2,494
Views
1
CrossRef citations to date
0
Altmetric
Research Paper

Cholestasis impairs gut microbiota development and bile salt hydrolase activity in preterm neonates

, , , , , , & ORCID Icon show all
Article: 2183690 | Received 28 Sep 2022, Accepted 16 Feb 2023, Published online: 26 Feb 2023

ABSTRACT

Cholestasis refers to impaired bile flow from the liver to the intestine. In neonates, cholestasis causes poor growth and may progress to liver failure and death. Normal bile flow requires an intact liver-gut-microbiome axis, whereby liver-derived primary bile acids are transformed into secondary bile acids. Microbial bile salt hydrolase (BSH) enzymes are responsible for the first step, deconjugating glycine- and taurine-conjugated primary bile acids. Cholestatic neonates often are treated with the potent choleretic bile acid ursodeoxycholic acid (UDCA), although interactions between UDCA, gut microbes, and other bile acids are poorly understood. To gain insight into how the liver-gut-microbiome axis develops in extreme prematurity and how cholestasis alters this maturation, we conducted a nested case-control study collecting 124 stool samples longitudinally from 24 preterm infants born at mean 27.2 ± 1.8 weeks gestation and 946 ± 249.6 g, half of whom developed physiologic cholestasis. Samples were analyzed by whole metagenomic sequencing, in vitro BSH enzyme activity assays optimized for low biomass fecal samples, and quantitative mass spectrometry to measure the bile acid metabolome. In extremely preterm neonates, acquisition of the secondary bile acid biosynthesis pathway and BSH genes carried by Clostridium perfringens are the most prominent features of early microbiome development. Cholestasis interrupts this developmental pattern. BSH gene abundance and enzyme activity are profoundly reduced in cholestatic neonates, resulting in decreased quantities of unconjugated bile acids. UDCA restores total fecal bile acid levels in cholestatic neonates, but this is due to a 522-fold increase in fecal UDCA. A majority of bile acids in early development are atypical positional and stereo-isomers of bile acids. We report novel associations linking isomeric bile acids and BSH activity to neonatal growth trajectories. These data highlight deconjugation of bile acids as a key microbial function that is acquired in early neonatal development and impaired by cholestasis.

Introduction

Preterm birth, a live birth occurring before 37 weeks of gestation, affects 11% of global births and contributes to more deaths in children under 5 years of age than any other cause.Citation1 Preterm infants are at high risk of sepsis, necrotizing enterocolitis (NEC), feeding difficulties, and liver diseaseCitation2–4 due to structural and functional immaturity of the gastrointestinal tract. Cholestasis, impaired bile flow from the liver to the intestine, affects 1:2,500 of all live births and 10–20% of preterm births.Citation5 Cholestasis can have multiple etiologies, and many cases (13–30%) are idiopathic.Citation6 Risk factors include prematurity, low birth weight, and parenteral nutrition.Citation7,Citation8 There are no specific treatments for neonatal cholestasis and most interventions do not address underlying causes.Citation9 Ursodeoxycholic acid (UDCA) often is administered to cholestatic infants to stimulate bile flow.Citation7 However, its effects are poorly understood, especially in the preterm intestine. Cholestasis can lead to poor neonatal growth and may progress to liver failure or death.Citation9 Thus, there is an urgent need to understand how cholestasis impacts the developing neonate, specifically extremely preterm infants who are at high risk of mortality.

Synthesized in the liver from cholesterol, bile acids provide the primary driving force for the production and secretion of bile, are critical for the digestion and absorption of dietary lipids,Citation10 and serve as key mediators of gut-liver communication. They act as signaling molecules and tightly regulate their pool size through an efficient enterohepatic circulation.Citation11 Bile acids affect the composition of the gut microbiome through bactericidal and bacteriostatic effects, and in turn, gut bacteria influence the bile acid pool.Citation12,Citation13 Microbial bile salt hydrolase (BSH) enzymes, present in many commensal microbes,Citation14 perform the first step in transforming liver-derived primary bile acids into secondary bile acids, which is hydrolysis of the amino acid moiety.Citation13 The gut microbiome influences the growth and development of preterm infants, and gestational age (GA) influences microbiome maturation.Citation15–17 There is a knowledge gap regarding how gut bacteria and bile acids interact over time and contribute to somatic growth in extremely premature neonates. Addressing this knowledge gap could provide new therapeutic opportunities to address the comorbidities that are associated with prematurity.

We aimed to test the hypotheses that: 1) The gut microbiome and bile acid composition of extremely premature neonates develop in a predictable manner over time; 2) Cholestasis interrupts this pattern of development; and 3) UDCA treatment quantitatively modifies bile acid and microbiota profiles. In order to test these hypotheses, neonates that developed cholestasis and controls that did not develop cholestasis were matched by GA and birth weight in a single-center nested case-control study. Stool samples were collected longitudinally from birth to hospital discharge. Whole metagenomic sequencing, quantitative mass spectrometry (MS), and a unique in vitro BSH enzymatic activity assay optimized for low microbial biomass samples revealed that cholestasis disrupts the development of the neonatal gut microbiome and impairs bile acid deconjugation. Furthermore, we identified novel associations between bile acid biomarkers and neonatal growth. These results suggest that acquisition of microbial bile acid deconjugation activity is a critical event in early neonatal development and that the liver-gut-microbiome axis could serve diagnostic or therapeutic roles to improve neonatal outcomes.

Results

Patient characteristics

Twenty-four preterm neonates were included in this nested case-control study. The mean GA for all infants was 27.2 ± 1.8 weeks gestation and the mean birth weight was 946 ± 249.6 g. Baseline clinical characteristics were similar between the 12 neonates that developed cholestasis (mean peak serum conjugated bilirubin 7.0 mg/dL) during their hospitalization and the 12 matched controls that did not develop cholestasis (). Neonates in both groups had similar patterns of antibiotic use over time (Supplementary Figure S4). Additionally, parameters describing the intrauterine environment prior to birth, including the presence of maternal sepsis, preterm premature rupture of membranes, and maternal antibiotic use, were not significantly different between groups. Birth mode did not significantly affect microbiota communities (Suplementary Figure S3). Timing of serum bilirubin testing, antibiotic, and UDCA use, and fecal sample collection are depicted in Supplementary Figures S1 and S2.

Table 1. Baseline demographics.

The extremely preterm neonatal gut microbiome develops over time

To determine how gut microbial communities of extremely preterm neonates develop over time, we performed whole metagenomic sequencing on the 64 stool samples collected from the 12 neonates that did not develop cholestasis (controls) from birth to 6 weeks of age. Consistent with previous reports,Citation18,Citation19 microbial community richness increased with postnatal age (, P = 0.056). Proteobacteria and Firmicutes were the most abundant phyla throughout early development (). Staphylococcus was the most prominent genus initially, but its abundance decreased over time. In contrast, Klebsiella gradually increased in abundance over the first month of postnatal life (). Individual microbiota proportions are depicted in Supplementary Figure S3.

Figure 1. Development of the preterm gut microbiome in control neonates. (a) Observed OTUs increase with age over the first 6 weeks of life (n = 18–25). (b) The gut microbiome changes at both phylum and genus levels in the first 6 weeks of life (n = 64). (c) At the species level, Clostridium perfringens is most significantly increased and Staphylococcus epidermidis and Gemella sanguinis are most significantly decreased with increasing PMA (n = 13–31). (d) LEfSe analysis identified secondary bile acid biosynthesis as the most enriched pathway in control neonates 32–40 weeks PMA relative to control neonates 25–28 weeks PMA (n = 20–31). (e) The abundance of the secondary bile acid biosynthesis pathway increases with PMA (n = 13–31). BA, bile acid; LEfSe, linear discriminate analysis effect size; OTU, operational taxonomic unit; PMA, post-menstrual age.

**** P<0.0001; *** P<0.001; ** P<0.01; * P<0.05.
Figure 1. Development of the preterm gut microbiome in control neonates. (a) Observed OTUs increase with age over the first 6 weeks of life (n = 18–25). (b) The gut microbiome changes at both phylum and genus levels in the first 6 weeks of life (n = 64). (c) At the species level, Clostridium perfringens is most significantly increased and Staphylococcus epidermidis and Gemella sanguinis are most significantly decreased with increasing PMA (n = 13–31). (d) LEfSe analysis identified secondary bile acid biosynthesis as the most enriched pathway in control neonates 32–40 weeks PMA relative to control neonates 25–28 weeks PMA (n = 20–31). (e) The abundance of the secondary bile acid biosynthesis pathway increases with PMA (n = 13–31). BA, bile acid; LEfSe, linear discriminate analysis effect size; OTU, operational taxonomic unit; PMA, post-menstrual age.

In addition to these known patterns of early postnatal gut microbiota maturation, we identified new longitudinal developmental patterns among control neonates based on postmenstrual age (PMA), which is the sum of postnatal age and GA at birth (Supplementary Figure S5). Among all species, Clostridium perfringens increased the most in relative abundance over time (FDR adjusted global P = 0.01, ). In contrast, the abundance of Gemella sanguinis and Staphylococcus epidermidis decreased with increasing PMA (FDR adjusted global P < 0.05, ).

To determine how microbial functional genomic potential develops with increasing PMA, we performed linear discriminant analysis effect size (LEfSe) pathway analysis of the metagenomic signatures. The most enriched microbial pathway in stool samples from the most mature (32–40 weeks PMA) relative to the least mature (25–28 weeks PMA) neonates was secondary bile acid biosynthesis (). The relative abundance of this pathway increased 28-fold from early to late neonatal development (). In contrast, stool from the least mature neonates was enriched in microbial genes that participate in valine, leucine, and isoleucine degradation; tryptophan metabolism; and Staphylococcus aureus infection. Together, these data indicate that microbiome profiles from extremely preterm neonates mature with increasing PMA, and that the most prominent developmental features are the acquisition of C. perfringens and the secondary bile acid biosynthesis pathway.

Cholestasis disrupts microbiome development and bile acid deconjugation

In order to determine whether these neonatal developmental patterns are interrupted by cholestasis, we plotted developmental trajectories of gut microbiome signatures from both control and cholestatic neonates using principal coordinate (PC) analysis. PC 1, which accounted for the greatest amount of variation in the composition of the gut microbiome, was strongly associated with PMA in the control cohort (, P < 0.0001). In contrast, PC 1 was not significantly correlated with increasing PMA among cholestatic neonates (, P = 0.32), suggesting that gut microbiome development is disrupted in preterm neonates with cholestasis.

Figure 2. Cholestasis disrupts microbiota maturation. (a) Position on PC Axis 1 increases linearly with increasing PMA in control neonates; this pattern is absent in cholestatic neonates (n = 49–64). (b) LEfSe analysis identified secondary bile acid biosynthesis as the most enriched pathway in control neonates compared to cholestatic neonates 32–40 weeks PMA (n = 13–17). Relative abundances of (c) the secondary bile acid biosynthesis pathway, (D) the BSH gene, and (e) the BSH-carrying bacterium Clostridium perfringens are reduced in cholestatic neonates at 32–40 weeks PMA (n = 13–17). (f) Schematic of BSH activity assay. (g) Glycine deconjugation is preferred over taurine deconjugation in stool from control neonates; BSH activity is reduced in stool from cholestatic neonates (n = 5–6). (h) Total fecal unconjugated bile acids are reduced in cholestatic neonates, although the quantity of fecal conjugated bile acids is unchanged between groups (n = 16–18). BA, bile acid; BSH, bile salt hydrolase; LDA, linear discriminate analysis; LEfSe, linear discriminate analysis effect size; PC, principal coordinate; PMA, post-menstrual age.

*** P<0.001; ** P<0.01; * P<0.05
Figure 2. Cholestasis disrupts microbiota maturation. (a) Position on PC Axis 1 increases linearly with increasing PMA in control neonates; this pattern is absent in cholestatic neonates (n = 49–64). (b) LEfSe analysis identified secondary bile acid biosynthesis as the most enriched pathway in control neonates compared to cholestatic neonates 32–40 weeks PMA (n = 13–17). Relative abundances of (c) the secondary bile acid biosynthesis pathway, (D) the BSH gene, and (e) the BSH-carrying bacterium Clostridium perfringens are reduced in cholestatic neonates at 32–40 weeks PMA (n = 13–17). (f) Schematic of BSH activity assay. (g) Glycine deconjugation is preferred over taurine deconjugation in stool from control neonates; BSH activity is reduced in stool from cholestatic neonates (n = 5–6). (h) Total fecal unconjugated bile acids are reduced in cholestatic neonates, although the quantity of fecal conjugated bile acids is unchanged between groups (n = 16–18). BA, bile acid; BSH, bile salt hydrolase; LDA, linear discriminate analysis; LEfSe, linear discriminate analysis effect size; PC, principal coordinate; PMA, post-menstrual age.

Next, we sought to identify the specific microbial genomic pathways that cholestatic neonates failed to acquire during early development. LEfSe analysis revealed that secondary bile acid biosynthesis was the most enriched pathway in stool from the control cohort relative to the cholestatic cohort at 32–40 weeks PMA (, P = 0.04). In accord with this finding, the BSH gene, which encodes the enzyme responsible for the initial step in the microbial transformation of primary into secondary bile acids, was reduced by 55% in cholestatic neonates (, P = 0.04). We next sought to determine which species of bacteria were responsible for acquisition of the BSH gene in the control group. Within our metagenomic dataset, we identified seven BSH-carrying bacterial species, of which only C. perfringens was depleted in stool from cholestatic neonates relative to controls (, P = 0.0008; Supplementary Figure S6). These data indicate that cholestasis interrupts the acquisition of C. perfringens, the BSH gene, and the potential to synthesize secondary bile acids, all of which are prominent features of gut microbiome development in our extremely preterm control cohort.

To determine whether delayed acquisition of the BSH gene translates to impaired deconjugation of primary bile acids in cholestatic neonates, we optimized a BSH enzyme activity assay for low microbial biomass stool samples by quantifying the nanomoles of glycine or taurine liberated from glycocholic or taurocholic acid, respectively (). Quantifying BSH enzyme activity in stool from control neonates revealed that glycine-conjugated cholic acid was metabolized at twice the rate of taurine-conjugated cholic acid (, blue shapes). Given that C. perfringens preferentially hydrolyzes glycine over taurine conjugates,Citation20,Citation21 this result supports the notion that C. perfringens is the primary BSH carrier in the control cohort. As expected, BSH enzyme activity was markedly decreased in stool from cholestatic neonates (P < 0.05, ). To obtain further evidence that BSH activity is impaired in cholestatic neonates, we quantified by MS all products of BSH activity, the total unconjugated bile acids, in stool from control and cholestatic neonates. Indeed, there was a median 86.4% reduction of unconjugated fecal bile acids in the cholestatic cohort (P = 0.0099, ). This result is in accord with both decreased microbial BSH activity and with decreased flow of bile into the intestine in cholestasis. Taken together, these data indicate that cholestatic neonates have delayed colonization of C. perfringens, delayed acquisition of the BSH gene, and markedly impaired capacity to deconjugate primary bile acids.

Fecal bile acid profiles mature over time and are altered by cholestasis

Based on our finding that acquisition of the capacity for secondary bile acid biosynthesis was the most prominent feature of gut microbiome development in extremely preterm neonates, we sought to determine whether fecal bile acid profiles mature in parallel during the same developmental period. Thus, we quantified concentrations of individual and total fecal bile acids from 124 samples using MS based on previously described methods with modifications.Citation22 At the earliest developmental stage (25–28 weeks PMA), approximately 67% of fecal bile acids identified in the extremely preterm control cohort were positional and stereoisomers of trihydroxy-cholanoic acids that were 6-hydroxylated (). These isomers typically are present in very low levels in healthy adult fecesCitation22 but are a common feature of bile acid synthesis in early life.Citation23–25 Consistent with previous reportsCitation26,Citation27 and with our finding that BSH genes are acquired during early neonatal development, fecal unconjugated bile acids became increasingly abundant over time, accounting for only 4% of all bile acids at 25–28 weeks to 98% of the bile acids by 32–40 weeks PMA. Thus, fecal bile acid profiles develop over time and in accordance with acquisition of microbial BSH in extremely preterm neonates.

Figure 3. Fecal bile acid profiles are altered during cholestasis. Bile acid stereoisomers dominate in early development, then primary unconjugated bile acids increase in abundance in control neonates. Bile acid stereoisomers are less abundant and bile acid deconjugation is impaired in cholestatic neonates (excluding samples obtained during UDCA treatment) (n = 14–26). α-MCA, alpha-muricholic acid; CA, cholic acid; CDCA, chenodeoxycholic acid; DCA, deoxycholic acid; GCA, glycocholic acid; GCDCA, glycochenodeoxycholic acid; GHyoCA, glycohyocholic acid; HyoCA, hyocholic acid; Isomer2-T-Triol, taurine conjugate of an unidentified trihydroxy-cholanoic acid; PMA, post-menstrual age; Tα-MCA, tauro-α-muricholic acid; TCA, taurocholic acid; TCDCA, taurochenodeoxycholic acid; THyoCA, taurohyocholic acid; THyoDCA, taurohyodeoxycholic acid; UDCA, ursodeoxycholic acid.

Figure 3. Fecal bile acid profiles are altered during cholestasis. Bile acid stereoisomers dominate in early development, then primary unconjugated bile acids increase in abundance in control neonates. Bile acid stereoisomers are less abundant and bile acid deconjugation is impaired in cholestatic neonates (excluding samples obtained during UDCA treatment) (n = 14–26). α-MCA, alpha-muricholic acid; CA, cholic acid; CDCA, chenodeoxycholic acid; DCA, deoxycholic acid; GCA, glycocholic acid; GCDCA, glycochenodeoxycholic acid; GHyoCA, glycohyocholic acid; HyoCA, hyocholic acid; Isomer2-T-Triol, taurine conjugate of an unidentified trihydroxy-cholanoic acid; PMA, post-menstrual age; Tα-MCA, tauro-α-muricholic acid; TCA, taurocholic acid; TCDCA, taurochenodeoxycholic acid; THyoCA, taurohyocholic acid; THyoDCA, taurohyodeoxycholic acid; UDCA, ursodeoxycholic acid.

Cholestasis altered these bile salt developmental trajectories in two key respects (). First, cholestatic neonates not given UDCA treatment had decreased proportions of 6-hydroxylated cholanoic acid isomers (29.6% vs. 66.9%) relative to control neonates at 25–28 weeks PMA. Interestingly, this difference was noted prior to the onset of cholestasis in most cases, suggesting that these isomers could potentially predict outcomes. Second, the majority of total bile acids quantified in stool from cholestatic neonates not given enteral UDCA remained conjugated (54% vs 2% in control neonates) at 32–40 weeks PMA (), providing further evidence of impaired acquisition of bile acid deconjugation activity.

Ursodeoxycholic acid treatment modifies the bile acid pool and microbiota composition

Cholestatic neonates often are treated with enteral UDCA to stimulate bile flow, although the impact of this drug on intestinal bile acid composition and gut microbial communities is unclear. In the present study, 5 of the 12 cholestatic neonates received UDCA; the remaining 7 were not treated due to enteral feeding intolerance. There were no significant differences in baseline characteristics between enrolled infants treated with UDCA, untreated cholestatic infants, and control infants (Supplementary Table S3). To understand how UDCA affects fecal bile acid profiles, we generated PC plots based on our quantitative bile acid data from all 124 fecal samples. Samples from cholestatic infants on UDCA therapy clustered separately from all other samples (). None of the cholestatic infants were treated with UDCA from 25 to 28 weeks PMA, thus further analyses examined samples from 29 to 40 weeks PMA. As expected, given that cholestasis represents a lack of bile flow into the intestine, total fecal bile acid concentrations were reduced in cholestasis (, P = 0.0002). Interestingly, although total fecal bile acid levels were much higher with UDCA treatment, this increase reflects unabsorbed drug. UDCA is relatively insoluble and absorbed in the proximal jejunum by nonionic passive diffusion, unlike conjugated bile acids that require the ileal bile acid transporter for reabsorption.Citation28,Citation29 Stool from cholestatic neonates on UDCA therapy had 522-fold higher concentrations of UDCA compared to untreated cholestatic neonates (, P < 0.0001). Indeed, UDCA accounted for 90% of total fecal bile acids among cholestatic neonates treated with UDCA (). Taken together, these data indicate that UDCA treatment dramatically alters fecal bile acid composition via the presence of unabsorbed drug.

Figure 4. UDCA treatment alters microbiota and fecal bile acid profiles. (a) PC analysis of bile acid quantification reveals that stools obtained during UDCA treatment cluster distinctively (n = 124). (b) UDCA treatment restores fecal bile acid concentrations to those of control infants 29–40 weeks PMA (n = 7–20). (c) There are higher concentrations of UDCA in treated samples compared to untreated samples from cholestatic neonates 29–40 weeks PMA (n = 7–20). (d) UDCA treatment shifts the fecal bile acid pool to predominately UDCA in neonates 29–40 weeks PMA (n = 7–20). (e) UDCA administration increases the relative abundance of Firmicutes and decreases the relative abundance of Proteobacteria in infants 29–40 weeks PMA (n = 5–20). (F) BSH-carrying Clostridium perfringens is increased with UDCA treatment, while BSH-carrying Bifidobacterium breve is decreased with UDCA treatment in cholestatic infants 29–40 weeks PMA (n = 5–20). BA, bile acid; CA, cholic acid; CDCA, chenodeoxycholic acid; DCA, deoxycholic acid; GCA, glycocholic acid; GCDCA, glycochenodeoxycholic acid; HyoCA, hyocholic acid; Isomer2-T-Triol, taurine conjugate of an unidentified trihydroxy-cholanoic acid; ω-MCA, omega-muricholic acid; PC, principal coordinate; TCA, taurocholic acid; TCDCA, taurochenodeoxycholic acid; THyoCA, taurohyocholic acid; UDCA, ursodeoxycholic acid.

**** P<0.01; ** P<0.01; * P<0.05
Figure 4. UDCA treatment alters microbiota and fecal bile acid profiles. (a) PC analysis of bile acid quantification reveals that stools obtained during UDCA treatment cluster distinctively (n = 124). (b) UDCA treatment restores fecal bile acid concentrations to those of control infants 29–40 weeks PMA (n = 7–20). (c) There are higher concentrations of UDCA in treated samples compared to untreated samples from cholestatic neonates 29–40 weeks PMA (n = 7–20). (d) UDCA treatment shifts the fecal bile acid pool to predominately UDCA in neonates 29–40 weeks PMA (n = 7–20). (e) UDCA administration increases the relative abundance of Firmicutes and decreases the relative abundance of Proteobacteria in infants 29–40 weeks PMA (n = 5–20). (F) BSH-carrying Clostridium perfringens is increased with UDCA treatment, while BSH-carrying Bifidobacterium breve is decreased with UDCA treatment in cholestatic infants 29–40 weeks PMA (n = 5–20). BA, bile acid; CA, cholic acid; CDCA, chenodeoxycholic acid; DCA, deoxycholic acid; GCA, glycocholic acid; GCDCA, glycochenodeoxycholic acid; HyoCA, hyocholic acid; Isomer2-T-Triol, taurine conjugate of an unidentified trihydroxy-cholanoic acid; ω-MCA, omega-muricholic acid; PC, principal coordinate; TCA, taurocholic acid; TCDCA, taurochenodeoxycholic acid; THyoCA, taurohyocholic acid; UDCA, ursodeoxycholic acid.

UDCA treatment also altered the fecal microbiota. At the phylum level, the relative abundance of Firmicutes increased, while the relative abundance of Proteobacteria decreased (, P < 0.05) among cholestatic neonates receiving UDCA compared to untreated cholestatic neonates. At the species level, the relative abundance of two BSH carriers was altered by UDCA. C. perfringens was enriched (, P = 0.04) and Bifidobacterium breve was depleted (, P = 0.13) in the stool of UDCA recipients. The loss of B. breve could explain why total BSH gene abundance did not differ significantly between cholestatic infants treated with UDCA and those untreated (Supplementary Figure S7). These data indicate that the dramatic fecal bile acid composition changes attributed to UDCA therapy also are associated with microbial community changes.

Associations between microbial bile acid transformations and growth

The significance of delayed acquisition of microbial bile salt deconjugation activity is not yet known. In an exploratory analysis within our cohort, we identified novel associations between markers of microbial bile acid transformations () and neonatal growth. Neonates with fecal BSH gene abundance > 0.005% at 32–40 weeks PMA exhibited 1.2-fold increased length and weight velocities compared to those with fecal BSH < 0.005% (, P < 0.05). In accord with increased BSH gene abundance, neonates with a fecal cholic acid composition of > 30% demonstrated increased length (mean increase 14%), weight (mean increase 18%), and head circumference (mean increase 15.8%) velocities (, P < 0.05) compared to those with fecal cholic acid ≤ 30%. These data suggest that fecal markers of increased BSH activity may be associated with increased neonatal growth.

Figure 5. Fecal bile acids and BSH are associated with neonatal growth outcomes. (a) Schematic of BSH transformation of TCA and TCDCA into CA and CDCA, respectively. (b) In neonates 32–40 weeks PMA, the relative proportions of the BSH gene and of fecal cholic acid are associated positively with growth (n = 12, all groups). (c) In neonates 25–28 weeks PMA, the proportion of combined fecal TCA and TCDCA is associated negatively with growth, and the proportion of combined fecal Isomer2-T-Triol and GHyoCA is associated positively with growth (n = 12, all groups). BSH, bile salt hydrolase; CA, cholic acid; CDCA, chenodeoxycholic acid; GHyoCA, glycohyocholic acid; Isomer2-T-Triol, taurine conjugate of an unidentified trihydroxy-cholanoic acid; PMA, post-menstrual age; TCA, taurocholic acid; TCDCA, taurochenodeoxycholic acid.

* P<0.05
Figure 5. Fecal bile acids and BSH are associated with neonatal growth outcomes. (a) Schematic of BSH transformation of TCA and TCDCA into CA and CDCA, respectively. (b) In neonates 32–40 weeks PMA, the relative proportions of the BSH gene and of fecal cholic acid are associated positively with growth (n = 12, all groups). (c) In neonates 25–28 weeks PMA, the proportion of combined fecal TCA and TCDCA is associated negatively with growth, and the proportion of combined fecal Isomer2-T-Triol and GHyoCA is associated positively with growth (n = 12, all groups). BSH, bile salt hydrolase; CA, cholic acid; CDCA, chenodeoxycholic acid; GHyoCA, glycohyocholic acid; Isomer2-T-Triol, taurine conjugate of an unidentified trihydroxy-cholanoic acid; PMA, post-menstrual age; TCA, taurocholic acid; TCDCA, taurochenodeoxycholic acid.

Finally, we sought to determine whether fecal bile acids present in the earliest developmental window (25–28 weeks PMA) might serve as markers of growth across the newborn hospitalization. Remarkably, we found that stools containing < 30% taurocholic acid and taurochenodeoxycholic acid (substrates for BSH enzymes) were associated with increased length (mean 22.1% increase) and increased head circumference (mean 13.6% increase) velocities (, P < 0.05) at 36 weeks PMA. Additionally, stools containing > 7% of two 6-hydroxylated bile acid isomersCitation28 (taurine conjugate of an unidentified trihydroxy-cholanoic acid [Isomer 2-T-triol] and glycohyocholic acid [GHyoCA]) were associated with a mean 22.6% increased length velocity compared to stools containing ≤ 7% (, P < 0.05). These findings suggest that an impaired enterohepatic environment, including altered BSH activity (reflected by compositional changes in fecal bile acids) is associated with reduced neonatal growth velocity.

Discussion

By characterizing fecal metagenomic profiles longitudinally among extremely preterm infants discordant for cholestasis, we have shown that gut microbiome development coincides with PMA but is compromised by cholestasis. We found that cholestasis delays the acquisition of C. perfringens and bile acid deconjugation activity, and that treating cholestatic neonates with UDCA increases total fecal bile acid levels by dramatically increasing the amount of fecal UDCA (). We also identified novel associations between microbial bile acid transformation and somatic growth, with early-life bile acid metabolites potentially predicting growth trajectories throughout the neonatal intensive care unit admission.

Figure 6. The most distinctive features of preterm gut microbiome development are increasing abundance of Clostridium perfringens and BSH, which increases total and relative proportions of unconjugated BAs. Cholestasis disrupts the acquisition of C. perfringens, BSH enzyme activity, and the capacity to form unconjugated bile acids. Enteral UDCA administration increases the abundance of C. perfringens and dramatically increases UDCA in the stool. BA, bile acid; BSH, bile salt hydrolase; UDCA, ursodeoxycholic acid. Created with BioRender.Com.

Figure 6. The most distinctive features of preterm gut microbiome development are increasing abundance of Clostridium perfringens and BSH, which increases total and relative proportions of unconjugated BAs. Cholestasis disrupts the acquisition of C. perfringens, BSH enzyme activity, and the capacity to form unconjugated bile acids. Enteral UDCA administration increases the abundance of C. perfringens and dramatically increases UDCA in the stool. BA, bile acid; BSH, bile salt hydrolase; UDCA, ursodeoxycholic acid. Created with BioRender.Com.

There are relatively few published studies on the extremely preterm neonatal gut microbiome. This is in part due to the medical complexity of this fragile population, the logistical challenges related to sequencing very low microbial biomass samples that are infrequently produced (often less than one stool per week), and the challenges of profiling the bile acid metabolome in stools.Citation30 Previous reports suggest that gut microbial community diversity decreases according to the degree of prematurity.Citation31 During full-term development, neonates typically are colonized by potentially beneficial genera including Bifidobacterium and Bacteroides.Citation32 However, premature neonates initially are colonized by members of other phyla, specifically Firmicutes followed by Proteobacteria.Citation33 Several studies also have identified the genus Clostridium as a highly abundant member of the microbial community in preterm infants.Citation16,Citation34 We observed each of these known patterns of development in our control cohort of preterm neonates.

Our whole metagenome data extend this knowledge by identifying secondary bile acid biosynthesis and BSH genes as the most striking features of microbiome development over time. In mice, the relative abundance of the BSH gene gradually increases between birth and 28 days of life.Citation35 Similarly, most healthy full-term infants are able to deconjugate bile acids by one month of age.Citation36 In our cohort, we attributed the acquisition of BSH enzymes to colonization by the BSH-carrying C. perfringens. Often considered a pathogen due to its associations with NEC, gas gangrene, and food poisoning,Citation37 others have reported that C. perfringens is present in the microbiota of healthy infants and children.Citation38,Citation39 A recent study of 247 C. perfringens isolates found increased numbers of virulence genes and higher necrotic capacity in vitro in isolates obtained from preterm infants with NEC compared to strains isolated from preterm infants without NEC.Citation40 Therefore, the increase in C. perfringens over time among our control neonates, none of whom developed NEC, likely reflects colonization by less virulent or nonpathogenic strains.

There also is very limited data on how the bile acid metabolome matures over time in extremely preterm neonates.Citation30 In full-term infant meconium, < 1%–16% of bile acids are unconjugated, but by 24 months 61–95% of all fecal bile acids are unconjugated.Citation27 Similarly, in late preterm newborns, the majority of serum bile acids are conjugated.Citation41 Precisely when unconjugated bile acids become predominant in neonates was not yet known. Our data confirm that extremely preterm newborns have mostly conjugated bile acids, and we further delineate the timeline by which the abundance of primary conjugated bile acids decreases from 96% to 2%. This timeline coincides precisely with acquisition of microbial BSH activity. Our data also highlight 6-hydroxylated bile acid isomers, which are not typically measured in most bile acid assays, as the majority of bile acids in control neonates at 25–28 weeks PMA. Gallbladder bile of fetuses between the 14th and 20th week of gestation contains an abundance of polyhydroxylated “atypical bile acids” with hydroxyl groups at positions C-1 and C-6.Citation24,Citation42 These bile acid metabolites result from bile acid biosynthetic pathways that involve cytochrome P450 enzymesCitation43 that are active in the fetal liverCitation24 and also have been observed in adults with advanced cholestatic liver disease.Citation44 Not surprisingly, atypical and short-chain bile acids also are found in meconium.Citation25 Therefore, it can be hypothesized that the bile acid composition in the most immature preterm neonates resembles that in fetal development, with a highly hydrophilic bile acid pool designed to protect hepatocytes from injury and to facilitate urinary excretion. Further studies are required to confirm their intriguing association with increased rate of growth in preterm infants.

We leveraged this new understanding of microbiome and bile acid maturation dynamics in extremely preterm neonates to gain new insight into how cholestasis alters this maturation process. We found that total fecal bile acid concentrations are markedly reduced in cholestasis, which is expected in the context of decreased bile flow from the liver into the intestine. However, our discovery of impaired formation of unconjugated bile acids, coinciding with delayed acquisition of C. perfringens and its BSH genes, may offer new insights into disease pathogenesis or therapeutic opportunities. Others have proposed BSH-carrying probiotics as a potential strategy to stimulate bile flow.Citation45–47 BSH-carrying Lactobacillus rhamnosus GG prevents hepatic fibrosis in bile duct ligated mice by increasing fecal bile acid excretion and enhancing enterohepatic circulation via the FXR/FGF15 signaling pathways.Citation48 Bile acid secretion is stimulated by similar mechanisms in a mouse model of drug-induced cholestasis by L. rhamnosus GG and other probiotic bacteria.Citation49 Although a causal relationship between decreased BSH activity and neonatal cholestasis remains unknown, BSH-based probiotic therapy is an intriguing potential therapeutic avenue. It is notable that our cholestatic neonates were on total parenteral nutrition for a longer duration of time than the control cohort and that half of the cholestatic infants developed NEC (2/6 patients required surgery). The etiology of NEC is unknown and may be multifactorial.Citation50 Future work will examine larger cohorts to study gut microbiome and bile acid interactions in relation to the development of NEC in cholestatic infants. These factors could contribute to the abnormal microbiota development in the cholestatic cohort.Citation51,Citation52

Oral UDCA therapyCitation28 has been an almost universal approach to the management of neonatal cholestasis because UDCA is highly hydrophilic and generates a bicarbonate-rich hypercholeresis, but how this drug affects the liver-gut-microbiome axis is poorly understood. In our cholestatic neonates, UDCA administration altered the microbiome and increased fecal UDCA more than 500-fold to represent 90% of total fecal bile acids. In contrast, UDCA and its conjugates represent just 40% of total bile acids in duodenal contents from healthy adults given similar weight-adjusted doses,Citation53 likely because it is metabolized to chenodeoxycholic and lithocholic acids.Citation54 We did not detect increases in conjugated forms of UDCA in stool in part because UDCA is relatively insoluble and with a pKa of around 5 is non-ionized at intestinal pH and therefore shows limited transport to the liver by nonionic passive diffusion.Citation28 In cholestasis of pregnancy, UDCA treatment increases the abundance of BSH-carrying bacteria,Citation55 similar to our finding that UDCA increases BSH-carrying C. perfringens in cholestatic neonates. Mechanisms by which UDCA promotes the growth of BSH-carrying bacteria remain unclear.

Bile acids play a critical role in the intestine to promote the digestion and absorption of lipids.Citation56 In this regard, bile acids are especially important for growth and development in the neonatal period to aid in digestion of fat, the most abundant macronutrient in their human milk diet. Preterm infants fed a human milk diet show improved gastrointestinal development, improved feeding tolerance, and decreased infection rates.Citation57,Citation58 Furthermore, preterm infants weighing ≤ 1250 grams at birth fed an exclusive human milk diet exceed targeted standards for weight gain.Citation59 Optimal growth has been linked to improved neurodevelopment, thus emphasizing the importance of adequate nutrition in preterm infants.Citation60 We have reported that preterm cholestatic neonates have reduced quantities of fecal bile acids. Failure to secrete bile acids into the intestine results in fat malabsorption and fat-soluble vitamin deficiency.Citation61 Therefore, augmenting bile flow to improve lipid absorption in cholestatic neonates may improve growth outcomes.

Similar to neonatal cholestasis, child malnutrition decreases intestinal bile acids to further impair growth.Citation62 A sub-clinical cholestasis was present in malnourished Pakistani infants aged 3–9 months; this study also identified the primary conjugated bile acid glycocholic acid, but not its unconjugated metabolite cholic acid, as a marker predicting growth faltering.Citation63 Similarly, we found high proportions of primary conjugated bile acid substrates of BSH and low proportions of unconjugated products of BSH to be associated with lower head circumference, weight, and length velocities in preterm neonatal development. The potential role of these bile acids and BSH genes as predictive biomarkers must be validated in a larger independent cohort, and a potential growth-promoting causal role of these bile acids should be explored in preclinical models.

Limitations of our study include the small sample size and single-center study design. Furthermore, study participants were recruited over 4 years, which may result in environmental microbiota shifts in the neonatal intensive care unit. We performed PC analysis to confirm there was no distinct clustering of samples by year collected (Suplementary Figure S3). Also, we chose to focus on BSH as the initial and essential step in secondary bile acid biosynthesis because the bioinformatics pipeline used for this study annotates a single gene (BSH) to the Secondary Bile Acid Biosynthesis KEGG pathway. Future studies will broaden this investigation to determine whether other microbial genes (e.g., bai genes, hydroxysteroid dehydrogenases)Citation64,Citation65 and their bacterial carriers are similarly affected by cholestasis. Additionally, blood samples were not obtained from this cohort to measure serum bile acids. Future studies will analyze serum bile acids to understand how cholestasis may impact the circulating bile acid environment. Our study was not designed to determine causal links between microbial bile acid transformations and clinical outcomes; however, longitudinal sampling allowed us to describe for the first time how gut microbial communities and bile acid profiles mature in tandem in extremely preterm neonates. Additionally, by integrating metagenomic, enzyme activity, and metabolite analyses, we were able to characterize this developmental maturation in greater detail. Additional strengths include the sensitivity of our MS-based bile acid quantification, which allowed us to identify and quantify atypical bile acid metabolites in stool samples, as well as the sensitivity of our bile salt hydrolase activity assay, which facilitates the detection of quantitative changes in BSH activity from only 15 µg of fecal protein. This assay can be applied to other low microbial biomass samples including meconium.

In summary, these data lend new insights into how cholestasis affects the intestinal microbiota and bile acid composition of extremely preterm neonates. Delayed acquisition of microbial bile acid deconjugation warrants further study as a predictive marker or possibly even as a contributing factor to cholestasis and neonatal growth failure.

Patients and methods

Study participants and sample collection

In this nested case-control study, we selected 24 preterm infants of less than 30 weeks gestation born at Texas Children’s Hospital-Pavilion for Women (Houston, TX, United States of America). Subjects were enrolled between September 2015 and August 2019. Fecal samples were collected by the subject’s bedside nurse into sterile fecal collection containers, stored at 4°C, and transferred within 48 h into aliquots of 0.5 mL by research study personnel and stored at −80°C until analysis.

Subjects were selected from two larger observational prospective cohort studies involving preterm infants. Both studies collected serial stool samples to be used for microbiome analysis. Studies were approved by the Institutional Review Board at Baylor College of Medicine (protocols H-36828 and H-43075). All study procedures were in accordance with the ethical standards of Baylor College of Medicine. Written parental informed consent was provided for each infant enrolled in each study. Eligible study participants from Study 1Citation66 included premature infants born at < 1500 g with no barriers to enteral milk feedings enrolled within the first 72 h of life. Exclusion criteria included anomalies or birth defects that precluded enteral feeding, severe perinatal hypoxia, or < 50% projected survival based on the National Institute of Child Health and Human Development Neonatal Research Network Extremely Preterm Birth Outcome Data calculator. For Study 2, eligible study participants included premature infants born between 24 and 34 weeks GA, birth weight ≤ 1250 g, and enrolled within the first 72 h of life. Infants were excluded for > 1250 g birth weight, low likelihood of survival, major congenital anomalies or clinically significant congenital heart disease, intestinal perforation, Bell’s Stage 2 or greater NEC, early transfer to another institution, or failure to achieve full enteral feedings by 28 days of life.

Cholestasis was defined by a serum conjugated bilirubin level ≥ 1 mg/dL and > 20% of the total bilirubin at any point during hospital admission. Elevated conjugated bilirubin represents impaired bile formation or flow.Citation9 All neonates were screened within the first 48 h of life and again as clinically indicated (e.g., concerns for sepsis or new-onset jaundice). Each cholestatic infant was matched to a control (non-cholestatic) infant with emphasis on matching by birthweight, sex, delivery by Cesarean section, antibiotic exposure, % of feeds mother’s milk, and multiple fecal samples obtained. Anthropometrics were obtained at birth and approximately 36 weeks PMA by a trained research nurse. Growth velocities (weight, length, and head circumference) were calculated from birth to approximately 36 weeks PMA. The timing of patient fecal sampling, serum conjugated bilirubin measurements, antibiotic use, and UDCA treatment (if applicable) are shown in Supplementary Figures S1 and S2.

Fecal microbiome analysis

Complete methodological details are presented in the STORMSCitation67 (Strengthening the Organization and Reporting of Microbiome Studies) checklist (Table S1, Tables S4-S12). Briefly, whole metagenome shotgun sequencing was performed at the Alkek Center for Metagenomics and Microbiome Research at Baylor College of Medicine. Fecal samples were collected and stored at −80°C until DNA was extracted using the DNeasy 96 PowerSoil Pro QIAcube HT Kit (Qiagen) according to the manufacturer’s protocol. DNA was quantified using Qubit fluorometric quantitation prior to library preparation. Sequencing was performed on the Illumina NovaSeq S4 platform using the 150 bp paired-end read protocol (average sequencing depth = 41518559; minimum sequencing depth = 33936). Negative controls were tubes with reagents from DNA extraction and water only, and positive controls were from the MSA-2002 and MSA-1003 mock communities (ATCC). Quality filtering and trimming were performed using BBTools (sourceforge.net/projects/bbmap/; BBMap version 38.82). Trimming parameters were set to a k-mer length of 19 and a minimum Phred quality score of 25. Reads with a minimum average Phred quality score below 23 and length shorter than 50 bp after trimming were discarded. Final sample size after quality control was 113 (original total = 124). The taxonomic composition of the filtered reads was determined using the MetaPhlAn3 software package.Citation68

Functional metagenomic analysis was performed with the HUMAnN3 package.Citation68 Microbial genes and metabolic pathways were annotated to the Kyoto Encyclopedia of Genes and Genomes (KEGG) database.Citation69 Raw data files have been submitted to the NCBI SRA database under the accession number PRJNA870588.

Bile acid profiling by ultra-performance liquid chromatography coupled with tandem mass spectrometry (UPLC-MS/MS)

The fecal bile acid metabolome was determined by ultra-performance liquid chromatography coupled with tandem mass spectrometry (UPLC-MS/MS). Bile acid extraction was carried out on wet feces. Samples were weighed, homogenized, and sonicated and bile acids extracted based on previous methodsCitation22,Citation70 using 80% methanol/water followed by chloroform/methanol (2:1, v/v). The supernatants were combined and dried under nitrogen. The dried extract was dissolved in 1 mL of methanol and a 1/20th aliquot was used for sample clean-up and bile acid extraction using a C18 solid phase cartridge. Internal standards comprising 15 stable-isotopically labeled bile acids were added to the fecal extract prior to the extraction. The bile acid final extract was reconstituted in 200 µL of methanol/water (50/50 v/v) and analyzed by UPLC-MS/MS.

Quantitative analysis of the individual major bile acids and their isomers in the fecal extracts was conducted by UPLC-MS/MS with electrospray ionization (ESI) using a Waters Xevo TQ-S triple quadruple mass spectrometer interfaced with an Aquity UPLC system (Waters Corporation, Milford, MA). Individual bile acid species were separated on a Kinetex C18 (2.6 µm, 100 × 3.0 mm) column (Phenomenex, Torrance, CA) with gradient elution consisting of mobile phase A (20% acetonitrile/water with 10 mM ammonium acetate) and mobile phase B (80% acetonitrile/water with 10 mM ammonium acetate) programmed from 5% mobile phase B to 100% mobile phase B, with a total run time of 20 min. Calibration curves used for quantification were built with 23 of the bile acid reference compounds and detected by multiple reaction monitoring (MRM) function operated with negative ion detection.

Bile salt hydrolase activity assay

A colorimetric ninhydrin BSH activity assay optimized for stool samples was adapted from Tanaka et al. and Gopal-Srivastava et al.Citation20,Citation71 The experimental samples varied in fluidity, had low microbial and protein content, and contained insoluble particles that interfered with colorimetric estimation of glycine or taurine. Given these limitations, we optimized the following protocol for precise measurements of BSH activity. First, stool samples were dried at 37°C and dry weight was used for optimal microbial protein recovery. After suspending 300 mg dried stool samples in 3 mL suspension buffer (PBS with 10% glycerol, 5 mM DTT, and protease inhibitor cocktail), the solution was sonicated on ice for 6 cycles of 30 s on/off cycles. Sonicated stool suspensions were centrifuged at 4000 g for 10 min at 4°C. Supernatants were concentrated using Amicon Ultra-5 10K centrifugal filters. Approximately 300 µL of concentrated sample was further centrifuged at 16,000 g for 10 min at 4°C. The protein extract was stored at −80°C until use. Assay buffer (10 mM citrate buffer, 10 mM DDT; pH 5.8) was added to 15 µg of fecal protein, then 10 mM of GCA or TCA was added for a final volume of 200 µL. Negative control samples contained 200 µL of the reaction mixture with 200 µL of 15% TCA. Reactions were carried out at 37°C, and 100 µL was collected after 30 and 60 min of incubation. Immediately after sample collection, samples were mixed with an equal volume of 15% TCA and placed on ice. Samples were centrifuged (16,000 g, 4°C, 15 min) and the supernatant was collected. Ninhydrin reagent (263 µL ninhydrin, 632 µL glycerol, 105 µL 0.5 M citrate buffer; pH 5.5) was added to the samples, which were boiled for 15 min. Tubes were cooled under running water; from each tube 200 µL of volume was further diluted to 1.2 mL with water and the absorbance at 570 nm was determined. Nanomoles of glycine or taurine were calculated against a standard curve and background absorbance for each sample was subtracted using the negative control values.

Statistical analysis

Statistical analyses were performed in GraphPad Prism (v. 9.3.0) and RStudio (v. 1.4.1106). Codes for data analysis and complete metadata can be accessed at https://github.com/laurenevelynch/Preterm_Cholestasis.git.

Community abundance and alpha diversity metrics were obtained from count data using the phyloseq package.Citation72 Relative abundances were used for further microbiota analyses and calculated using the total-sum scaling (TSS) technique. Kruskal-Wallis (KW) tests with FDR adjustment were applied to the dataset of control infants to determine differences in species relative abundance. LEfSe was performed to detect differentially abundant pathways with an effect size cut-off of 2.0 and KW P < 0.05.Citation73 Differences in pathway and species abundances by PMA in control infants were analyzed with KW tests followed by Dunn’s multiple comparisons test. PC analysis with Bray-Curtis dissimilarity was performed in the following manner using the vegan package.Citation74 First, the PC 1 axis that accounted for the greatest variation among microbiota communities from control neonates was determined. Next, this PC 1 axis was used to generate a second PC plot for cholestatic neonates based on weighted PC analysis with the wcmdscale function in vegan. For this second plot, samples from cholestatic neonates were assigned a weight of 0 and their position on PC 1 relative to control samples (weight = 1) was computed. Finally, the relationship between PC 1 and PMA for both groups was determined via linear regression.

All data were tested for normality, and non-parametric Mann-Whitney tests were applied for non-normally distributed data to test significance between the control and cholestatic groups at 32–40 weeks PMA. BSH activity assay data were normally distributed and assessed via ANOVA with correction for multiple comparisons by the Benjamini and Hochberg method. Bile acid relative abundance is depicted as median values in pie charts. PC coordinates for bile acid data were generated via the prcomp package in R. Total fecal bile acids between control and treatment groups were analyzed via KW tests with correction by Dunn’s. Phyla and species differences between treatment groups were assessed by Mann-Whitney tests.

Clinical characteristics were tested for normality by the Shapiro-Wilk test. Comparisons were analyzed with t-tests for normally distributed continuous variables, Mann-Whitney tests for non-normally distributed data, and Fisher’s exact test for categorical variables. For comparisons with more than two groups, ANOVA or KW tests were used. To analyze associations between microbial bile acid transformations and growth, data were visualized with histograms and inflection points within the data set were identified. Significance was assessed at the α < 0.05 level.

Abbreviations

α-MCA, alpha-muricholic acid, 3α,6β,7α-trihydroxy-5β-cholanoic acid; BSH, bile salt hydrolase; CA, cholic acid, 3α,7α,12α-trihydroxy-5β-cholanoic acid; CDCA, chenodeoxycholic acid, 3α,7α-dihydroxy-5β-cholanoic acid; DCA, deoxycholic acid, 3α,12α-dihydroxy-5β-cholanoic acid; FDR, false discovery rate; GA, gestational age; GCA, glycocholic acid, glyco-3α,7α-dihydroxy-5β-cholanoic acid; GCDCA, glycochenodeoxycholic acid, glyco-3α,7α-dihydroxy-5β-cholanoic acid; GHyoCA, glycohyocholic acid, glyco-3α,6α,7α-trihydroxy-5β-cholanoic acid; HyoCA, hyocholic acid, 3α,6α,7α-trihydroxy-5β-cholanoic acid; isomer 2-T-triol, taurine conjugate of an unidentified trihydroxy-cholanoic acid; KW, Kruskal-Wallis; LDA, linear discriminant analysis; LEfSe, linear discriminant analysis effect size; MS, mass spectrometry; NEC, necrotizing enterocolitis; ω-MCA, omega-muricholic acid, 3α,6α,7α-trihydroxy-5β-cholanoic acid; PC, principal coordinate; PMA, post menstrual age; Tα-MCA, tauro- alpha-muricholic acid, tauro-3α,6,7α-trihydroxy-5β-cholanoic acid; TCA, taurocholic acid, tauro-3α,7α,12α-trihydroxy-5β-cholanoic acid; TCDCA, taurochenodeoxycholic acid, tauro-3α,7α-dihydroxy-5β-cholanoic acid; THyoCA, taurohyocholic acid, tauro-3α,6α,7α-trihydroxy-5β-cholanoic acid; THyoDCA, taurohyodeoxychloic acid, tauro-3α,6α-dihydroxy-5β-cholanoic acid; UDCA, ursodeoxycholic acid, 3α,7α-dihydroxy-5β-cholanoic acid; UPLC-MS/MS, ultra-performance liquid chromatography coupled with tandem mass spectrometry.

Availability of data

The data are openly available in the National Center for Biotechnology Information Sequence Read Archive under the accession number PRJNA870588.

Supplemental material

Supplemental Material

Download MS Excel (24.3 KB)

Supplemental Material

Download MS Word (790.5 KB)

Disclosure statement

KDRS discloses equity in Asklepion Pharmaceuticals, LLC, Baltimore and Aliveris s.r.l. Italy, and is a consultant to Travere Therapeutics and Mirum Pharmaceuticals. No potential conflict of interest was reported by the other authors.

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/19490976.2023.2183690

Additional information

Funding

This project was supported by the U.S. National Institute of Diabetes and Digestive and Kidney Diseases grant/award K08 DK113114 to G.A.P., the U.S. Public Health Service grant/award P30 DK056338 which funds the Texas Medical Center Digestive Diseases Center, the U.S. National Institute of General Medical Sciences T32 GM136554 training grant to L.E.L., and the Men of Distinction grant/award to A.B.H. and G.A.P.

References

  • Walani SR. Global burden of preterm birth. Int J Gynecol Obstet. 2020;150(1):31–18. doi:10.1002/ijgo.13195.
  • Purisch SE, Gyamfi-Bannerman C. Epidemiology of preterm birth. Semin Perinatol. 2017;41(7):387–391. doi:10.1053/j.semperi.2017.07.009.
  • Neu J. Gastrointestinal development and meeting the nutritional needs of premature infants. Am J Clin Nutr. 2007;85(2):629S–634S. doi:10.1093/ajcn/85.2.629S.
  • Henderickx JGE, Zwittink RD, van Lingen RA, Knol J, Belzer C. The preterm gut microbiota: an inconspicuous challenge in nutritional neonatal care. Front Cell Infect Microbiol. 2019; 9:85. doi:10.3389/fcimb.2019.00085.
  • Nastasio S, Maggiore G. Cholestasis in preterm infants: when is a yellow alert?. Ital J Pediatr. 2014;40(S2). A12, 1824-7288-40-S2–A12. doi:10.1186/1824-7288-40-S2-A12.
  • Gotze T, Blessing H, Grillhosl C, Gerner P, Hoerning A. 2015. Neonatal cholestasis – differential diagnoses, current diagnostic procedures, and treatment. Front Pediatr [Internet]. [accessed 2022 Apr 25]. doi:10.3389/fped.2015.00043
  • Satrom K, Gourley G. Cholestasis in preterm infants. Clin Perinatol. 2016;43(2):355–373. doi:10.1016/j.clp.2016.01.012.
  • Christensen RD, Henry E, Wiedmeier SE, Burnett J, Lambert DK. Identifying patients, on the first day of life, at high-risk of developing parenteral nutrition-associated liver disease. J Perinatol. 2007;27(5):284–290. doi:10.1038/sj.jp.7211686.
  • Feldman AG, Sokol RJ. Recent developments in diagnostics and treatment of neonatal cholestasis. Semin Pediatr Surg. 2020;29(4):150945. doi:10.1016/j.sempedsurg.2020.150945.
  • de Aguiar Vallim Tq, Tarling EJ, Edwards PA. Pleiotropic roles of bile acids in metabolism. Cell Metab. 2013;17(5):657–669. doi:10.1016/j.cmet.2013.03.013.
  • Hofmann AF. The continuing importance of bile acids in liver and intestinal disease. Arch Intern Med. 1999;159(22):2647. doi:10.1001/archinte.159.22.2647.
  • Guo C, Li Y, Wang P, Li Y, Qiu C, Li M, Wang D, Zhao R, Li D, Wang Y, et al. Alterations of gut microbiota in cholestatic infants and their correlation with hepatic function. Front Microbiol. 2018;9:2682. doi:10.3389/fmicb.2018.02682.
  • Ridlon JM, Harris SC, Bhowmik S, Kang D-J, Hylemon PB. Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes. 2016;7(1):22–39. doi:10.1080/19490976.2015.1127483.
  • Chand D, Avinash VS, Yadav Y, Pundle AV, Suresh CG, Ramasamy S. Molecular features of bile salt hydrolases and relevance in human health. Biochim Biophys Acta (BBA) - Gen Sub. 2017;1861(1):2981–2991. doi:10.1016/j.bbagen.2016.09.024.
  • Younge NE, Newgard CB, Cotten CM, Goldberg RN, Muehlbauer MJ, Bain JR, Stevens RD, O’connell TM, Rawls JF, Seed PC, et al. Disrupted maturation of the microbiota and metabolome among extremely preterm infants with postnatal growth failure. Sci Rep. 2019;9(1):8167. doi:10.1038/s41598-019-44547-y.
  • Grier A, Qiu X, Bandyopadhyay S, Holden-Wiltse J, Kessler HA, Gill AL, Hamilton B, Huyck H, Misra S, Mariani TJ, et al. Impact of prematurity and nutrition on the developing gut microbiome and preterm infant growth. Microbiome. 2017;5(1):158. doi:10.1186/s40168-017-0377-0.
  • La Rosa PS, Warner BB, Zhou Y, Weinstock GM, Sodergren E, Hall-Moore CM, Stevens HJ, Bennett WE, Shaikh N, Linneman LA, et al. Patterned progression of bacterial populations in the premature infant gut. Proc Natl Acad Sci U S A. 2014;111(34):12522–12527. doi:10.1073/pnas.1409497111.
  • Korpela K, Blakstad EW, Moltu SJ, Strømmen K, Nakstad B, Rønnestad AE, Brække K, Iversen PO, Drevon CA, de Vos W. Intestinal microbiota development and gestational age in preterm neonates. Sci Rep. 2018;8(1):2453. doi:10.1038/s41598-018-20827-x.
  • Ttb H, Groer MW, Kane B, Yee AL, Torres BA, Gilbert JA, Maheshwari A. Dichotomous development of the gut microbiome in preterm infants. Microbiome. 2018;6(1):157. doi:10.1186/s40168-018-0547-8.
  • Gopal-Srivastava R, Hyelemon PB. Purification and characterization of bile salt hydrolase from Clostridium perfringens. J Lipid Res. 1988;29(8):1079–1085. doi:10.1016/S0022-2275(20)38464-9.
  • Coleman JP, Hudson LL. Cloning and characterization of a conjugated bile acid hydrolase gene from Clostridium perfringens. Appl Environ Microbiol. 1995;61(7):2514–2520. doi:10.1128/aem.61.7.2514-2520.1995.
  • Setchell KD, Lawson AM, Tanida N, Sjövall J. General methods for the analysis of metabolic profiles of bile acids and related compounds in feces. J Lipid Res. 1983;24(8):1085–1100. doi:10.1016/S0022-2275(20)37923-2.
  • Nakagawa M, Setchell KD. Bile acid metabolism in early life: studies of amniotic fluid. J Lipid Res. 1990;31(6):1089–1098. doi:10.1016/S0022-2275(20)42749-X.
  • Colombo C, Zuliani G, Ronchi M, Breidenstein J, Setchel KDR. Biliary bile acid composition of the human fetus in early gestation. Pediatr Res. 1987;21(2):197–200. doi:10.1203/00006450-198702000-00017.
  • Lester R, St Pyrek J, Little JM, Adcock EW. Diversity of bile acids in the fetus and newborn infant. J Pediatr Gastroenterol Nutr. 1983;2(2):355–364. doi:10.1097/00005176-198305000-00026.
  • Niijima S-I. Studies on the conjugating activity of bile acids in children. Pediatr Res. 1985;19(3):302–307. doi:10.1203/00006450-198503000-00010.
  • Jönsson G, Midtvedt A-C, Norman A, Midtvedt T. Intestinal microbial bile acid transformation in healthy infants. J Pediatr Gastroenterol Nutr. 1995;20(4):394–402. doi:10.1097/00005176-199505000-00004.
  • Crosignani A, Setchell KD, Invernizzi P, Larghi A, Rodrigues CM, Podda M. Clinical pharmacokinetics of therapeutic bile acids. Clin Pharmacokinet. 1996;30(5):333–358. doi:10.2165/00003088-199630050-00002.
  • Setchell KDR, Heubi JE, Shah S, Lavine JE, Suskind D, Al-Edreesi M, Potter C, Russell DW, O’connell NC, Wolfe B, et al. Genetic defects in bile acid conjugation cause fat-soluble vitamin deficiency. Gastroenterology. 2013;144(5):945-955.e6; quiz e14–15. doi:10.1053/j.gastro.2013.02.004.
  • Setchel KDR, Kritchevsky D, Nair P. The bile acids, methods, and applications. New York: Plenum Press; 1988.
  • Chernikova DA, Madan JC, Housman ML, Zain-Ul-Abideen M, Lundgren SN, Morrison HG, Sogin ML, Williams SM, Moore JH, Karagas MR, et al. The premature infant gut microbiome during the first 6 weeks of life differs based on gestational maturity at birth. Pediatr Res. 2018;84(1):71–79. doi:10.1038/s41390-018-0022-z.
  • Saturio S, Nogacka AM, Suárez M, Fernández N, Mantecón L, Mancabelli L, Milani C, Ventura M, de Los Reyes-Gavilán CG, Solís G, et al. Early-life development of the bifidobacterial community in the infant gut. IJMS. 2021;22(7):3382. doi:10.3390/ijms22073382.
  • Underwood MA, Sohn K. The microbiota of the extremely preterm infant. Clin Perinatol. 2017;44(2):407–427. doi:10.1016/j.clp.2017.01.005.
  • Ferraris L, Butel MJ, Campeotto F, Vodovar M, Rozé JC, Aires J. Clostridia in premature neonates’ gut: incidence, antibiotic susceptibility, and perinatal determinants influencing colonization. Forestier C, editor. Plos One. 2012;7(1):e30594. doi:10.1371/journal.pone.0030594.
  • van Best N, Rolle-Kampczyk U, Schaap FG, Basic M, Olde Damink SWM, Bleich A, Savelkoul PHM, von Bergen M, Penders J, Hornef MW. Bile acids drive the newborn’s gut microbiota maturation. Nat Commun. 2020;11(1):3692. doi:10.1038/s41467-020-17183-8.
  • Tanaka M, Sanefuji M, Morokuma S, Yoden M, Momoda R, Sonomoto K, Ogawa M, Kato K, Nakayama J. The association between gut microbiota development and maturation of intestinal bile acid metabolism in the first 3 y of healthy Japanese infants. Gut Microbes. 2020;11(2):205–216. doi:10.1080/19490976.2019.1650997.
  • Kiu R, Hall LJ. An update on the human and animal enteric pathogen Clostridium perfringens. Emerg Microbes Infect. 2018;7(1):1–15. doi:10.1038/s41426-018-0144-8.
  • Kiu R, Sim K, Shaw A, Cornwell E, Pickard D, Kroll JS, Hall LJ. Genomic analysis of clostridium perfringens BEC/CPILE-positive, toxinotype D and E strains isolated from healthy children. Toxins. 2019;11(9):543. doi:10.3390/toxins11090543.
  • Nagpal R, Ogata K, Tsuji H, Matsuda K, Takahashi T, Nomoto K, Suzuki Y, Kawashima K, Nagata S, Yamashiro Y. Sensitive quantification of Clostridium perfringens in human feces by quantitative real-time PCR targeting alpha-toxin and enterotoxin genes. BMC Microbiol. 2015;15(1):219. doi:10.1186/s12866-015-0561-y.
  • Kiu R, Shaw A, Sim K, Bedwell H, Cornwell E, Pickard D, Belteki G, Malsom J, Philips S, Young GR, et al. 2021. Dissemination and pathogenesis of toxigenic Clostridium perfringens strains linked to neonatal intensive care units and Necrotising Enterocolitis [Internet]. [place unknown]. Microbiology. [accessed 2022 Jun 22]. doi:10.1101/2021.08.03.454877
  • Zöhrer E, Resch B, Scharnagl H, Schlagenhauf A, Fauler G, Stojakovic T, Hofer N, Lang U, Jahnel J. Serum bile acids in term and preterm neonates: a case–control study determining reference values and the influence of early-onset sepsis. Medicine. 2016;95(44):e5219. doi:10.1097/MD.0000000000005219.
  • Setchell KD, Dumaswala R, Colombo C, Ronchi M. Hepatic bile acid metabolism during early development revealed from the analysis of human fetal gallbladder bile. J Biol Chem. 1988;263(32):16637–16644. doi:10.1016/S0021-9258(18)37438-6.
  • Russell DW, Setchell KDR. Bile acid biosynthesis. Biochemistry. 1992;31(20):4737–4749. doi:10.1021/bi00135a001.
  • Bremmelgaard A, Sjövall J. Bile acid profiles in urine of patients with liver diseases. Eur J Clin Invest. 1979;9(5):341–348. doi:10.1111/j.1365-2362.1979.tb00894.x.
  • Degirolamo C, Rainaldi S, Bovenga F, Murzilli S, Moschetta A. Microbiota modification with probiotics induces hepatic bile acid synthesis via downregulation of the Fxr-Fgf15 axis in mice. Cell Rep. 2014;7(1):12–18. doi:10.1016/j.celrep.2014.02.032.
  • Hou G, Peng W, Wei L, Li R, Yuan Y, Huang X, Yin Y. Lactobacillus delbrueckii interfere with bile acid enterohepatic circulation to regulate cholesterol metabolism of growing–finishing pigs via its bile salt hydrolase activity. Front Nutr. 2020; 7:617676. doi:10.3389/fnut.2020.617676.
  • Begley M, Hill C, Gahan CGM. Bile salt hydrolase activity in probiotics. Appl Environ Microbiol. 2006;72(3):1729–1738. doi:10.1128/AEM.72.3.1729-1738.2006.
  • Liu Y, Chen K, Li F, Gu Z, Liu Q, He L, Shao T, Song Q, Zhu F, Zhang L, et al. Probiotic Lactobacillus rhamnosus GG prevents liver fibrosis through inhibiting hepatic bile acid synthesis and enhancing bile acid excretion in mice. Hepatology. 2020;71(6):2050–2066. doi:10.1002/hep.30975.
  • Chen Q, Li Q, Cao M, Yan J, Zhang X. Hierarchy-assembled dual probiotics system ameliorates cholestatic drug-induced liver injury via gut-liver axis modulation. Adv Sci. 2022;9(17):2200986. doi:10.1002/advs.202200986.
  • Tanner SM, Berryhill TF, Ellenburg JL, Jilling T, Cleveland DS, Lorenz RG, Martin CA. Pathogenesis of necrotizing enterocolitis. Am J Pathol. 2015;185(1):4–16. doi:10.1016/j.ajpath.2014.08.028.
  • Wang Y, Hoenig JD, Malin KJ, Qamar S, Petrof EO, Sun J, Antonopoulos DA, Chang EB, Claud EC. 16S rRNA gene-based analysis of fecal microbiota from preterm infants with and without necrotizing enterocolitis. Isme J. 2009;3(8):944–954. doi:10.1038/ismej.2009.37.
  • Dahlgren AF, Pan A, Lam V, Gouthro KC, Simpson PM, Salzman NH, Nghiem-Rao TH. Longitudinal changes in the gut microbiome of infants on total parenteral nutrition. Pediatr Res. 2019;86(1):107–114. doi:10.1038/s41390-019-0391-y.
  • Dilger K, Hohenester S, Winkler-Budenhofer U, Bastiaansen BAJ, Schaap FG, Rust C, Beuers U. Effect of ursodeoxycholic acid on bile acid profiles and intestinal detoxification machinery in primary biliary cirrhosis and health. J Hepatol. 2012;57(1):133–140. doi:10.1016/j.jhep.2012.02.014.
  • Lepercq P, Gã©rard P, Bã©guet F, Raibaud P, Grill J-P, Relano P, Cayuela C, Juste C. Epimerization of chenodeoxycholic acid to ursodeoxycholic acid by Clostridium baratii isolated from human feces. FEMS Microbiol Lett. 2004;235(1):65–72. doi:10.1111/j.1574-6968.2004.tb09568.x.
  • Ovadia C, Perdones-Montero A, Fan HM, Mullish BH, McDonald JAK, Papacleovoulou G, Wahlström A, Ståhlman M, Tsakmaki A, Clarke LCD, et al. Ursodeoxycholic acid enriches intestinal bile salt hydrolase-expressing Bacteroidetes in cholestatic pregnancy. Sci Rep. 2020;10(1):3895. doi:10.1038/s41598-020-60821-w.
  • Hofmann AF. Bile acids: the good, the bad, and the ugly. Physiology. 1999;14(1):24–29. doi:10.1152/physiologyonline.1999.14.1.24.
  • Schanler RJ. Outcomes of human milk-fed premature infants. Semin Perinatol. 2011;35(1):29–33. doi:10.1053/j.semperi.2010.10.005.
  • Sisk PM, Lovelady CA, Gruber KJ, Dillard RG, O’shea TM. Human milk consumption and full enteral feeding among infants who weigh ≤1250 grams. Pediatrics. 2008;121(6):e1528–1533. doi:10.1542/peds.2007-2110.
  • Hair AB, Hawthorne KM, Chetta KE, Abrams SA. Human milk feeding supports adequate growth in infants ≤ 1250 grams birth weight. BMC Res Notes. 2013;6(1):459. doi:10.1186/1756-0500-6-459.
  • Ehrenkranz RA, Dusick AM, Vohr BR, Wright LL, Wrage LA, Poole WK. For the national institutes of child health and human development neonatal research network. Growth in the neonatal intensive care unit influences neurodevelopmental and growth outcomes of extremely low birth weight infants. Pediatrics. 2006;117(4):1253–1261. doi:10.1542/peds.2005-1368.
  • Best C, Gourley GR. Management of neonatal cholestasis. Therapy. 2009;6(1):75–81. doi:10.2217/14750708.6.1.75.
  • Moreau GB, Ramakrishnan G, Cook HL, Fox TE, Nayak U, Ma JZ, Colgate ER, Kirkpatrick BD, Haque R, Petri WA. Childhood growth and neurocognition are associated with distinct sets of metabolites. EBioMedicine. 2019.44: 597–606. doi:10.1016/j.ebiom.2019.05.043.
  • Zhao X, Setchell KDR, Huang R, Mallawaarachchi I, Ehsan L, Dobrzykowski IE, Zhao J, Syed S, Ma JZ, Iqbal NT, et al. Bile acid profiling reveals distinct signatures in undernourished children with environmental enteric dysfunction. J Nutr. 2021;151(12):3689–3700. doi:10.1093/jn/nxab321.
  • Ridlon JM, Kang D-J, Hylemon PB. Isolation and characterization of a bile acid inducible 7α-dehydroxylating operon in Clostridium hylemonae TN271. Anaerobe. 2010;16(2):137–146. doi:10.1016/j.anaerobe.2009.05.004.
  • Lee J-Y, Arai H, Nakamura Y, Fukiya S, Wada M, Yokota A. Contribution of the 7β-hydroxysteroid dehydrogenase from Ruminococcus gnavus N53 to ursodeoxycholic acid formation in the human colon. J Lipid Res. 2013;54(11):3062–3069. doi:10.1194/jlr.M039834.
  • Ford SL, Lohmann P, Preidis GA, Gordon PS, O’donnell A, Hagan J, Venkatachalam A, Balderas M, Luna RA, Hair AB. Improved feeding tolerance and growth are linked to increased gut microbial community diversity in very-low-birth-weight infants fed mother’s own milk compared with donor breast milk. Am J Clin Nutr. 2019;109(4):1088–1097. doi:10.1093/ajcn/nqz006.
  • Mirzayi C, Renson A, Standards Consortium G, Analysis M, Control Society Q, Zohra F, Elsafoury S, Geistlinger L, Kasselman LJ, Eckenrode K, et al. Reporting guidelines for human microbiome research: the STORMS checklist. Nat Med. 2021;27(11):1885–1892. doi:10.1038/s41591-021-01552-x.
  • Beghini F, McIver LJ, Blanco-Míguez A, Dubois L, Asnicar F, Maharjan S, Mailyan A, Manghi P, Scholz M, Thomas AM, et al. Integrating taxonomic, functional, and strain-level profiling of diverse microbial communities with bioBakery 3. eLife. 2021;10:e65088. doi:10.7554/eLife.65088.
  • Kanehisa M. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30. doi:10.1093/nar/28.1.27.
  • Jose S, Mukherjee A, Horrigan O, Setchell KDR, Zhang W, Moreno-Fernandez ME, Andersen H, Sharma D, Haslam DB, Divanovic S, et al. Obeticholic acid ameliorates severity of Clostridioides difficile infection in high fat diet-induced obese mice. Mucosal Immunol. 2021;14(2):500–510. doi:10.1038/s41385-020-00338-7.
  • Tanaka H, Hashiba H, Kok J, Mierau I. Bile salt hydrolase of bifidobacterium longum —biochemical and genetic characterization. Appl Environ Microbiol. 2000;66(6):2502–2512. doi:10.1128/AEM.66.6.2502-2512.2000.
  • McMurdie PJ, Holmes S. Phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. Plos One. 2013;8(4):e61217. doi:10.1371/journal.pone.0061217.
  • Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, Huttenhower C. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12(6):R60. doi:10.1186/gb-2011-12-6-r60.
  • Oksanen J, Blanchet FG, Friendly M, Kindt R, Legendre P, McGlinn D, Minchin PR, O’hara RB, Simpson GL, Solymos P, et al. Vegan: Community Ecology Package. R package version 2.5-7. 2020. https://CRAN.R-project.org/package=vegan