1,957
Views
0
CrossRef citations to date
0
Altmetric
Research Paper

Tunable control of B. infantis abundance and gut metabolites by co-administration of human milk oligosaccharides

, , , , , , , & show all
Article: 2304160 | Received 19 Sep 2023, Accepted 08 Jan 2024, Published online: 18 Jan 2024

ABSTRACT

Precision engineering of the gut microbiome holds promise as an effective therapeutic approach for diseases associated with a disruption in this microbial community. Engrafting a live biotherapeutic product (LBP) in a predictable, controllable manner is key to the consistent success of this approach and has remained a challenge for most LBPs under development. We recently demonstrated high-level engraftment of Bifidobacterium longum subsp. infantis (B. infantis) in adults when co-dosed with a specific prebiotic, human milk oligosaccharides (HMO). Here, we present a cellular kinetic-pharmacodynamic approach, analogous to pharmacokinetic-pharmacodynamic-based analyses of small molecule- and biologic-based drugs, to establish how HMO controls expansion, abundance, and metabolic output of B. infantis in a human microbiota-based model in gnotobiotic mice. Our data demonstrate that the HMO dose controls steady-state abundance of B. infantis in the microbiome, and that B. infantis together with HMO impacts gut metabolite levels in a targeted, HMO-dependent manner. We also found that HMO creates a privileged niche for B. infantis expansion across a 5-log range of bacterial inocula. These results demonstrate remarkable control of both B. infantis levels and the microbiome community metabolic outputs using this synbiotic approach, and pave the way for precision engineering of desirable microbes and metabolites to treat a range of diseases.

Introduction

The use of live biotherapeutic products (LBPs) to manipulate gut microbiomes is a successful strategy for treating recurrent Clostridioides difficile infection (rCDI), as evidenced by the recent FDA approval of two different therapies, REBYOTATM (FDA STN 125,739) and VOWSTTM (FDA STN 125,757). LBPs, an emerging class of therapeutic microbes manufactured following rigorous safety standards,Citation1,Citation2 are also currently under development for other therapeutic uses including treatment of inflammatory bowel and graft-versus-host diseases (e.g. Clinical Trial IDs: NCT06102213, NCT05370885, NCT04995653), and as a co-therapeutic for cancer immunotherapies (Clinical Trial IDs: NCT05122546, NCT04988841, NCT05354102, NCT03686202). Several studies have demonstrated that therapeutic success for rCDI is dependent on stable engraftment of the delivered microbes.Citation3,Citation4 However, the determinants of successful engraftment are not well-definedCitation5–7 and many introduced bacteria do not stably engraft.Citation8–15 Furthermore, relative abundance of the engrafted microbes within the microbiome, regardless of how it has been measured and defined, may also influence efficacy.Citation3,Citation16

The beneficial impacts of therapeutic microbes are thought to be largely driven by modulation of host-impacting metabolites, which are known to be important in health and disease.Citation17,Citation18 Net metabolite output within the gastrointestinal tract is tied to microbial abundance and substrate availability. Inter-individual functional outputs of the microbiome, such as bile acid production and resistance to C. difficile colonization,Citation19,Citation20 organic acid production with a corresponding decreased fecal pH,Citation21 and cytokine responses,Citation22 have been tied to the abundance of specific beneficial bacterial species, many of which are under investigation as candidate LBPs. In humans, modulation of bile acids and short chain fatty acids (SCFAs) by a therapeutic consortium occurred specifically after multiple doses that led to high levels of engraftment.Citation23 In another human study, administration of indigestible starches altered levels of branched-chain fatty acids and SCFAs only at the highest doses of starch.Citation24

Given that abundance, and not just presence, of a therapeutic microbe appears to be a key factor impacting the levels of efficacy-mediating metabolites or molecules,Citation3,Citation4,Citation16 the design of novel LBPs should incorporate strategies to maximize efficacy by optimizing abundance. This concept is analogous to the pharmacokinetic and structure-activity relationship analyses central to medicinal chemistry efforts in which potency, bioavailability, and metabolism of small-molecule drugs is optimized. However, given that LBPs are live organisms and that they primarily act through gastrointestinal sites, the types of pharmacokinetic analyses used for tracking the absorption and metabolism of small molecules or biologics are not directly applicable and are not widely applied during LBP development.Citation25 Recently, advances in whole metagenome sequencing-based strain tracking methods and quantitative systems pharmacology modeling approaches have improved LBP abundance data, generating what has been termed “cellular kinetic-pharmacodynamic” (CK-PD) data,Citation3,Citation23,Citation26 which link dosing strategy, microbial abundance, and metabolite production. These types of analyses have the potential to inform optimal LBP dosing strategies, provide a framework for evaluating interactions of the LBP within the microbiome and metabolome, and ultimately facilitate beneficial impacts of an LBP on patient health.

We aimed to generate CK-PD data for a novel LBP, Bifidobacterium longum, subsp. infantis (B. infantis), an infant-associated microbe selected for its ability to proliferate in the microbiome by efficiently internalizing and metabolizing human milk oligosaccharides (HMO).Citation27–29 We recently reported engraftment of B. infantis into both unperturbed and antibiotic-treated gut microbiomes of healthy adult volunteers when co-dosed with a mixture of HMOCitation30,Citation31; dosing of the paired microbe and nutrient source is termed a synbiotic treatment. In unperturbed adults, fewer became engrafted with B. infantis when lower amounts of HMO were administered, suggesting that there may be a nutrient threshold for B. infantis engraftment.Citation30 Untargeted global metabolomics of fecal samples from unperturbed adults before and after treatment with B. infantis and HMO showed statistically significant changes in 305 metabolites, whereas in adults treated with HMO alone, only 86 metabolites were changed, suggesting that the synbiotic combination of B. infantis and HMO induces more consistent changes across treated individuals than does HMO alone.Citation30

In unperturbed adults treated with 18 g of HMO daily, B. infantis reached an abundance that averaged 5% but ranged from 0.004–25% across individuals.Citation30 In adults treated with antibiotics and each given the same dose of 18 g of HMO, B. infantis abundance ranged from 0.02–81%.Citation31 This strongly suggests that individual microbiome communities are an important variable impacting abundance. For antibiotic-treated adults, even though B. infantis abundance varied between individuals, consistent changes in a number of therapeutically relevant metabolites, including lactate, acetate, indoles, and tyrosine metabolites, and robust impacts on the microbiome, characterized by increases in the lactate-utilizing, short-chain fatty acid-producing genus Veillonella, were observed.Citation31 Antibiotic treatment has been suggested as a strategy to enhance engraftment and abundance of LBPs,Citation32 but it can also disrupt other taxa in the microbiota and compromise patient health,Citation26,Citation33,Citation34 making it undesirable in general practice. Therefore, maximizing levels of abundance through other means, such as by dosing as a synbiotic or by altering the dosing strategy, may be critical for therapeutic impact, and this demonstrates the relevance of CK-PD data to facilitate optimization of B. infantis abundance across individuals in our approach.

In this study, we defined the impact of different HMO and B. infantis dosing regimens on bacterial abundance and metabolite levels using gnotobiotic mice colonized with healthy, unperturbed human stool microbiota and a highly sensitive and rapid qPCR assay for B. infantis. We found that the abundance of B. infantis was dependent on the dose of HMO administered, and that B. infantis expansion occurred in the presence of HMO independent of either the starting bacterial inoculum levels or the number of bacterial doses. Additionally, we observed distinct impacts on metabolite levels across groups that were administered different doses of HMO, suggesting that B. infantis abundance influences metabolite output. Together, these data demonstrate that for the synbiotic pairing of HMO and B. infantis, HMO creates a privileged niche for B. infantis replication and enables tunable control of abundance, opening the door to precision engineering of microbes and metabolites for specific therapeutic applications and individualized treatment of patients.

Results

B. infantis steady-state abundance is dependent on the administered dose of HMO

We previously demonstrated that B. infantis colonizes at higher levels in healthy adults when they are also treated with antibiotics,Citation31 likely due to antibiotic-mediated clearing of microbiota niches. However, B. infantis is thought to proliferate in the microbiome due to a unique ability to metabolize HMO, thereby creating its own nutritional niche. We hypothesized that administration of different amounts of HMO would impact the size of the niche, and consequently the abundance of B. infantis. We therefore developed a gnotobiotic mouse model, in which germ-free mice were colonized with stool microbiota from a single human and maintained on a polysaccharide-free diet.Citation30 Mice colonized with stool from a healthy adult human were treated with B. infantis and a range of doses of HMO for three days, followed by continued dosing with HMO alone at different doses to support continued metabolism of B. infantis (). Fecal samples were analyzed for total bacterial abundance, as measured by 16S rRNA gene qPCR, and for B. infantis abundance using a subspecies-specific qPCR assay.Citation35 Total bacterial abundance did not significantly differ across the treatments (; no significant difference using a mixed effects model for repeated measures of log-transformed data), although it trended numerically lower at certain timepoints.

Figure 1. HMO dose determines steady-state abundance of B. infantis. (a) Gnotobiotic mice were colonized with healthy human adult stool #R01 and allowed to acclimate for 1 week, prior to treatment with 1 × 108 CFU B. infantis and different amounts of HMO twice daily. Control arms included B. infantis treatment with vehicle (no HMO) and 32 mg HMO per dose without B. infantis. (b,c) 16S gene copy number, and B. infantis abundance in fecal pellets relative to total 16S gene copy number, were determined using qPCR. For samples with B. infantis signal at or below the limit of detection, values were imputed as one-half the lowest observed abundance. Data are from two independent experiments carried out under the same conditions. Data are geometric mean and geometric standard deviation, n = 4–8 per group. Significance between groups was calculated using a mixed effects model for repeated measures of log-transformed data with the Bonferroni-Sidak correction for multiple comparisons; asterisks represent statistically significant differences in the time versus treatment interaction factor for the indicated pair of conditions. (d) Individual traces for each mouse. (e) Steady state B. infantis abundance for groups receiving 1 mg HMO or higher per dose, determined as the geometric mean of values for days 12, 14, and 17 for each mouse in the indicated HMO dose groups. p-values were calculated for log-transformed data using one-way ANOVA.

Figure 1. HMO dose determines steady-state abundance of B. infantis. (a) Gnotobiotic mice were colonized with healthy human adult stool #R01 and allowed to acclimate for 1 week, prior to treatment with 1 × 108 CFU B. infantis and different amounts of HMO twice daily. Control arms included B. infantis treatment with vehicle (no HMO) and 32 mg HMO per dose without B. infantis. (b,c) 16S gene copy number, and B. infantis abundance in fecal pellets relative to total 16S gene copy number, were determined using qPCR. For samples with B. infantis signal at or below the limit of detection, values were imputed as one-half the lowest observed abundance. Data are from two independent experiments carried out under the same conditions. Data are geometric mean and geometric standard deviation, n = 4–8 per group. Significance between groups was calculated using a mixed effects model for repeated measures of log-transformed data with the Bonferroni-Sidak correction for multiple comparisons; asterisks represent statistically significant differences in the time versus treatment interaction factor for the indicated pair of conditions. (d) Individual traces for each mouse. (e) Steady state B. infantis abundance for groups receiving 1 mg HMO or higher per dose, determined as the geometric mean of values for days 12, 14, and 17 for each mouse in the indicated HMO dose groups. p-values were calculated for log-transformed data using one-way ANOVA.

During the initial phase of the experiment, when mice were actively dosed with B. infantis, abundance was not statistically different across all groups regardless of HMO dose, including control animals that did not receive HMO (, one-way ANOVA). After bacterial dosing ended, B. infantis abundance numerically declined to below the limit of detection in groups receiving 2 mg or less of HMO per dose, with kinetics not significantly different from those of animals receiving no HMO, suggesting that this was below the threshold necessary to enable HMO-dependent B. infantis engraftment. At intermediate doses of HMO (4 and 8 mg), B. infantis abundance declined for approximately 5 days, at a rate numerically slower than with no HMO, and plateaued at an abundance of 0.00008% and 0.0004% respectively (average across the last three timepoints). With either 12 or 16 mg of HMO per dose, B. infantis abundance did not decline, and plateaued at approximately 0.06%, a level slightly above that observed during the initial bacterial dosing period (0.015% across all mice); no significant difference was observed between these doses. In contrast, over the course of a week, abundance in mice receiving 32 mg of HMO per dose increased 200-fold, plateauing at approximately 3% of the microbiome for an additional week, and was significantly different from the 16 mg dose. Over the last week of the study, when abundance appeared to have plateaued in all groups, we observed that B. infantis increased as the amount of dosed HMO increased (). These results suggest that tuning of B. infantis abundance within a particular microbiome community may be achievable by tuning the dose of HMO.

B. infantis abundance, not just presence, influences the metabolite profile

We and others have previously demonstrated that B. infantis produces acetate and lactate via fermentation of carbon sources and produces indole-3-lactate from tryptophan.Citation30,Citation31,Citation36 We hypothesized that higher levels of B. infantis abundance would lead to higher production of these metabolites, which could be quantified in cecal contents (). Levels of lactate, acetate, and indole-3-lactate were significantly higher in mice receiving B. infantis in combination with the highest dose of HMO (32 mg) compared to mice that did not receive HMO (). Significant increases in acetate and indole-3-lactate were also observed at the second highest HMO dose (16 mg), and lactate was numerically increased, although this change was not statistically significant. B. infantis-produced metabolites were numerically, but not significantly, increased in mice receiving intermediate doses of HMO (12 mg and 8 mg). This observation is particularly surprising for the group that received 12 mg, as the abundance of B. infantis was similar to that observed for the 16 mg HMO group. One possible explanation is that the number of animals in this group (n = 4) was not sufficient to detect statistically significant differences.

Figure 2. Metabolite analysis of cecal contents from mice treated with B. infantis and different doses of HMO. The indicated metabolites were quantified in cecal contents. (a) Heatmaps displaying the geometric mean of the metabolite quantification for each HMO dose group on day 17, log-transformed and normalized to that of the group receiving 0 mg HMO. Asterisks represent p-values compared to the 0 mg HMO group calculated using one-way ANOVA with Sidak’s posttest. (b) Geometric mean and geometric standard deviation of metabolites in cecal contents, and p-values between the indicated dose groups calculated by one-way ANOVA on all groups with Sidak’s multiple comparison test.

Figure 2. Metabolite analysis of cecal contents from mice treated with B. infantis and different doses of HMO. The indicated metabolites were quantified in cecal contents. (a) Heatmaps displaying the geometric mean of the metabolite quantification for each HMO dose group on day 17, log-transformed and normalized to that of the group receiving 0 mg HMO. Asterisks represent p-values compared to the 0 mg HMO group calculated using one-way ANOVA with Sidak’s posttest. (b) Geometric mean and geometric standard deviation of metabolites in cecal contents, and p-values between the indicated dose groups calculated by one-way ANOVA on all groups with Sidak’s multiple comparison test.

Significant changes in lactate and acetate, but not indole-3-lactate, were also observed upon treatment with 32 mg HMO alone compared to 0 mg HMO (). When we compared groups receiving 32 mg HMO with and without B. infantis (), we found significant differences for lactate and indole-3-lactate, supporting the idea that B. infantis robustly and specifically produces these metabolites, but not for acetate, suggesting that other members of this particular healthy microbiome utilize HMO to produce acetate.

Lactate and acetate production enables crossfeeding of other microbiome members that consequently produce propionate and butyrate.Citation30,Citation31 Similarly, we observed increased propionate in the B. infantis and HMO treatment group relative to no HMO treatment (). Treatment with HMO alone also significantly increased propionate compared to no HMO (), and the level of propionate was significantly higher for treatment with HMO alone than treatment with HMO and B. infantis (). Treatment with HMO and B. infantis did not impact levels of butyrate, but treatment with HMO alone did significantly increase butyrate levels (), although interestingly there was no statistically significant difference in butyrate between groups treated with HMO with or without B. infantis ().

We also quantified the levels of indole-3-propionate, a metabolite that can be produced from indole-3-lactate by certain microbes and which may have host-beneficial impacts.Citation37,Citation38 Indole-3-propionate was significantly increased by treatment with B. infantis and either 16 or 32 mg HMO, and was also increased during treatment with 32 mg HMO without B. infantis (), although to a statistically significantly lower extent than with B. infantis ().

B. infantis expansion rate is similar regardless of delivered inoculum or number of bacterial doses

Repeated dosing or varying the inoculum of LBPs has been suggested to increase the frequency of individuals that become engrafted, the abundance of the delivered microbes, and therapeutic efficacy for rCDI,Citation3,Citation16,Citation23,Citation32 though one study found no impact of repeated dosing.Citation39 We therefore next employed our gnotobiotic model to evaluate the kinetics of B. infantis expansion while varying the initial inoculum or the total number of doses of bacteria administered (). Twice-daily doses of B. infantis () ranging from 1 × 103 to 1 × 108 CFU were administered for either 1 day (2 total doses) or 3 days (6 total doses). In one study, HMO or vehicle dosing continued for a total of 14 days; in a second study, HMO dosing ceased after 10 days but sampling continued until 14 days. Total bacterial abundance, as measured by 16S rRNA gene qPCR, was not statistically significantly different by treatment (; no significant difference using a mixed effects model for repeated measures of log-transformed data).

Figure 3. B. infantis expands at a similar rate regardless of starting inoculum level. (a) Gnotobiotic mice colonized with healthy human adult stool #R02 were treated with the indicated CFU of B. infantis along with HMO (16 mg) or PBS twice daily. (b,c,d) 16S gene copy number, and B. infantis abundance in DNA extracted from fecal pellets relative to total 16S gene copy number, were determined using qPCR. Data are geometric mean and geometric standard deviation for mice receiving B. infantis with HMO (b,c) or with PBS (b,d); n = 8 for groups dosed over days 0–2 from two independent experiments (solid line HMO; dotted line PBS); n = 4 for groups dosed only on day 0 in a single experiment (hatched line HMO). For samples with B. infantis signal at or below the limit of detection, values were imputed as one-half the lowest observed abundance. Significance between groups was calculated using a mixed effects model for repeated measures of log-transformed data with the Bonferroni-Sidak correction for multiple comparisons; asterisks represent statistically significant differences in the time versus treatment interaction factor for the indicated pair of conditions. (e,f) Individual traces for each mouse dosed over days 0–2 from the data plotted in (c); 4 mice in each dose cohort continued receiving HMO to day 14 (e), and the other 4 received vehicle from days 11–14 (f). (g) Linear regression of log-transformed abundance data for all mice dosed over days 0–2 with HMO dosing through day 10. Timepoints for which B. infantis was below limit of detection in the 1 × 103 or 5 × 104 dose groups were omitted to improve regression. (h) The number of doublings of B. infantis per day for each group of mice was captured from the slope of the regression lines presented in (e). (i) Quantification of B. infantis inoculum for each dose in both studies.

Figure 3. B. infantis expands at a similar rate regardless of starting inoculum level. (a) Gnotobiotic mice colonized with healthy human adult stool #R02 were treated with the indicated CFU of B. infantis along with HMO (16 mg) or PBS twice daily. (b,c,d) 16S gene copy number, and B. infantis abundance in DNA extracted from fecal pellets relative to total 16S gene copy number, were determined using qPCR. Data are geometric mean and geometric standard deviation for mice receiving B. infantis with HMO (b,c) or with PBS (b,d); n = 8 for groups dosed over days 0–2 from two independent experiments (solid line HMO; dotted line PBS); n = 4 for groups dosed only on day 0 in a single experiment (hatched line HMO). For samples with B. infantis signal at or below the limit of detection, values were imputed as one-half the lowest observed abundance. Significance between groups was calculated using a mixed effects model for repeated measures of log-transformed data with the Bonferroni-Sidak correction for multiple comparisons; asterisks represent statistically significant differences in the time versus treatment interaction factor for the indicated pair of conditions. (e,f) Individual traces for each mouse dosed over days 0–2 from the data plotted in (c); 4 mice in each dose cohort continued receiving HMO to day 14 (e), and the other 4 received vehicle from days 11–14 (f). (g) Linear regression of log-transformed abundance data for all mice dosed over days 0–2 with HMO dosing through day 10. Timepoints for which B. infantis was below limit of detection in the 1 × 103 or 5 × 104 dose groups were omitted to improve regression. (h) The number of doublings of B. infantis per day for each group of mice was captured from the slope of the regression lines presented in (e). (i) Quantification of B. infantis inoculum for each dose in both studies.

As expected, the initial abundance of B. infantis during the bacterial dosing period varied according to the dose administered (). For mice receiving vehicle, B. infantis abundance numerically declined rapidly after the end of bacterial dosing (). However, with sustained administration of HMO, B. infantis abundance was maintained or increased in all mice, regardless of the bacterial dose administered or the initial abundance (). The two highest dose groups appeared to converge on a similar maximum abundance before the end of the study of approximately 1% (). In one experiment, B. infantis levels were monitored over time after cessation of HMO dosing and rapidly declined, as expected ().

We next performed regression analysis to determine the expansion rate of the B. infantis population (). Groups receiving 1 × 10,3 5 × 10,4 or 2 × 106 CFU B. infantis had similar doubling rates (; no statistically significant differences by one-way ANOVA with Tukey’s posttest for multiple comparisons), suggesting that rate of growth is independent of starting abundance. At the highest dose of 1 × 108 CFU per dose, it is possible that the measured expansion rate is masked by a high passage of B. infantis through the gut during dosing, and that expansion slows when B. infantis approaches carrying capacity in the gut, perhaps due to a limited supply of HMO or other forms of inter-species competition. Indeed, the groups receiving 2 × 106 CFU per dose also appear to have a slower rate of expansion after day 10.

We also examined the impact of repeated dosing with B. infantis. For the three highest dose groups, there were no statistically significant differences in the expansion curve between same-dose paired groups receiving 2 total doses or 6 total doses (; mixed effects model). However, in the groups receiving 1 × 103 CFU per dose, the group that received 6 total doses displayed a significant, 2-day reduction in the time to achieve comparable abundance with the group that received 2 total doses, although there was no apparent difference in the rate of expansion.

Abundance of B. infantis varies in different healthy stools treated with the same dose of HMO

As shown in , in mice colonized with stool #R01 and treated with B. infantis and 16 mg HMO, B. infantis abundance plateaued at 0.065% (average of the last three timepoints). As shown in , mice colonized with stool #R02, sourced from a different healthy adult, and treated with the same B. infantis dose and HMO dose of 16 mg, B. infantis abundance exceeded 1% (). This comparison aligns with our previous observations in humans, where we observed steady-state abundances of B. infantis ranging from 0.004–81% in different engrafted individuals.Citation30,Citation31

Discussion

Defining the effects of dosing permutations on engraftment of LBPs has been a suggested alternative for traditional pharmacokinetic (PK) studies,Citation23 and has the potential to inform dosing strategiesCitation26 that may improve efficacy and decrease cost. Here we report two kinds of CK-PD studies, in which we tested the effect of varying the dose of each component of a synbiotic product, comprising B. infantis and its preferred carbon source HMO, on abundance and metabolite levels in gnotobiotic mice harboring a healthy human microbiota. We found that altering the dose of HMO led to corresponding changes in the abundance of B. infantis at steady state, and that the levels of lactate, acetate, propionate, indole-3-lactate, and indole-3-propionate were only altered at the highest doses of HMO tested. This synbiotic pairing of LBP and prebiotic substrate is expected to enable highly reproducible impacts on the microbiome and gut metabolites. Indeed, we previously observed consistent microbiome and metabolite changes (e.g. expansion of Veillonella due to crossfeeding by B. infantis-produced lactate) across multiple antibiotic-treated adult humans harboring high levels of B. infantis during treatment with HMO.Citation31 Here, we also found that altering the B. infantis inoculum did not change B. infantis expansion rate in the microbiome (1–1.2 doublings per day) but did affect the time for B. infantis to reach steady-state abundance. Our data are consistent with the idea that HMO creates a privileged niche for B. infantis and that B. infantis expansion in the microbiome is limited primarily by access to carbon. Indeed, B. infantis is known to outcompete other microbes for use of HMO by selfishly importing HMOs and retaining degradation products intracellularly.Citation27–29

We found a 15-fold difference in B. infantis abundance across the two different donor stools used in our studies, when the same B. infantis and HMO dosing conditions were compared. This result suggests that the composition of a particular microbiota community impacts the size of the niche available to B. infantis, as we have previously observed in humans.Citation30,Citation31 Although germ-free mice colonized with human stool microbiota are widely used as a model, it is important to note that retention of inoculated taxa varies widely across different inocula, and the distribution of retained taxa is generally distinct from that observed in the inoculumCitation40–48 due to physiological and sample handling factors; for example, differences in mouse and human gastrointestinal anatomy and diet, and storage time/temperature/oxygen exposure of fresh stool prior to cryopreservation. Additionally, microbial composition of the stool inoculum is typically determined via DNA sequencing, which detects DNA from both viable and non-viable cells and complicates measurements of retention of inoculated taxa. Despite these limitations of using germ-free mice to model human microbiota communities, we have previously observed microbiome-mediated recapitulation of human colonization phenotypes using our mouse model,Citation30 which supports the idea that the impacts of synbiotic dosing strategies observed here are likely to also apply to other communities in humans and mice.

Our synbiotic strategy enables LBP engraftment by using a prebiotic substrate to create a privileged nutritional niche for B. infantis in the absence of antibiotic pretreatment.Citation30 Successful engraftment remains a key challenge for LBPs, as it only occurs in a subset of human subjects,Citation3,Citation6,Citation11,Citation15,Citation16,Citation23,Citation32,Citation49 likely due to competitive exclusion by related microbes or limited resource availability.Citation6 Additionally, LBPs comprising a consortium of microbes may require stepwise cooperation, where the success of initial colonizers is required to enable engraftment of all strains, and this has been demonstrated in mouse models.Citation50 To achieve engraftment in the absence of antibiotic conditioning, altering the bacterial dose regimen, by either increasing the dose or dosing repeatedly, has been suggested to increase the frequency of individuals that become engrafted and increase therapeutic efficacy for rCDI,Citation3,Citation16,Citation23,Citation32 though one study found no impact of repeated dosing.Citation39 Here, we found that, in the presence of HMO, doses of B. infantis as low as 2 × 103 total CFU were sufficient to enable expansion and engraftment. Our synbiotic strategy not only enables engraftment of B. infantis, but also allows for precise control of abundance by co-administration with different doses of HMO.

Other microbiome modulation strategies that rely solely on prebiotics may promote the expansion of particular taxa in humans and in mouse models, sometimes in a dose-dependent manner.Citation24,Citation30,Citation51–57 Similarly, we observed increased production of acetate, propionate, and butyrate in mice treated with HMO alone, without B. infantis, suggesting that other members of the particular healthy microbiome used in this study may utilize HMO. Other members of genus Bifidobacterium, as well as members of Lactobacillus and Bacteroides, are known to secrete HMO-degrading enzymes, making monosaccharides available for consumption.Citation58–61 However, our previously published human data indicates that dosing with HMO alone did not lead to consistent, global changes in metabolite profiles across individuals, while dosing with B. infantis and HMO together did.Citation30 The number of significantly changed metabolites in adults receiving B. infantis and HMO together (305) compared with the number observed in adults receiving HMO only (86) suggest that the impact of HMO alone is not consistently strong across individual microbiomes. Indeed, prebiotics as sole agents are reliant on the preexistence of responding microbes, the presence of which likely varies to a high degree across individuals. Prebiotics useable by multiple species of bacteria are likely to have less impact on the abundance of any particular taxa and therefore minimal therapeutic relevance.Citation24,Citation62–64 In contrast, highly targeted prebiotics support the expansion of only the microbes able to utilize that substrate.Citation63,Citation65,Citation66 For our synbiotic of HMO and B. infantis, there was a strong effect of HMO dose on B. infantis abundance and metabolites, suggesting targeted expansion of this species. This strategy enables exquisite control of B. infantis engraftment, because not only did dosing with HMO maintain B. infantis in the microbiome, but withdrawal of HMO led to B. infantis clearance.

In addition to supporting an increase in abundance of B. infantis, HMO supports B. infantis-produced metabolic products that cross-feed other bacteria in the microbiome, resulting in additional beneficial metabolites. Given that metabolites are key mediators through which LBPs are expected to impact the host, it is important to understand the relationship between LBP abundance and the desired metabolomic and therapeutic outcomes. In mice, high levels of particular taxa induced via administration of high doses of prebiotics have been shown to reduce measures of diversity and abundance of other potentially beneficial taxa,Citation24,Citation56,Citation57 raising the question of whether maximizing LBP abundance is the best therapeutic strategy or whether tuning LBP abundance to produce ideal levels of metabolites would be a better approach.

For B. infantis, the CK-PD data collected in this study support the idea that HMO dosing enables tunable control of abundance and metabolite levels within the microbiome. This feature could facilitate precision engineering of metabolites and microbes to create a microbiome profile for specific therapeutic applications. HMO-enabled tuning of B. infantis abundance may also allow for personalized dosing strategies to achieve optimal impact for individual patients harboring distinct microbiome communities or needing particular metabolite profiles. More broadly, we anticipate that CK-PD analyses have the potential to impact design of LBPs and improve the clinical application of microbiome-based therapeutics.

Methods

Bacterial culture

B. infantis PBI001 was propagated for mouse studies in Lactobacilli MRS Broth (BD Difco # 288130) or in media containing 20 g/L pea peptone, 5 g/L yeast extract, 2 g/L potassium phosphate dibasic, 5 g/L sodium acetate, 1 g/L sodium chloride, 20 g/L dextrose, and 0.1% polysorbate-80. Cultures were centrifuged, and cell pellets were resuspended in phosphate-buffered saline (PBS) with glycerol at a final concentration of 15%, aliquoted, and frozen at −80°C for long term storage. Viable colony-forming units (CFU) were determined by diluting and plating onto Brucella Blood Agar (Anaerobe Systems) and growing anaerobically at 37°C. In studies delivering a range of B. infantis doses, cryovials were thawed, diluted in 15% glycerol, and refrozen prior to thawing and use in animal studies.

Preparation of fecal slurries

Healthy adult stool was sourced from two self-reported healthy adults with a body mass index < 25 and no history of antibiotic use in the three months prior to collection. Stools were frozen within 4 hours of production. The stools were collected and processed using methods designed to remove food particulates while maximizing recovery of viable bacteria. To prepare stool slurries, frozen stools were anaerobically thawed, weighed, and combined with sterile PBS. Stool #R01 was simultaneously homogenized and large particulates were removed using a BagMixer 400 (Interscience) with a BagPage XR (280 µm porosity) filter bag. Stool #R02 was homogenized in a blender and centrifuged at 200 × g for two minutes to remove large particulates. The two methods for homogenizing and removing particulates are both recognized as appropriate methods for stool processing.Citation67–71 The resulting filtrate or supernatant was combined with glycerol in PBS for a final concentration of 15% glycerol (w/w). The stool slurries (8–10% stool w/w) were aliquoted and frozen at −80°C for experimental use. Viable CFU in aliquots of stool were determined by diluting and plating onto Brucella Blood Agar (Anaerobe Systems AS-141) and growing aerobically and anaerobically at 37°C.

Preparation of HMO concentrate

The HMO concentrate used in this study was manufactured by Prolacta Bioscience (Duarte, CA). Prolacta Bioscience makes human milk nutritional products from donated breast milk, intended for use by extremely premature infants in the neonatal intensive care unit and by term infants born with congenital malformations requiring immediate surgery. The milk is obtained from healthy donors who produce excess milk beyond the needs of their own infants. The donors are carefully screened for a variety of infectious diseases and lifestyle elements, such as drug use including nicotine, similar to what occurs in a blood bank. All potential donors provide attestation from their baby’s pediatrician that the baby will not be adversely affected if the mother donates her excess breast milk. Individual milk donations are also screened for a variety of pathogens of concern. All donors sign informed consent as part of their contract with Prolacta prior to donation and are aware that a portion of their milk may be used for the development of new products or other research purposes. Multiple lots of pooled human milk, from up to 200 donors per lot, were used to prepare the HMO product. Human milk permeate, a co-product of ultrafiltration used during the manufacture of commercial human milk-based human milk fortifier and ready-to-feed products, consists primarily of water, lactose, HMO and minerals. HMO concentrate was generated by subsequently removing lactose from the permeate and carrying out further ultrafiltration and pasteurization steps. The test product was supplied as a frozen liquid, stored at ≤-20°C, and thawed prior to administration.

Murine studies

Germ-free female C57BL/6 mice (age 5–6 weeks, Taconic) were divided into individual cages and inoculated with 0.1 mL of a 5% fecal slurry from a healthy human and maintained on a polysaccharide-deficient diet (Bio-serv, F5805–1, AIN-93 G, 68% glucose, irradiated 40–55 kGy).Citation72,Citation73 After a 7-day stabilization period, animals were randomized based on the original group cage and orally gavaged with B. infantis PBI001 resuspended in 0.2 mL of either HMO or sterile phosphate buffered saline (PBS) every 10–14 hours over a period of one or three days, followed by administration of HMO or PBS twice daily until study end. Dosage of B. infantis and HMO for each study is indicated in the figures. Groups receiving different doses of B. infantis or HMO were dosed in ascending dosages. Fecal samples were collected from the bottom of the cage for each individual animal throughout the study and flash frozen for DNA extraction. At the end of the study, cecal contents were collected and snap-frozen for metabolite analysis. Individuals handling animals and samples were blinded to the dose of HMO or B. infantis being administered.

Animals were housed in a positive-pressure clean room with HEPA filtration (bioBubble) in solid-bottom micro-isolator cages (Innovive Innocage MVX6 with M-feed food hoppers), given sterile water ad libitum (Innovive Aquavive M-WB-300A), and provided with sterile 1/8” corn cob bedding (Teklad 7902). Environmental conditions were maintained at temperature 20–26°C, humidity 30–70%, with a 12/12 hour light/dark cycle. Health checks were performed at least twice daily and included observation of behavior and appearance. The number of animals per group was determined to be sufficient to detect changes in B. infantis abundance as small as 4.1-fold (#R01) or 2.7-fold (#R02) with a significance level of 0.05 with 80% power based on the test for the ratio of two means [log normal data])Citation74 using B. infantis abundance data from pilot studies in germ-free mice colonized with stools #R01 (coefficient of variation = 0.65) or #R02 (coefficient of variation = 0.27). Animal work was carried out in accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. All protocols were approved by the TransPharm PreClinical Solutions Institutional Animal Care and Use Committee.

DNA extraction from fecal samples and B. infantis-specific and 16S rRNA gene qPCR

Genomic DNA extraction from fecal pellets was performed using the ZymoBiomics 96 MagBead DNA Kit (Zymo Research, Inc), following the manufacturer’s instructions. The concentration of the extracted DNA was measured using a Nanodrop 8000 (Thermo Scientific). To quantify B. infantis or total bacterial copy number in DNA samples, qPCR was performed using 2 µl undiluted gDNA with primers specific to a B. infantis sialidase gene (5’-ATACAGCAGAACCTTGGCCT-3’; 5’-GCGATCACATGGACGAGAAC-3’; 5’-FAM/TTTCACGGA/ZEN/TCAC CGG ACCATACG-3IABkFQ), as previously described,Citation35 or using 3 ng gDNA as template with primers targeted to the 16S rRNA gene (5’-CGGTGAATACGTTCCCGG-3’; 5’-TACGGCTACCTTGTTACGACT T3’),Citation75 respectively.

Metabolite analysis of mouse cecal contents

Metabolite analysis was performed by Precion, Inc. (Morrisville, NC). Snap-frozen mouse cecal contents were thawed and 20–50 mg of material was transferred into a 2 mL cryotube containing three stainless steel 1/8” cone balls, and the exact weight of the sample was recorded. A solution of deuterium-labeled internal standards in water (50 μL) and 1.5 mL of methanol was added to the cryotube and the sample was homogenized by vortex mixing for 2 minutes. The resulting suspensions were centrifuged at 2,000 × g at 20°C for 10 minutes. Aliquots of the supernatants were transferred to a 96-well plate and derivatized using modified versions of published protocols (50 μL)Citation76 or (20 μL),Citation77 followed by liquid-chromatography mass spectrometry using an Exion UHPLC (Sciex) coupled to a 5500+ Triple Quadrupole Mass Spectrometer (Sciex) respectively in negative mode for lactate, acetate, butyrate, and propionate or in polarity switching mode for indole-3-lactate and indole-3-propionate. The peak areas of the respective parent to product ion transitions were measured against the peak area of the parent to product ion transitions of the corresponding labeled internal standards for the quantitated metabolites. Quantitation was performed with Sciex OS-MQ software (Sciex) based on fortified calibration standards prepared immediately prior to each run. Data were normalized to wet weight based on the exact wet weight of each sample.

Statistical analysis

Regressions and statistical analyses were performed using GraphPad Prism version 9.3.1 or higher for Windows, GraphPad Software, San Diego, California USA, www.graphpad,com. Significance values are noted as follows unless otherwise defined in figure legends: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Author contributions

ALR, JEB, GJM designed experiments. CMC developed methods. ALR, RS, OT performed experiments. ALR, SLB, and MLL analyzed data. ALR wrote the paper. AKS provided materials.

Acknowledgments

We would like to thank Shuning Rook Zheng, Richard Lavin, Aislinn Rowan-Nash, Chloe Autran, David Rechtman, Biranchi Patra, Shao-Hung Fred Tsen, Huiyu Jannela Xia, Kim Thu Tran, Tin Huynh, Thanhvan Vanessa Nguyen, Anita Wong, Jorge Ramirez, Gildardo Inzunza, Yesenia Martinez, and Mike Garelick for their support of and contributions to this study.

Data availability statement

The authors confirm that the data supporting the findings of this study are available within the article.

Disclosure statement

Prolacta Bioscience employees are also shareholders in the company. U.S. Patent No. 8,927,027, U.S. Patent Application No. 20200054035, International Application Pub. Nos. WO 2021/061991 and WO 2022/036225, and their corresponding family member patents and applications relate to aspects of this work.

Correction Statement

This article has been corrected with minor changes. These changes do not impact the academic content of the article.

Additional information

Funding

This work was funded by Prolacta Bioscience.

References

  • U.S. Food and Drug Administration, Center for Biologics Evaluation and Research. Early clinical trials with live biotherapeutic products: chemistry, manufacturing, and Control Information; Guidance for Industry. Silver Spring, MD; 2016. https://www.regulations.gov/docket/FDA-2010-D-0500.
  • Cordaillat-Simmons M, Rouanet A, Pot B. Live biotherapeutic products: the importance of a defined regulatory framework. Experimental & Molecular Medicine. 2020;52(9):1397–16. doi:10.1038/s12276-020-0437-6.
  • Aggarwala V, Mogno I, Li Z, Yang C, Britton GJ, Chen-Liaw A, Mitcham J, Bongers G, Gevers D, Clemente JC. et al. Precise quantification of bacterial strains after fecal microbiota transplantation delineates long-term engraftment and explains outcomes. Nat Microbiol. 2021;6(10):1309–1318. doi:10.1038/s41564-021-00966-0.
  • Feuerstadt P, Louie TJ, Lashner B, Wang EEL, Diao L, Bryant JA, Sims M, Kraft CS, Cohen SH, Berenson CS. et al. SER-109, an Oral Microbiome Therapy for Recurrent Clostridioides difficile Infection. N Engl J Med. 2022;386(3):220–229. doi:10.1056/NEJMoa2106516.
  • Danne C, Rolhion N, Sokol H. Recipient factors in faecal microbiota transplantation: one stool does not fit all. Nat Rev Gastroenterol Hepatol. 2021;18(7):503–513. doi:10.1038/s41575-021-00441-5.
  • Maldonado-Gómez MX, Martínez I, Bottacini F, O’Callaghan A, Ventura M, van Sinderen D, Hillmann B, Vangay P, Knights D, Hutkins RW. et al. Stable engraftment of Bifidobacterium longum AH1206 in the human gut depends on individualized features of the resident microbiome. Cell Host & Microbe. 2016;20(4):515–526. doi:10.1016/j.chom.2016.09.001.
  • Smillie CS, Sauk J, Gevers D, Friedman J, Sung J, Youngster I, Hohmann EL, Staley C, Khoruts A, Sadowsky MJ. et al. Strain tracking reveals the determinants of bacterial engraftment in the human gut following fecal microbiota transplantation. Cell Host & Microbe. 2018;23(2):229–240.e5. doi:10.1016/j.chom.2018.01.003.
  • Alander M, Mättö J, Kneifel W, Johansson M, Kögler B, Crittenden R, Mattila-Sandholm T, Saarela M. Effect of galacto-oligosaccharide supplementation on human faecal microflora and on survival and persistence of Bifidobacterium lactis Bb-12 in the gastrointestinal tract. Int Dairy J. 2001;11(10):817–825. doi:10.1016/S0958-6946(01)00100-5.
  • Charbonneau D, Gibb RD, Quigley EMM. Fecal excretion of Bifidobacterium infantis 35624 and changes in fecal microbiota after eight weeks of oral supplementation with encapsulated probiotic. Gut Microbes. 2013;4(3):201–211. doi:10.4161/gmic.24196.
  • Firmesse O, Mogenet A, Bresson J-L, Corthier G, Furet J-P. Lactobacillus rhamnosus R11 Consumed in a food supplement survived human digestive transit without modifying microbiota equilibrium as assessed by real-time polymerase chain reaction. J Mol Microbiol Biotechnol. 2008;14(1–3):90–99. doi:10.1159/000106087.
  • Frese SA, Hutkins RW, Walter J. Comparison of the colonization ability of autochthonous and allochthonous strains of lactobacilli in the human gastrointestinal tract. Adv Microbiol. 2012;2(03):399–409. doi:10.4236/aim.2012.23051.
  • Malinen E, Matto J, Salmitie M, Alander M, Saarela M, Palva A. PCR-ELISAII: analysis of Bifidobacterium populations in human faecal samples from a consumption trial with Bifidobacterium lactis bb-12 and a galacto-oligosaccharide preparation. Syst Appl Microbiol. 2002;25(2):249–258. doi:10.1016/S0723-2020(04)70109-5.
  • Rattanaprasert M, Roos S, Hutkins RW, Walter J. Quantitative evaluation of synbiotic strategies to improve persistence and metabolic activity of Lactobacillus reuteri DSM 17938 in the human gastrointestinal tract. J Funct Foods. 2014;10:85–94. doi:10.1016/j.jff.2014.05.017.
  • Rochet V, Rigottier-Gois L, Levenez F, Cadiou J, Marteau P, Bresson J-L, Goupil-Feillerat N, Doré J. Modulation of Lactobacillus casei in ileal and fecal samples from healthy volunteers after consumption of a fermented milk containing Lactobacillus casei DN-114 001 Rif. Can J Microbiol. 2008;54(8):660–667. doi:10.1139/W08-050.
  • Zmora N, Zilberman-Schapira G, Suez J, Mor U, Dori-Bachash M, Bashiardes S, Kotler E, Zur M, Regev-Lehavi D, Brik R-Z. et al. Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell. 2018;174(6):1388–1405.e21. doi:10.1016/j.cell.2018.08.041.
  • Louie T, Golan Y, Khanna S, Bobilev D, Erpelding N, Fratazzi C, Carini M, Menon R, Ruisi M, Norman JM. et al. VE303, a defined bacterial consortium, for prevention of recurrent Clostridioides difficile infection: a randomized clinical trial. JAMA. 2023;329(16):1356. doi:10.1001/jama.2023.4314.
  • Wong AC, Levy M. New approaches to microbiome-based therapies. mSystems. 2019;4:e00122–19. doi:10.1128/mSystems.00122-19.
  • Levy M, Blacher E, Elinav E. Microbiome, metabolites and host immunity. Curr Opin Microbiol. 2017;35:8–15. doi:10.1016/j.mib.2016.10.003.
  • Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, No D, Liu H, Kinnebrew M, Viale A. et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature. 2015;517(7533):205–208. doi:10.1038/nature13828.
  • Staley C, Kelly CR, Brandt LJ, Khoruts A, Sadowsky MJ, Blaser MJ. Complete microbiota engraftment is not essential for recovery from recurrent Clostridium difficile infection following fecal microbiota transplantation. mBio. 2016;7(6):e01965–16. doi:10.1128/mBio.01965-16.
  • Frese SA, Hutton AA, Contreras LN, Shaw CA, Palumbo MC, Casaburi G, Xu G, Davis JCC, Lebrilla CB, Henrick BM. et al. Persistence of supplemented Bifidobacterium longum subsp infantis EVC001 in breastfed infants. Infantis. 2017;2(6):e00501–17. doi:10.1128/mSphere.00501-17. EVC001 in Breastfed Infants. mSphere 2017.
  • Henrick BM, Rodriguez L, Lakshmikanth T, Pou C, Henckel E, Arzoomand A, Olin A, Wang J, Mikes J, Tan Z. et al. Bifidobacteria-mediated immune system imprinting early in life. Cell. 2021;184(15):3884–3898.e11. doi:10.1016/j.cell.2021.05.030.
  • Dsouza M, Menon R, Crossette E, Bhattarai SK, Schneider J, Kim Y-G, Reddy S, Caballero S, Felix C, Cornacchione L. et al. Colonization of the live biotherapeutic product VE303 and modulation of the microbiota and metabolites in healthy volunteers. Cell Host & Microbe. 2022;30(4):583–598.e8. doi:10.1016/j.chom.2022.03.016.
  • Deehan EC, Yang C, Perez-Muñoz ME, Nguyen NK, Cheng CC, Triador L, Zhang Z, Bakal JA, Walter J. Precision microbiome modulation with discrete dietary fiber structures directs short-chain fatty acid production. Cell Host & Microbe. 2020;27(3):389–404.e6. doi:10.1016/j.chom.2020.01.006.
  • Chopra T, Hecht G, Tillotson G. Gut microbiota and microbiota-based therapies for Clostridioides difficile infection. Front Med. 2023;9:1093329. doi:10.3389/fmed.2022.1093329.
  • Renardy M, Prokopienko AJ, Maxwell JR, Flusberg DA, Makaryan S, Selimkhanov J, Vakilynejad M, Subramanian K, Wille L. A quantitative systems pharmacology model describing the cellular kinetic‐pharmacodynamic relationship for a live biotherapeutic product to support microbiome drug development. Clin Pharma And Therapeutics. 2023;114(3):633–643. doi:10.1002/cpt.2952.
  • Sela DA, Chapman J, Adeuya A, Kim JH, Chen F, Whitehead TR, Lapidus A, Rokhsar DS, Lebrilla CB, German JB. et al. The genome sequence of Bifidobacterium longum subsp. infantis reveals adaptations for milk utilization within the infant microbiome. Proc Natl Acad Sci U S A. 2008;105(48):18964–18969. doi:10.1073/pnas.0809584105.
  • Sela DA, Garrido D, Lerno L, Wu S, Tan K, Eom H-J, Joachimiak A, Lebrilla CB, Mills DA. Bifidobacterium longum subsp. infantis ATCC 15697 α-fucosidases are active on fucosylated human milk oligosaccharides. Appl Environ Microbiol. 2012;78(3):795–803. doi:10.1128/AEM.06762-11.
  • Garrido D, Kim JH, German JB, Raybould HE, Mills DA, Uversky V. Oligosaccharide binding proteins from Bifidobacterium longum subsp. infantis reveal a preference for host glycans. PloS ONE. 2011;6(3):e17315. doi:10.1371/journal.pone.0017315.
  • Button JE, Autran CA, Reens AL, Cosetta CM, Smriga S, Ericson M, Pierce JV, Cook DN, Lee ML, Sun AK. et al. Dosing a synbiotic of human milk oligosaccharides and B. infantis leads to reversible engraftment in healthy adult microbiomes without antibiotics. Cell Host & Microbe. 2022;30(5):712–725.e7. doi:10.1016/j.chom.2022.04.001.
  • Button JE, Cosetta CM, Reens AL, Brooker SL, Rowan-Nask AD, Lavin RC, Saur R, Zheng S, Autran CA, Lee ML. et al. Precision modulation of dysbiotic microbiomes with a synbiotic of human milk sugars and B. infantis reshapes gut microbial composition and metabolites; [Manuscript in press]. Cell Host Microbe:2023p. 31.
  • Mocanu V, Rajaruban S, Dang J, Kung JY, Deehan EC, Madsen KL. Repeated fecal microbial transplantations and antibiotic pre-treatment are linked to improved clinical response and remission in inflammatory bowel disease: a systematic review and pooled proportion meta-analysis. JCM. 2021;10(5):959. doi:10.3390/jcm10050959.
  • Smith M, Dai A, Ghilardi G, Amelsberg KV, Devlin SM, Pajarillo R, Slingerland JB, Beghi S, Herrera PS, Giardina P. et al. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat Med. 2022;28(4):713–723. doi:10.1038/s41591-022-01702-9.
  • Peled JU, Gomes ALC, Devlin SM, Littmann ER, Taur Y, Sung AD, Weber D, Hashimoto D, Slingerland AE, Slingerland JB. et al. Microbiota as predictor of mortality in allogeneic hematopoietic-cell transplantation. N Engl J Med. 2020;382(9):822–834. doi:10.1056/NEJMoa1900623.
  • Lawley B, Munro K, Hughes A, Hodgkinson AJ, Prosser CG, Lowry D, Zhou SJ, Makrides M, Gibson RA, Lay C. et al. Differentiation of Bifidobacterium longum subspecies longum and infantis by quantitative PCR using functional gene targets. PeerJ. 2017;5:e3375. doi:10.7717/peerj.3375.
  • Laursen MF, Sakanaka M, von Burg N, Mörbe U, Andersen D, Moll JM, Pekmez CT, Rivollier A, Michaelsen KF, Mølgaard C. et al. Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut. Nat Microbiol. 2021;6(11):1367–1382. doi:10.1038/s41564-021-00970-4.
  • Flannigan KL, Nieves KM, Szczepanski HE, Serra A, Lee JW, Alston LA, Ramay H, Mani S, Hirota SA. The pregnane X receptor and indole-3-propionic acid shape the intestinal mesenchyme to restrain inflammation and fibrosis. Cell Mol Gastroenterol Hepatol. 2022;15(3):S765–795. doi:10.1016/j.jcmgh.2022.10.014.
  • Dodd D, Spitzer MH, Van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, Le A, Cowan TM, Nolan GP, Fischbach MA. et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551(7682):648–652. doi:10.1038/nature24661.
  • Dubberke ER, Lee CH, Orenstein R, Khanna S, Hecht G, Gerding DN. Results from a randomized, placebo-controlled clinical trial of a RBX2660—A microbiota-based drug for the prevention of recurrent Clostridium difficile infection. Clin Infect Dis. 2018;67(8):1198–1204. doi:10.1093/cid/ciy259.
  • Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med Available from. 2009;1(6). [[cited 2023 Nov 27]] doi:10.1126/scitranslmed.3000322.
  • Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, Beaumont M, Van Treuren W, Knight R, Bell JT. et al. Human genetics shape the gut microbiome. Cell. 2014;159(4):789–799. doi:10.1016/j.cell.2014.09.053.
  • Zhang L, Bahl MI, Roager HM, Fonvig CE, Hellgren LI, Frandsen HL, Pedersen O, Holm J-C, Hansen T, Licht TR. Environmental spread of microbes impacts the development of metabolic phenotypes in mice transplanted with microbial communities from humans. ISME J. 2017;11(3):676–690. doi:10.1038/ismej.2016.151.
  • Griffin NW, Ahern PP, Cheng J, Heath AC, Ilkayeva O, Newgard CB, Fontana L, Gordon JI. Prior dietary practices and connections to a human gut microbial metacommunity alter responses to diet interventions. Cell Host & Microbe. 2017;21(1):84–96. doi:10.1016/j.chom.2016.12.006.
  • Planer JD, Peng Y, Kau AL, Blanton LV, Ndao IM, Tarr PI, Warner BB, Gordon JI. Development of the gut microbiota and mucosal IgA responses in twins and gnotobiotic mice. Nature. 2016;534(7606):263–266. doi:10.1038/nature17940.
  • Van Den Ham KM, Little MR, Bednarski OJ, Fusco EM, Mandal RK, Mitra R, Li S, Doumbo S, Doumtabe D, Kayentao K. et al. Creation of a non-Western humanized gnotobiotic mouse model through the transplantation of rural African fecal microbiota. Microbiol Spectr. 2023;11(6):e01554–23. doi:10.1128/spectrum.01554-23.
  • Wang Y, Zhang Z, Liu B, Zhang C, Zhao J, Li X, Chen L. A study on the method and effect of the construction of a humanized mouse model of fecal microbiota transplantation. Front Microbiol. 2022;13:1031758. doi:10.3389/fmicb.2022.1031758.
  • Hutchison ER, Kasahara K, Zhang Q, Vivas EI, Cross T-W, Rey FE. Dissecting the impact of dietary fiber type on atherosclerosis in mice colonized with different gut microbial communities. NPJ Biofilms Microbio. 2023;9(1):31. doi:10.1038/s41522-023-00402-7.
  • Li Y, Cao W, Gao NL, Zhao X-M, Chen W-H. Consistent alterations of human fecal microbes after transplantation into germ-free mice. Genomics, Proteomics & Bioinformatics. 2022;20(2):382–393. doi:10.1016/j.gpb.2020.06.024.
  • Xiao Y, Zhao J, Zhang H, Zhai Q, Chen W. Mining Lactobacillus and Bifidobacterium for organisms with long-term gut colonization potential. Clin Nutr. 2020;39(5):1315–1323. doi:10.1016/j.clnu.2019.05.014.
  • Caballero S, Kim S, Carter RA, Leiner IM, Sušac B, Miller L, Kim GJ, Ling L, Pamer EG. Cooperating commensals restore colonization resistance to vancomycin-resistant Enterococcus faecium. Cell Host & Microbe. 2017;21(5):592–602.e4. doi:10.1016/j.chom.2017.04.002.
  • Davis LMG, Martínez I, Walter J, Hutkins R. A dose dependent impact of prebiotic galactooligosaccharides on the intestinal microbiota of healthy adults. Int J Food Microbiol. 2010;144(2):285–292. doi:10.1016/j.ijfoodmicro.2010.10.007.
  • Finegold SM, Li Z, Summanen PH, Downes J, Thames G, Corbett K, Dowd S, Krak M, Heber D. Xylooligosaccharide increases bifidobacteria but not lactobacilli in human gut microbiota. Food Funct. 2014;5(3):436. doi:10.1039/c3fo60348b.
  • Tandon D, Haque MM, Gote M, Jain M, Bhaduri A, Dubey AK, Mande SS. A prospective randomized, double-blind, placebo-controlled, dose-response relationship study to investigate efficacy of fructo-oligosaccharides (FOS) on human gut microflora. Sci Rep. 2019;9(1):5473. doi:10.1038/s41598-019-41837-3.
  • Costabile A, Deaville ER, Morales AM, Gibson GR, Riedel CU. Prebiotic potential of a maize-based soluble fibre and impact of dose on the human gut microbiota. PloS ONE. 2016;11(1):e0144457. doi:10.1371/journal.pone.0144457.
  • Tran TTT, Cousin FJ, Lynch DB, Menon R, Brulc J, Brown J-M, O’Herlihy E, Butto LF, Power K, Jeffery IB. et al. Prebiotic supplementation in frail older people affects specific gut microbiota taxa but not global diversity. Microbiome. 2019;7(1):39. doi:10.1186/s40168-019-0654-1.
  • Cui S, Gu J, Liu X, Li D, Mao B, Zhang H, Zhao J, Chen W. Lactulose significantly increased the relative abundance of Bifidobacterium and Blautia in mice feces as revealed by 16S rRNA amplicon sequencing. J Sci Food Agric. 2021;101(13):5721–5729. doi:10.1002/jsfa.11181.
  • Wang L, Hu L, Yan S, Jiang T, Fang S, Wang G, Zhao J, Zhang H, Chen W. Effects of different oligosaccharides at various dosages on the composition of gut microbiota and short-chain fatty acids in mice with constipation. Food Funct. 2017;8(5):1966–1978. doi:10.1039/C7FO00031F.
  • Asakuma S, Hatakeyama E, Urashima T, Yoshida E, Katayama T, Yamamoto K, Kumagai H, Ashida H, Hirose J, Kitaoka M. Physiology of consumption of human milk oligosaccharides by infant gut-associated bifidobacteria. J Biol Chem. 2011;286(40):34583–34592. doi:10.1074/jbc.M111.248138.
  • Marcobal A, Barboza M, Froehlich JW, Block DE, German JB, Lebrilla CB, Mills DA. Consumption of human milk oligosaccharides by gut-related microbes. J Agric Food Chem. 2010;58(9):5334–5340. doi:10.1021/jf9044205.
  • Bajic D, Wiens F, Wintergerst E, Deyaert S, Baudot A, Van Den Abbeele P. HMOs exert marked bifidogenic effects on children’s gut microbiota ex vivo, due to age-related Bifidobacterium species composition. Nutrients. 2023;15(7):1701. doi:10.3390/nu15071701.
  • Yu Z-T, Chen C, Newburg DS. Utilization of major fucosylated and sialylated human milk oligosaccharides by isolated human gut microbes. Glycobiology. 2013;23(11):1281–1292. doi:10.1093/glycob/cwt065.
  • Walter J, Maldonado-Gómez MX, Martínez I. To engraft or not to engraft: an ecological framework for gut microbiome modulation with live microbes. Curr Opin Biotechnol. 2018;49:129–139. doi:10.1016/j.copbio.2017.08.008.
  • Cantu-Jungles TM, Hamaker BR. Erratum for Cantu-Jungles and Hamaker, “New view on dietary fiber selection for predictable shifts in gut microbiota”. mBio. 2020;11(3):e02179–19. doi:10.1128/mBio.00747-20.
  • Patnode ML, Beller ZW, Han ND, Cheng J, Peters SL, Terrapon N, Henrissat B, Le Gall S, Saulnier L, Hayashi DK. et al. Interspecies competition impacts targeted manipulation of human gut bacteria by fiber-derived glycans. Cell. 2019;179(1):59–73.e13. doi:10.1016/j.cell.2019.08.011.
  • Shepherd ES, DeLoache WC, Pruss KM, Whitaker WR, Sonnenburg JL. An exclusive metabolic niche enables strain engraftment in the gut microbiota. Nature. 2018;557(7705):434–438. doi:10.1038/s41586-018-0092-4.
  • Cantu-Jungles TM, Bulut N, Chambry E, Ruthes A, Iacomini M, Keshavarzian A, Johnson TA, Hamaker BR, Zambrano MM. Dietary fiber hierarchical specificity: the missing link for predictable and strong shifts in gut bacterial communities. mBio. 2021;12(3):e01028–21. doi:10.1128/mBio.01028-21.
  • van Lingen E, Terveer EM, van der Meulen-de Jong AE, Vendrik KEW, Verspaget HW, Kuijper EJ, Kassam Z, Keller JJ. Advances in stool banking. Microbiota In Health And Disease [Internet]. [cited 2023 Nov 30]; 2020; 2. 10.26355/mhd_20201_182. Available from
  • Cui B, Li P, Xu L, Peng Z, Zhao Y, Wang H, He Z, Zhang T, Ji G, Wu K. et al. Fecal microbiota transplantation is an effective rescue therapy for refractory inflammatory bowel disease. Inflamm Cell Signal. 2015;2:e757.
  • Varga A, Kocsis B, Sipos D, Kása P, Vigvári S, Pál S, Dembrovszky F, Farkas K, Péterfi Z. How to apply FMT more effectively, conveniently and flexible – a comparison of FMT methods. Front Cell Infect Microbiol. 2021;11:657320. doi:10.3389/fcimb.2021.657320.
  • Halkjær SI, Christensen AH, BZS L, Browne PD, Günther S, Hansen LH, Petersen AM. Faecal microbiota transplantation alters gut microbiota in patients with irritable bowel syndrome: results from a randomised, double-blind placebo-controlled study. Gut. 2018;67(12):2107–2115. doi:10.1136/gutjnl-2018-316434.
  • Lee P-C, Chang T-E, Wang Y-P, Lee K-C, Lin Y-T, Chiou J-J, Huang C-W, Yang U-C, Li F-Y, Huang H-C. et al. Alteration of gut microbial composition associated with the therapeutic efficacy of fecal microbiota transplantation in Clostridium difficile infection. J Formos Med Assoc. 2022;121(9):1636–1646. doi:10.1016/j.jfma.2021.11.001.
  • Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, Fukuda S, Saito T, Narushima S, Hase K. et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500(7461):232–236. doi:10.1038/nature12331.
  • Marcobal A, Barboza M, Sonnenburg ED, Pudlo N, Martens EC, Desai P, Lebrilla CB, Weimer BC, Mills DA, German JB. et al. Bacteroides in the infant gut consume milk oligosaccharides via mucus-utilization pathways. Cell Host & Microbe. 2011;10(5):507–514. doi:10.1016/j.chom.2011.10.007.
  • NCSS. PASS 2023 power analysis and sample size software [Internet]. 2023; Available from: ncss.com/software/pass
  • Furet J-P, Firmesse O, Gourmelon M, Bridonneau C, Tap J, Mondot S, Doré J, Corthier G. Comparative assessment of human and farm animal faecal microbiota using real-time quantitative PCR: human and farm animal faecal microbiota. FEMS Microbiol Ecol. 2009;68(3):351–362. doi:10.1111/j.1574-6941.2009.00671.x.
  • Han J, Lin K, Sequeira C, Borchers CH. An isotope-labeled chemical derivatization method for the quantitation of short-chain fatty acids in human feces by liquid chromatography–tandem mass spectrometry. Anal Chim Acta. 2015;854:86–94. doi:10.1016/j.aca.2014.11.015.
  • Xu L, Spink DC. Analysis of steroidal estrogens as pyridine-3-sulfonyl derivatives by liquid chromatography electrospray tandem mass spectrometry. Anal Biochem. 2008;375(1):105–114. doi:10.1016/j.ab.2007.11.028.