of energy homeostasis and ectopic fat deposition: mechanisms and implications for metabolic disorders Curr Opin Lipidol 2010 21 76 83 Google Scholar OpenURL Placeholder Text WorldCat 98. Nagura T , Hachimura S, Hashiguchi M, Ueda Y, Kanno T, Kikuchi H, Sayama K, Kaminogawa S Suppressive effect of dietary raffinose on T-helper 2 cell-mediated immunity Br J Nutr 2002 88 421 426 Google Scholar OpenURL Placeholder Text WorldCat 99. Newburg DS , Walker WA Protection of the neonate by the innate immune system of developing gut and of human milk Pediatr Res 2007 61 2 8 Google Scholar OpenURL Placeholder Text WorldCat 100. Niness KR Inulin and oligofructose: what are they? J Nutr 1999 129 1402S 1406S Google Scholar OpenURL Placeholder Text WorldCat 101. Obermeier S , Rudloff S, Pohlentz G, Lentze MJ, Kunz C Secretion of 13C-labelled oligosaccharides into human milk and infant’s urine after an oral [13C]galactose load Isotopes Environ Health Stud 1999 35 119 125 Google Scholar OpenURL Placeholder Text WorldCat 102. Ofek I , Beachey EH Mannose binding and epithelial cell adherence of Escherichia coli Infect Immunity 1978 22 247 254 Google Scholar OpenURL Placeholder Text WorldCat 103. Okazaki M , Fujikawa S, Matsumoto N Effect of xylooligosaccharide on the growth of bifidobacteria Bifidobact Microflora 1990 9 77 86 Google Scholar OpenURL Placeholder Text WorldCat 104. Osman N , Adawi D, Molin G, Ahrne S, Berggren A, Jeppsson B Bifidobacterium infantis strains with and without a combination of oligofructose and inulin (OFI) attenuate inflammation in DSS-induced colitis in rats BMC Gastroenterol 2006 6 31 1634862 Google Scholar OpenURL Placeholder Text WorldCat 105. Paineau D , Payen F, Panserieu S, Coulombier G, Sobaszek A, Lartigau I, Brabet M, Galmiche J-P, Tripodi D, Sacher-Huvelin S, Chapalain V, Zourabichvili O, Respondek F, Wagner A, Bornet FRJ The effects of regular consumption of short-chain fructo-oligosaccharides on digestive comfort of subjects with minor functional bowel disorders Br J Nutr 2008 99 311 318 Google Scholar OpenURL Placeholder Text WorldCat 106. Pant K , Yadav AK, Gupta P, Islam R, Saraya A, Venugopal SK Butyrate induces ROS-mediated apoptosis by modulating miR-22/SIRT-1 pathway in hepatic cancer cells Redox Biol 2017 12 340 349 5350572 Google Scholar OpenURL Placeholder Text WorldCat 107. Parnell JA , Reimer RA Effect of prebiotic fibre supplementation on hepatic gene expression and serum lipids: a dose-response study in JCR:LA-cp rats Br J Nutr 2010 103 1577 1584 Google Scholar OpenURL Placeholder Text WorldCat 108. Pereira FC , Berry D Microbial nutrient niches in the gut Environ Microbiol 2017 19 1366 1378 5412925 Google Scholar OpenURL Placeholder Text WorldCat 109. Perugino G , Trincone A, Rossi M, Moracci M Oligosaccharide synthesis by glycosynthases Trends Biotechnol 2004 22 31 37 Google Scholar OpenURL Placeholder Text WorldCat 110. Phelps CF The physical properties of inulin solutions Biochem J 1965 95 41 47 1215175 Google Scholar OpenURL Placeholder Text WorldCat 111. Plante OJ , Palmacci ER, Seeberger PH Automated solid-phase synthesis of oligosaccharides Science 2001 291 1523 1527 Google Scholar OpenURL Placeholder Text WorldCat 112. Pourghassem Gargari B , Dehghan P, Aliasgharzadeh A, Asghari Jafar-Abadi M Effects of high performance inulin supplementation on glycemic control and antioxidant status in women with type 2 diabetes Diabetes Metab J 2013 37 140 148 3638225 Google Scholar OpenURL Placeholder Text WorldCat 113. Priem B , Gilbert M, Wakarchuk WW, Heyraud A, Samain E A new fermentation process allows large-scale production of human milk oligosaccharides by metabolically engineered bacteria Glycobiology 2002 12 235 240 Google Scholar OpenURL Placeholder Text WorldCat 114. Propst EL , Flickinger EA, Bauer LL, Merchen NR, Fahey GC Jr A dose-response experiment evaluating the effects of oligofructose and inulin on nutrient digestibility, stool quality, and fecal protein catabolites in healthy adult dogs J Anim Sci 2003 81 3057 3066 Google Scholar OpenURL Placeholder Text WorldCat 115. Prudden AR , Chinoy ZS, Wolfert MA, Boons G-J A multifunctional anomeric linker for the chemoenzymatic synthesis of complex oligosaccharides Chem Commun 2014 50 7132 7135 Google Scholar OpenURL Placeholder Text WorldCat 116. Quintero M , Maldonado M, Perez-Munoz M, Jimenez R, Fangman T, Rupnow J, Wittke A, Russell M, Hutkins R Adherence inhibition of Cronobacter sakazakii to intestinal epithelial cells by prebiotic oligosaccharides Curr Microbiol 2011 62 1448 1454 Google Scholar OpenURL Placeholder Text WorldCat 117. Rastall RA , Gibson GR Recent developments in prebiotics to selectively impact beneficial microbes and promote intestinal health Curr Opin Biotechnol 2015 32 42 46 Google Scholar OpenURL Placeholder Text WorldCat 118. dos Reis SA , da Conceição LL, Rosa DD, Dias MMDS, Peluzio MDCG Mechanisms used by inulin-type fructans to improve the lipid profile Nutr Hosp 2014 31 528 534 Google Scholar OpenURL Placeholder Text WorldCat 119. Roberfroid MB Introducing inulin-type fructans Br J Nutr 2005 93 S13 S25 Google Scholar OpenURL Placeholder Text WorldCat 120. Rogler G , Andus T Cytokines in inflammatory bowel disease World J Surg 1998 22 382 389 Google Scholar OpenURL Placeholder Text WorldCat 121. Roller M , Rechkemmer G, Watzl B Prebiotic inulin enriched with oligofructose in combination with the probiotics Lactobacillus rhamnosus and Bifidobacterium lactis modulates intestinal immune functions in rats J Nutr 2004 134 153 156 Google Scholar OpenURL Placeholder Text WorldCat 122. Ross GD , Větvička V CR122 (CD11b, CD18): a phagocyte and NK cell membrane receptor with multiple ligand specificities and functions Clin Exp Immunol 2008 92 181 184 Google Scholar OpenURL Placeholder Text WorldCat 123. Rowland IR , Rumney CJ, Coutts JT, Lievense LC Effect of Bifidobacterium longum and inulin on gut bacterial metabolism and carcinogen-induced aberrant crypt foci in rats Carcinogenesis 1998 19 281 285 Google Scholar OpenURL Placeholder Text WorldCat 124. Ruffing A , Mao Z, Ruizhen Chen R Metabolic engineering of Agrobacterium sp. for UDP-galactose regeneration and oligosaccharide synthesis Metab Eng 2006 8 465 473 Google Scholar OpenURL Placeholder Text WorldCat 125. Ruiz-Palacios GM , Cervantes LE, Ramos P, Chavez-Munguia B, Newburg DS Campylobacter jejuni binds intestinal H(O) antigen (Fucα1, 2Galβ1, 4GlcNAc), and fucosyloligosaccharides of human milk inhibit its binding and infection J Biol Chem 2003 278 14112 14120 Google Scholar OpenURL Placeholder Text WorldCat 126. Salyers AA , Vercellotti JR, West SE, Wilkins TD Fermentation of mucin and plant polysaccharides by strains of Bacteroides from the human colon Appl Environ Microbiol 1977 33 319 322 170684 Google Scholar OpenURL Placeholder Text WorldCat 127. Salyers AA , West SE, Vercellotti JR, Wilkins TD Fermentation of mucins and plant polysaccharides by anaerobic bacteria from the human colon Appl Environ Microbiol 1977 34 529 533 242695 Google Scholar OpenURL Placeholder Text WorldCat 128. Saville BA , Saville S Xylooligosaccharides and arabinoxylanoligosaccharides and their application as prebiotics Appl Food Biotechnol 2018 5 121 130 Google Scholar OpenURL Placeholder Text WorldCat 129. Scholz-Ahrens KE , Schaafsma G, van den Heuvel EG, Schrezenmeir J Effects of prebiotics on mineral metabolism Am J Clin Nutr 2001 73 459S 464S Google Scholar OpenURL Placeholder Text WorldCat 130. Schuster M , Wang P, Paulson JC, Wong C-H Solid-Phase Chemical-Enzymic Synthesis of Glycopeptides and Oligosaccharides J Am Chem Soc 1994 116 1135 1136 Google Scholar OpenURL Placeholder Text WorldCat 131. Sears P , Wong CH Toward automated synthesis of oligosaccharides and glycoproteins Science 2001 291 2344 2350 Google Scholar OpenURL Placeholder Text WorldCat 132. Sender R , Fuchs S, Milo R Revised estimates for the number of human and bacteria cells in the body PLoS Biol 2016 14 e1002533 4991899 Google Scholar OpenURL Placeholder Text WorldCat 133. Sender R , Fuchs S, Milo R Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans Cell 2016 164 337 340 Google Scholar OpenURL Placeholder Text WorldCat 134. Shepherd ES , DeLoache WC, Pruss KM, Whitaker WR, Sonnenburg JL An exclusive metabolic niche enables strain engraftment in the gut microbiota Nature 2018 557 434 438 6126907 Google Scholar OpenURL Placeholder Text WorldCat 135. Sierra C , Bernal M-J, Blasco J, Martínez R, Dalmau J, Ortuño I, Espín B, Vasallo M-I, Gil D, Vidal M-L, Infante D, Leis R, Maldonado J, Moreno J-M, Román E Prebiotic effect during the first year of life in healthy infants fed formula containing GOS as the only prebiotic: a multicentre, randomised, double-blind and placebo-controlled trial Eur J Nutr 2015 54 89 99 Google Scholar OpenURL Placeholder Text WorldCat 136. Silk DBA , Davis A, Vulevic J, Tzortzis G, Gibson GR Clinical trial: the effects of a trans-galactooligosaccharide prebiotic on faecal microbiota and symptoms in irritable bowel syndrome Aliment Pharmacol Ther 2009 29 508 518 Google Scholar OpenURL Placeholder Text WorldCat 137. Smiricky-Tjardes MR , Flickinger EA, Grieshop CM, Bauer LL, Murphy MR, Fahey GC Jr In vitro fermentation characteristics of selected oligosaccharides by swine fecal microflora J Anim Sci 2003 81 2505 2514 Google Scholar OpenURL Placeholder Text WorldCat 138. Smith PM , Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garrett WS The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis Science 2013 341 569 573 Google Scholar OpenURL Placeholder Text WorldCat 139. Stewart ML , Timm DA, Slavin JL Fructooligosaccharides exhibit more rapid fermentation than long-chain inulin in an in vitro fermentation system Nutr Res 2008 28 329 334 Google Scholar OpenURL Placeholder Text WorldCat 140. Tang WHW , Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen SL Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk N Engl J Med 2013 368 1575 1584 3701945 Google Scholar OpenURL Placeholder Text WorldCat 141. Tan J , McKenzie C, Vuillermin PJ, Goverse G, Vinuesa CG, Mebius RE, Macia L, Mackay CR Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways Cell Rep 2016 15 2809 2824 Google Scholar OpenURL Placeholder Text WorldCat 142. Thilakarathna WW , Langille MGI, Rupasinghe HPV Polyphenol-based prebiotics and synbiotics: potential for cancer chemoprevention Curr Opin Food Sci 2018 20 51 57 Google Scholar OpenURL Placeholder Text WorldCat 143. Thompson WG , Longstreth GF, Drossman DA, Heaton KW, Irvine EJ, Müller-Lissner SA Functional bowel disorders and functional abdominal pain Gut 1999 45 II43 II47 1766683 Google Scholar OpenURL Placeholder Text WorldCat 144. Thongaram T , Hoeflinger JL, Chow J, Miller MJ Prebiotic galactooligosaccharide metabolism by probiotic lactobacilli and bifidobacteria J Agric Food Chem 2017 65 4184 4192 Google Scholar OpenURL Placeholder Text WorldCat 145. Tilg H , Adolph TE, Gerner RR, Moschen AR The intestinal microbiota in colorectal cancer Cancer Cell 2018 33 954 964 Google Scholar OpenURL Placeholder Text WorldCat 146. Tomás-Barberán FA , Selma MV, Espín JC Interactions of gut microbiota with dietary polyphenols and consequences to human health Curr Opin Clin Nutr Metab Care 2016 19 471 476 Google Scholar OpenURL Placeholder Text WorldCat 147. Trautwein EA , Rieckhoff D, Erbersdobler HF Dietary inulin lowers plasma cholesterol and triacylglycerol and alters biliary bile acid profile in hamsters J Nutr 1998 128 1937 1943 Google Scholar OpenURL Placeholder Text WorldCat 148. Treves DS , Manning S, Adams J Repeated evolution of an acetate-crossfeeding polymorphism in long-term populations of Escherichia coli Mol Biol Evol 1998 15 789 797 Google Scholar OpenURL Placeholder Text WorldCat 149. Tuohy KM , Probert HM, Smejkal CW, Gibson GR Using probiotics and prebiotics to improve gut health Drug Discov Today 2003 8 692 700 Google Scholar OpenURL Placeholder Text WorldCat 150. Turnbaugh PJ , Bäckhed F, Fulton L, Gordon JI Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome Cell Host Microbe 2008 3 213 223 Google Scholar OpenURL Placeholder Text WorldCat 151. Turroni F , Milani C, Duranti S, Mahony J, van Sinderen D, Ventura M Glycan utilization and cross-feeding activities by bifidobacteria Trends Microbiol 2018 26 339 350 Google Scholar OpenURL Placeholder Text WorldCat 152. Tzortzis G , Vulevic J Charalampopoulos D, Rastall RA Galacto-oligosaccharide prebiotics Prebiotics and probiotics science and technology 2009 New York Springer 207 244 Google Scholar Google Preview OpenURL Placeholder Text WorldCat COPAC 153. Underwood MA , Gaerlan S, De Leoz MLA, Dimapasoc L, Kalanetra KM, Lemay DG, German JB, Mills DA, Lebrilla CB Human milk oligosaccharides in premature infants: absorption, excretion, and influence on the intestinal microbiota Pediatr Res 2015 78 670 677 4689671 Google Scholar OpenURL Placeholder Text WorldCat 154. Underwood MA , German JB, Lebrilla CB, Mills DA Bifidobacterium longum subspecies infantis: champion colonizer of the infant gut Pediatr Res 2015 77 229 235 Google Scholar OpenURL Placeholder Text WorldCat 155. Vandeputte D , Falony G, Vieira-Silva S, Wang J, Sailer M, Theis S, Verbeke K, Raes J Prebiotic inulin-type fructans induce specific changes in the human gut microbiota Gut 2017 66 1968 1974 5739857 Google Scholar OpenURL Placeholder Text WorldCat 156. Vatanen T , Franzosa EA, Schwager R, Tripathi S, Arthur TD, Vehik K, Lernmark Å, Hagopian WA, Rewers MJ, She J-X, Toppari J, Ziegler A-G, Akolkar B, Krischer JP, Stewart CJ, Ajami NJ, Petrosino JF, Gevers D, Lähdesmäki H, Vlamakis H, Huttenhower C, Xavier RJ The human gut microbiome in early-onset type 1 diabetes from the TEDDY study Nature 2018 562 589 594 Google Scholar OpenURL Placeholder Text WorldCat 157. Vinolo MAR , Rodrigues HG, Nachbar RT, Curi R Regulation of inflammation by short chain fatty acids Nutrients 2011 3 858 876 3257741 Google Scholar OpenURL Placeholder Text WorldCat 158. Vogt L , Meyer D, Pullens G, Faas M, Smelt M, Venema K, Ramasamy U, Schols HA, De Vos P Immunological properties of inulin-type fructans Crit Rev Food Sci Nutr 2015 55 414 436 Google Scholar OpenURL Placeholder Text WorldCat 159. Vohra Y , Vasan M, Venot A, Boons G-J One-pot synthesis of oligosaccharides by combining reductive openings of benzylidene acetals and glycosylations Org Lett 2008 10 3247 3250 2814312 Google Scholar OpenURL Placeholder Text WorldCat 160. Vulevic J , Drakoularakou A, Yaqoob P, Tzortzis G, Gibson GR 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 2008 88 1438 1446 Google Scholar OpenURL Placeholder Text WorldCat 161. Waldecker M , Kautenburger T, Daumann H, Busch C, Schrenk D Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon J Nutr Biochem 2008 19 587 593 Google Scholar OpenURL Placeholder Text WorldCat 162. Wang C-C , Lee J-C, Luo S-Y, Kulkarni SS, Huang Y-W, Lee C-C, Chang K-L, Hung S-C Regioselective one-pot protection of carbohydrates Nature 2007 446 896 899 Google Scholar OpenURL Placeholder Text WorldCat 163. Weng M , Ganguli K, Zhu W, Shi HN, Walker WA Conditioned medium from Bifidobacterium infantis protects against Cronobacter sakazakii-induced intestinal inflammation in newborn mice Am J Physiol Gastrointest Liver Physiol 2014 306 G779 G787 4010653 Google Scholar OpenURL Placeholder Text WorldCat 164. Whistler RL , Bushway AA, Singh PP, Nakahara W, Tokuzen R Tipson RS Noncytotoxic, antitumor polysaccharides Advances in carbohydrate chemistry and biochemistry 1976 Cambridge Academic Press 235 275 Google Scholar Google Preview OpenURL Placeholder Text WorldCat COPAC 165. White BA , Lamed R, Bayer EA, Flint HJ Biomass utilization by gut microbiomes Annu Rev Microbiol 2014 68 279 296 Google Scholar OpenURL Placeholder Text WorldCat 166. Xiao L , Van’t Land B, Engen PA, Naqib A, Green SJ, Nato A, Leusink-Muis T, Garssen J, Keshavarzian A, Stahl B, Folkerts G Human milk oligosaccharides protect against the development of autoimmune diabetes in NOD-mice Sci Rep 2018 8 3829 5832804 Google Scholar OpenURL Placeholder Text WorldCat 167. Yamashita K , Kawai K, Itakura M Effects of fructo-oligosaccharides on blood glucose and serum lipids in diabetic subjects Nutr Res 1984 4 961 966 Google Scholar OpenURL Placeholder Text WorldCat 168. Yatsunenko T , Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, Heath AC, Warner B, Reeder J, Kuczynski J, Caporaso JG, Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R, Gordon JI Human gut microbiome viewed across age and geography Nature 2012 486 222 227 3376388 Google Scholar OpenURL Placeholder Text WorldCat 169. Yu J , Shin J, Park M, Seydametova E, Jung S-M, Seo J-H, Kweon D-H Engineering of α-1,3-fucosyltransferases for production of 3-fucosyllactose in Escherichia coli Metab Eng 2018 48 269 278 Google Scholar OpenURL Placeholder Text WorldCat 170. Yu S , Liu J-J, Yun EJ, Kwak S, Kim KH, Jin Y-S Production of a human milk oligosaccharide 2′-fucosyllactose by metabolically engineered Saccharomyces cerevisiae Microb Cell Fact 2018 17 101 6020385 Google Scholar OpenURL Placeholder Text WorldCat 171. Yu Y , Lasanajak Y, Song X, Hu L, Ramani S, Mickum ML, Ashline DJ, Prasad BVV, Estes MK, Reinhold VN, Cummings RD, Smith DF Human milk contains novel glycans that are potential decoy receptors for neonatal rotaviruses Mol Cell Proteom 2014 13 2944 2960 Google Scholar OpenURL Placeholder Text WorldCat 172. Zenhom M , Hyder A, de Vrese M, Heller KJ, Roeder T, Schrezenmeir J Prebiotic oligosaccharides reduce proinflammatory cytokines in intestinal Caco-2 cells via activation of PPARγ and peptidoglycan recognition protein 3 J Nutr 2011 141 971 977 Google Scholar OpenURL Placeholder Text WorldCat 173. Zeuner B , Vuillemin M, Holck J, Muschiol J, Meyer AS Loop engineering of an α-1,3/4-l-fucosidase for improved synthesis of human milk oligosaccharides Enzyme Microb Technol 2018 115 37 44 Google Scholar OpenURL Placeholder Text WorldCat 174. Zhang Z , Ollmann IR, Ye X-S, Wischnat R, Baasov T, Wong C-H Programmable one-pot oligosaccharide synthesis J Am Chem Soc 1999 121 734 753 Google Scholar OpenURL Placeholder Text WorldCat 175. Zivkovic AM , German JB, Lebrilla CB, Mills DA Human milk glycobiome and its impact on the infant gastrointestinal microbiota Proc Natl Acad Sci USA 2011 108 Suppl 1 4653 4658 Google Scholar OpenURL Placeholder Text WorldCat DP Degree of polymerization HMO Human milk oligosaccharide FOS Fructooligosaccharide GOS Galactooligosaccharides MOS Mannan-oligosaccharides SCFA Short-chain fatty acid XOS Xylooligosaccharides Publisher's Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. © Society for Industrial Microbiology 2019 This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model) © Society for Industrial Microbiology 2019 TI - Prebiotics: tools to manipulate the gut microbiome and metabolome JF - Journal of Industrial Microbiology and Biotechnology DO - 10.1007/s10295-019-02203-4 DA - 2019-10-01 UR - https://www.deepdyve.com/lp/oxford-university-press/prebiotics-tools-to-manipulate-the-gut-microbiome-and-metabolome-ExubVXtYsJ SP - 1445 EP - 1459 VL - 46 IS - 9-10 DP - DeepDyve ER -