ABSTRACT
Progression of chronic liver diseases is precipitated by hepatocyte loss, inflammation and fibrosis. This process results in the loss of critical hepatic functions, increasing morbidity and the risk of infection. Medical interventions that treat complications of hepatic failure, including antibiotic administration for systemic infections, impact gut microbiome composition and metabolite production. Using a multi-omics approach on 850 fecal samples from 263 patients with acute or chronic liver disease, we demonstrate that patients hospitalized for liver disease have reduced microbiome diversity and a paucity of bioactive metabolites. We find that patients treated with the orally administered but non-absorbable disaccharide lactulose have increased densities of intestinal Bifidobacteria and reduced incidence of systemic infections and mortality. Bifidobacteria metabolize lactulose, produce high concentrations of acetate and acidify the gut lumen, which, in combination, can reduce the growth of antibiotic-resistant pathobionts such as Vancomycin-resistant Enterococcus faecium. Our studies suggest that lactulose and Bifidobacteria serve as a synbiotic to reduce rates of infection in patients with severe liver disease.
INTRODUCTION
The incidence of chronic liver disease (CLD) is rising, in part resulting from increased rates of metabolic syndrome and exposure to toxins, including alcohol1–4. Left untreated, all etiologies of CLD converge on the final common endpoint of cirrhosis with similar complications resulting from portal hypertension. CLD, in its early stages, can be clinically silent until hepatic decompensation, characterized by the development of ascites, variceal bleeding or hepatic encephalopathy (HE). Decompensating events often recur with increasing frequency, resulting in death or the need for liver transplant5,6. Treatment of advanced CLD is largely supportive and the ability to modify the clinical course of cirrhosis remains limited. Patients with decompensated cirrhosis frequently receive antibiotic treatment for HE, which is exacerbated by the activity of gut microbes and infection. The current first line treatment for HE is the non-absorbable disaccharide lactulose7. The mechanism through which lactulose reduces serum ammonia levels is incompletely known, and its role as a prebiotic that modifies the microbiome remains controversial 8–12. Moreover, there are varying reports illustrating the effects of lactulose on development of systemic infections and other complications of CLD, including death13,14. Together, these reports indicate that further evaluation is needed to fully understand the effects of lactulose on gut microbiome compositions and function as well as important clinical outcomes in patients with liver disease, including development of systemic infections.
The gut microbiome impacts human health and a wide range of diseases. Given the bidirectional anatomic communication between the liver and the gut via the portal vein and biliary tree, it is postulated that the microbiome plays a role in liver disease pathogenesis. Preclinical studies implicate the gut microbiome as a potential driver of non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD)15–17. While robust clinical evidence linking the microbiome to progression of liver disease is lacking, multiple observational studies have reported various gut microbiome “signatures” of advanced fibrosis and cirrhosis9,18–21.
Observational studies have also associated gut microbiome compositions with complications of end stage liver disease, including death9,16,22,23. Different commensal taxa are implicated in these studies, but in general, patients with higher burdens of potentially pathogenic taxa (e.g. Proteobacteria or Enterococcus) and lower prevalence of obligate anaerobic commensals (e.g. Lachnospiraceae and Ruminococcus) have a poor prognosis. This general principle is consistent with studies in non-hepatic diseases as well, including for patients undergoing bone marrow transplantation and patients with respiratory failure from COVID-1924–26.
While some studies have identified a potential difference in microbiome composition, the nature of these changes and functional consequences through microbial-derived metabolites (i.e. “the metabolome”) remain unaddressed. Microbe-derived metabolites contribute to intestinal epithelial cell differentiation and barrier formation27–29 and also regulate mucosal innate and adaptive immune defenses 26,30,31. In the case of liver disease, fecal bile acid (BA) profiles have been correlated with progression of NAFLD to non-alcoholic steatohepatitis (NASH) and subsequently advanced fibrosis32–34. Additionally, both total serum bile acids and specific immunomodulatory circulating BA have been implicated in the progression and prognosis of liver disease35–37. While recent studies have identified bacterial species that generate health promoting metabolites, whether these reduce or enhance progression of chronic liver disease remains largely unexplored territory.
Systemic infections precipitate hepatic decompensation and contribute to morbidity and mortality in liver disease patients38,39. The gut microbiome has been implicated in the development of systemic infections in many disease states24,40, including cirrhosis41. A recent prospective study used untargeted serum metabolites and fecal 16S rRNA microbiota data to predict nosocomial infections42; however, many of the serum metabolites of interest were not microbially derived and did not substantially improve the ability to predict infection development beyond standard clinical metrics (including known infection, model of end stage liver disease (MELD) score, and leukocyte count)42. Antibiotics, while highly effective for treatment and prophylaxis against common infections in liver disease43,44, are associated with increasing antibiotic resistance genes in the gut microbiomes and subsequent poor clinical outcomes45–48. Small studies of probiotics and fecal microbiota transfer (FMT) have been performed primarily as a means to control HE with overall positive clinical results including improved cognitive tests and fewer episodes of overt HE 49–53. However, while generally safe, there are reports of FMT-mediated transmission of toxin-producing and drug-resistant E. coli to patients, including one treated for HE54,55. Thus, FMT has not been adopted as a current standard of care56.
To better understand the role of the gut microbiome in the development of CLD complications, including development of systemic infections, we performed a single center observational cohort study of hospitalized patients with liver disease. Metagenomic and metabolomic profiles of fecal samples obtained from hospitalized patients were correlated with liver disease outcomes. We demonstrate that the commonly prescribed disaccharide lactulose preferentially expands Bifidobacteria and, in the absence of systemic antibiotic administration, results in a protective fecal metabolome. Functional Bifidobacteria expansion is associated with decreased abundance of antibiotic-resistant pathobionts and improved patient outcomes, including reduced incidence of systemic infections and prolonged survival in hospitalized patients. Our study provides novel insights into the mechanism by which lactulose impacts outcome of patients with CLD and provides a rationale for optimizing gut microbiome compositions and functions to minimize the complications of liver disease.
METHODS
Study Design
This was a prospective cohort study of consecutive hospitalized adult hepatology patients at a single institution from April 2021 to April 2022. Inclusion criteria were age 218 years, ability to provide informed consent (either themselves or by proxy if unable to provide consent), and being treated on the hepatology consult service. Subjects who were younger than 18 years, unable to provide consent, had prior solid organ transplant, or a prior colectomy were excluded. Patients were enrolled as soon as possible upon hospital admission, most within 48 hours. Samples were obtained under a protocol approved by the Institutional Review Board at the University of Chicago (IRB21-0327). Written informed consent was obtained from all participants or their surrogate decision makers.
Specimen Collection and Analysis
Fecal samples were collected as soon as possible following study enrollment. If able to provide additional samples, fecal samples were collected approximately every 2 days during hospitalization. Samples were collected in a similar manner on re-hospitalization up until 1 year post-enrollment or until death or transplant. Demographic and clinical data were collected through review of the medical record and through the University of Chicago Center for Research Informatics (CRI). Clinical laboratory values, death and transplant information was obtained by chart review. Ascitic fluid infections without an evident intraabdominal source were all termed spontaneous bacterial peritonitis (SBP) and were diagnosed with the standard clinical definition of either polymorphonuclear cells (PMN) of 250cells/mm3 or greater or a positive ascites culture43,44. Cultures growing common contaminants (i.e. components of skin flora) were considered negative if PMN were < 250cells/mm3 and the clinical team did not treat for SBP. Samples were paired with the closest stool sample that was within 14 days prior to the ascitic sample or 3 days after the ascitic sample. If a sample was positive, all subsequent samples for the next 30 days were considered surveillance paracenteses and excluded from analysis. If a sample was negative, subsequent samples, which were typically done to for volume management, were also excluded. This approach mitigated observing the effects of targeted antibiotic use on the microbiome after diagnosis of SBP, over-representing a single patient for repeated negative paracenteses, and ensured that each analyzed stool sample was paired with only one ascites sample. Blood stream infection was defined as having a blood culture with bacterial or fungal growth. Skin contaminants were again excluded (i.e. considered “negative” cultures) if the clinical team did not treat for bacteremia. Blood cultures were paired to the closest stool sample within 30 days prior or 7 days after the blood culture. Blood cultures 30 days after a positive culture (i.e. “surveillance cultures”) were excluded from analysis, and blood cultures drawn within 30 days after a negative culture were also excluded as “duplicates.”
Metagenomic Analyses
Fecal samples underwent shotgun DNA sequencing. After undergoing mechanical disruptions with a bead beater (BioSpec Product), samples were further purified with QIAamp mini spin columns (Qiagen). Purified DNA was quantified with a Qubit 2.0 fluorometer and sequenced on the Illumina HiSeq platform, producing around 7 to 8 million PE reads per sample with read length of 150 bp. Adapters were trimmed off from the raw reads, and their quality were assessed and controlled using Trimmomatic (v.0.39)57, then human genome were removed by kneaddata (v0.7.10, https://github.com/biobakery/kneaddata). Taxonomy was profiled using metaphlan4 using the resultant high-quality reads58. Microbial reads then were assembled using MEGAHIT (v1.2.9)59, genes are called by prodigal (https://github.com/hyattpd/Prodigal). In addition, high-quality reads are queried against genes of interest, such as virulence factors, cazymes, and antibiotic resistance genes, using DIAMOND (v2.0.4)60, and hits are filtered with threshold > 80% identity, > 80% protein coverage, then abundance is tabulated into counts per million or reads per million mapped reads (RPKM).
Alpha-diversity of fecal samples was estimated using the Inverse Simpson Index, while beta diversity was assessed by using taxumap (https://github.com/jsevo/taxumap). Metagenomic information is publicly available on NCBI under BioProject ID PRJNA912122.
Mouse fecal DNA isolation
DNA was extracted using the QIAamp PowerFecal Pro DNA kit (Qiagen). Prior to extraction, samples were subjected to mechanical disruption using a bead beating method. Briefly, samples were suspended in a bead tube (Qiagen) along with lysis buffer and loaded on a bead mill homogenizer (Fisherbrand). Samples were then centrifuged, and supernatant was resuspended in a reagent that effectively removed inhibitors. DNA was then purified routinely using a spin column filter membrane and quantified using Qubit.
16S sequencing
V4-V5 region within 16S rRNA gene was amplified using universal bacterial primers – 563F (5’-nnnnnnnn-NNNNNNNNNNNN-AYTGGGYDTAAA-GNG-3’) and 926R (5’-nnnnnnnn-NNNNNNNNNNNN-CCGTCAATTYHT-TTRAGT-3’), where ‘N’ represents the barcodes, ‘n’ are additional nucleotides added to offset primer sequencing. Approximately ∼412bp region amplicons were then purified using a spin column-based method (Minelute,Qiagen), quantified, and pooled at equimolar concentrations. Illumina sequencing-compatible Unique Dual Index (UDI) adapters were ligated onto the pools using the QIAseq 1-step amplicon library kit (Qiagen). Library QC was performed using Qubit and Tapestation and sequenced on Illumina MiSeq platform to generate 2×250bp reads.
16S qPCR
Extracted DNA was diluted to 20ng/ul to ensure concentrations fell within measurable range. Degenerate primers were diluted to 5.5mM concentration. Primer sequences are as follows: 563F (5’-AYTGGGYDTAAAGNG-3’) and 926Rb (5’-CCGTCAATTYHTTTRAGT-3’). Standard curves were generates using linearized TOPO pcr2.1TA vector (containing V4-V5 region of the 16S rRNA gene) transformed into DH5α competent bacterial cells. Five-fold serial dilution was performed on the purified plasmid from 108 to 103 copies/□l per tube. PCR products were detected using PowerTrack SYBR Green Master Mix (A46109). qPCR was run on QuantStudio 6 Pro (Applied Biosystems) with the following cycling conditions: 95°C for 10 min, followed by 40 cycles of 95°C for 30 s, 52°C for 30s, and 72°C for 1min. Copy numbers for samples were calculated using the Design and Analysis v2 software
Metabolomic Analyses
Short chain fatty acids (SCFA, i.e. butyrate, acetate, propionate, and succinate) were derivatized with pentafluorobenzyl bromide (PFBBr) and analyzed via negative ion collision induced-gas chromatography-mass spectrometry ([–]CI-GC-MS, Agilent 8890)61. Eight bile acids (BA, i.e. primary: cholic acid; conjugated primary: glycocholic acid, taurocholic acid; secondary: deoxycholic acid, lithocholic acid [LCA], isodeoxycholic acid; modified secondary: alloisolithocholic acid [alloisoLCA] and 3-oxolithocholic acid [3-oxoLCA]) were quantified (µg/mL) by negative mode liquid chromatography-electrospray ionization-quadrupole time-of-flight-MS ([–]LC-ESI-QTOF-MS, Agilent 6546). Eleven indole metabolites were quantified by UPLC-QqQ LC-MS. Eighty-five additional compounds were relatively quantified using normalized peak areas relative to internal standards. Data analysis was performed using MassHunter Quantitative Analysis software (version B.10, Agilent Technologies) and confirmed by comparison to authentic standards. Quantitative fecal metabolomic information paired to fecal metagenomic information is publicly available on NCBI under BioProject ID PRJNA912122. Raw data files are publicly available on MetaboLights project ID MTBLS7046.
Bacterial culture
The Bifidobacteria longum strains MSK.11.12 and DFI.2.45 were previously derived from two distinct healthy donor stool samples and whole genome sequenced (BioSample ID: SAMN19731851 and SAMN22167409). The vancomycin resistant Enterococcus faecium (VRE) strain used in this study was obtained from ATCC (strain 700221). Both bacterial strains were grown in anaerobic conditions in Brain-heart infusion broth (BHI broth, BD 237500). The pH was adjusted to 7.0 with NaOH. Media was supplemented with lactulose (Thermo Scientific, J60160-22) or sucrose (Fisher BP220-1) where indicated. For growth in media supplemented with short chain fatty acids, media was supplemented with either sodium butyrate (Sigma 303410), sodium succinate (Sigma S2378) or sodium acetate (Sigma S5636). For studies of bile acid metabolism, 10μg/mL of conjugated primary bile acid, either glycocholic acid (Sigma T4009) or taurocholic acid (EMD Millipore, 360512), was added to the media. All growth curves were obtained in anaerobic culture conditions at 37°C on a BioTek EPOCH2 microplate reader with BioTek Gen 5 3.11 software. Growth curves were analyzed in GraphPad Prism Version 9.4.0. Lactulose concentrations in were measured using the EnzyChrom™ Lactulose Assay Kit (ELTL-100).
Mouse studies
All mouse studies were approved by The University of Chicago Institutional Animal Care and Use Committee (IACUC, Protocol 72599). For germ-free studies, 8-12 week-old male C57BL/6 mice were used. Mice were bred and raised in a germ-free isolator, and after removal from the germ-free isolator, mice were handled in a sterile manner and individually housed in sealed negative pressure (BCU) isolators. Mice were treated with either regular sterile water or sterile water supplemented with filter sterilized lactulose at a final concentration of 20g lactulose/L of water. B. longum was grown to steady state in BHIS, pelleted, and resuspended in an equal volume of PBS, and previously germ-free mice were gavaged with 200μL of a freshly prepared suspension on 3 consecutive days. Fecal pellets were collected prior to and after B. longum inoculation at the indicated timepoints for 16S rRNA metagenomic analysis and targeted metabolomic analysis.
Data sharing
In addition to the repositories specified above, all raw data included in the manuscript is publicly available at: https://github.com/yingeddi2008/DFILiverDiseaseMicrobiome.
RESULTS
Patient recruitment and disease characteristics
We enrolled 356 hospitalized patients with liver disease from April 2021 through April 2022. Of these 356 patients, 263 (73.8%) produced 850 stool samples that were analyzed by shotgun metagenomics and targeted metabolomics. The demographics and admission disease characteristics of patients who produced samples are shown in Table 1. Notably, alcohol use was the most common etiology of liver disease and accounted for 51.0% of patients. Patients had a median model of end stage liver disease MELD-sodium (MELD-Na) score of 18.69 (IQR 10.61 – 27.88). Of the 263 patients included in the analysis, 183 (69.6%) had known clinically significant portal hypertension upon hospitalization, which was defined either by hepatic venous pressure gradient (HVPG) ≥ 10mmHg, characteristic findings on imaging (enlarged portal vein, intraabdominal varices, splenomegaly, or ascites), or clinical features of ascites, varices, or hepatic encephalopathy. Over half (137, 52.1%) of the enrolled patients had some form of end organ dysfunction present on admission. End organ dysfunction included neurologic dysfunction (grade III or higher HE), circulatory dysfunction (mean arterial pressure < 60mmHg or requiring vasoactive agents), respiratory dysfunction (new or increasing oxygen requirement), or renal dysfunction (acute kidney injury or new hemodialysis needs). Taken together, these enrollment demographics and baseline disease characteristics indicate that, on average, this patient cohort has advanced liver disease with a poor prognosis.
Fecal microbiome and metabolite profiles of liver disease patients are highly variable
To determine the range of fecal microbiome compositions in patients with liver disease, we performed shotgun sequencing on DNA extracted from 850 fecal samples collected from 263 liver disease patients. We used MetaPhlan4 to assign taxonomic compositions and Bray-Curtis distance to assess microbial diversity within the resulting dataset (Figure 1A). Immediately apparent is the low taxonomic alpha diversity in the majority of fecal samples (inverse Simpson range: 1.00 – 36.41, mean = 6.22, median = 3.86) and the expansion of bacterial taxa belonging to the Enterococcus genus (forest green) and Enterobacteriaceae family (red), which are common hallmarks of dysbiosis and are similar to microbiome domination detected in patients undergoing allogeneic hematopoietic cell transplantation24,62. Another striking observation is the high number of samples with marked expansion of the genus Bifidobacteria (shown in purple), which is commonly reported in breast fed infants but rarely seen in adults, suggesting that this may be a liver disease-specific finding.
While microbiome-derived or modified metabolites mediate many of the beneficial impacts on mucosal immune defenses and epithelial barrier functions, little is known about their production in liver disease patients harboring vastly different microbial populations. We used targeted GC- and LC-MS platforms to determine relative amounts 82 metabolites in all 850 fecal samples within our dataset, and the first sample from each patient is shown (Figure S1). Ranking of fecal samples from lowest to highest alpha diversity demonstrates the correlation between microbiome diversity and the relative amounts of SCFAs, branched chain FAs, aminated FAs, secondary bile acids, and various indole compounds and tryptophan metabolites. As expected, low diversity fecal samples have markedly reduced amounts of secondary bile acids but also reduced concentrations of SCFAs and a subset of indole compounds.
In order to obtain a more quantitative picture of fecal metabolite concentrations, we measured the concentration of SCFAs and primary and secondary bile acids in fecal samples from patients with liver disease. Despite some variation, higher proportions of commensal anaerobes and alpha-diversity generally coincided with greater levels of all measured short chain fatty acids (towards the right side of Figure 1B). Additionally, samples with more commensal anaerobes tended to have greater capacity to deconjugate primary bile acids and generate both secondary and modified secondary bile acids (Figure 1C). These findings demonstrate that the fecal microbiome and metabolome is highly variable in hospitalized patients with liver disease.
Fecal metabolites correlate with expansion of distinct microbial taxa
To more precisely associate fecal metabolite concentrations with fecal microbiome compositions, fecal samples were assigned to 1 of 12 taxonomic groups on a taxonomic Uniform Manifold Approximation and Projection (taxUMAP) (Fig 2A). While all clusters displayed a wide range of short chain fatty acid production, our analysis presented in Figure 2 revealed that clusters that were characterized by samples with Enterococcus (forest green), Proteobacteria (red), or Lactobacillus (royal blue) as the most abundant taxa all had lower levels of short chain fatty acid production when compared to samples characterized by expansion of Bifidobacteria (purple), Lachnospiraceae (pink) or Bacteroidetes (aquamarine) (Fig 2A-D, S2A-C). On average, Bifidobacteria-expanded samples, which are prevalent in patients with liver disease, had higher acetate production (Fig 2B, S2A) but similar levels of butyrate (Fig 2C, S2B) and propionate (Fig 2D, S2C) compared to samples with high Bacteroidetes or Lachnospiraceae abundance. Consistent with a role for Bifidobacteria in acetate production, samples with greater than 10% Bifidobacteria abundance had a positive and statistically significant linear correlation with increasing acetate but not butyrate (Fig S2A-B, right-most panels). Propionate concentration negatively correlated with Bifidobacteria abundance (Fig S2C, right-most panel).
While all groups had detectable concentrations of primary bile acids, Enterococcus dominated samples had the highest (Fig S3). In contrast, conjugated primary bile acid levels were lower (Fig 2E, S3) and secondary (Fig 2F, S3) and modified secondary BA (Fig 2G, S3) levels were higher in samples with high levels of obligate anaerobes, including Bifidobacteria, Bacteroidetes, and Lachnospiraceae. Samples dominated by Bifidobacteria had higher levels of primary bile acids and lower levels of secondary bile acids compared to Bacteroidetes- and Lachnospiraceae-dominated samples, suggesting Bifidobacteria provide increased bile acid hydrolase activity and/or reduced efficiency of primary BA to secondary BA conversion. Taken together, our results indicate that a subset of patients with liver disease are predisposed to Bifidobacteria expansion with resultant acetate production and conjugated bile salt hydrolysis with reduced efficiency of secondary bile acid generation.
Lactulose administration is associated with increased Bifidobacteria abundance and reduced antibiotic-resistant pathogen density
As lactulose is a common therapy administered to liver disease patients and has been reported to act as a prebiotic, we next sought to address how it might be impacting microbiome composition. Fecal samples obtained from liver disease patients after lactulose administration (lactulose given within 7 days prior to fecal sample) were taxonomically diverse, with many having marked Bifidobacteria expansion. In many samples, Bifidobacteria were the predominant genus (Fig 3A, B; range: 0.0 – 94.7%, mean: 10.9%, stdev: 10.9%). This contrasted with fecal samples obtained from patients who were not treated with lactulose (Fig 3A, B; range: 0.0 – 50.1%, mean: 3.57%, stdev: 3.57%, p < 0.001). High abundances of potentially pathogenic bacteria including Enterobacteriaceae and Enterococcus were detected in lactulose treated and untreated patients (Fig 3A). Enterococcus expansion was largely restricted to low-Bifidobacteria abundance samples and was associated with the presence of the vancomycin-resistance gene vanA, a result that was more striking in samples obtained in the presence of lactulose (Fig 3C). These results suggest that lactulose-mediated Bifidobacteria expansion may limit the expansion of Vancomycin-resistant Enterococcus (VRE) in the gut. Alternatively, treatment with broad-spectrum antibiotics may reduce the density of Bifidobacterium species while also eliminating other commensal species mediating resistance to VRE in lactulose-treated patients (Fig 3B) 24,63. Although the osmotic laxative effect of lactulose may affect the fecal metagenomic composition, we were unable to detect an association between stool consistency and Bifidobacteria abundance (Supplemental Data).
Lactulose-mediated Bifidobacteria expansion is associated with increased levels of multiple bioactive fecal metabolites
Bifidobacteria expansion, defined here as relative abundance ≥10%, was associated with marked changes in fecal metabolite profiles only in samples obtained in the presence of lactulose (Fig 4, S4). In lactulose-treated patients with Bifidobacteria expansion, the fecal metabolome was characterized by increased SCFA concentrations (including acetate, butyrate, and propionate), reduced conjugated primary bile acid concentrations (taurocholic, glycocholic, taurochenodeoxycholic, and glycochenodeoxycholic acids), increased secondary (lithocholic, isodeoxycholic acid) and modified secondary (alloisolithocholic, 3-oxo-lithocholic acid) bile acids, and indole metabolites (indole-3-lactic acid and indole-3-carboxaldehyde) (Fig 4A-C). Consistent with a known property of Bifidobacteria to hydrolyze conjugated primary bile acids, ratios of primary bile acids to conjugated primary bile acids were substantially higher in lactulose-treated samples with Bifidobacteria expansion (Fig 4D). While secondary bile acid levels were also higher in these samples, this was primarily due to an increased capacity to deconjugate primary bile acids rather than convert primary BA to secondary BA (Fig 4D, middle and right-most panels). In the absence of lactulose treatment, fecal samples with greater than 10% Bifidobacteria also had increased concentrations of SCFA (acetate and butyrate) and decreased conjugated primary bile acids (Fig S4). However, these changes were all modest compared to those from patients receiving lactulose, suggesting that lactulose impacts the metabolic activities of the gut microbiome in patients with liver disease (Fig S4).
Lactulose enhances Bifidobacteria longum growth, acetate production and inhibition of VRE
Many fecal samples obtained from lactulose treated patients had high abundances of potentially pathogenic bacteria belonging to Enterobacteriaceae family (range: 0.0 – 99.2%, mean: 8.04%, median: 0.73%, stdev: 8.04%) and the Enterococcus genus (range: 0.0 – 100%, mean: 30.8%, median: 5.24%, stdev: 30.8%), with, on average, higher densities than those seen in fecal samples taken from patients not receiving lactulose (Proteobacteria mean: 6.38%, median: 1.25%; Enterococcus mean: 7.4%, median: 0.06%). While high abundances of Proteobacteria species were seen throughout a range of Bifidobacteria abundances, Enterococcus domination was seen predominantly in samples with reduced to absent Bifidobacteria abundance (Fig 3A, 5A).
To further investigate the inverse relationship between Bifidobacteria and VRE abundance, we isolated a Bifidobacterium longum strain from a healthy human donor and performed whole genome sequencing to demonstrate the presence of genes involved in lactulose metabolism and acetate production64,65. B. longum growth in culture was markedly augmented by addition of lactulose to media, and to a lesser extent by addition of sucrose (Fig 5B). Lactulose-induced growth augmentation was accompanied by increased media acidification and acetate, but not butyrate or propionate, production (Fig S5A, B). Similar to patient fecal samples dominated by Bifidobacteria and consistent with coding for bile salt hydrolase genes, monocultures of B. longum efficiently deconjugated conjugated primary bile acids but did not convert these to secondary bile acids (Fig S5C, D).
We next colonized germ-free (GF) mice with B. longum in the presence or absence of lactulose (Fig S6A). Lactulose was detectable in the stool of GF mice and led to increase stool water content (Fig S6B, C). B. longum efficiently colonized and persisted in the intestines of gnotobiotic mice independent of lactulose (Fig S6D). Consistent with in vitro functions, B. longum-colonized mice had measurable fecal acetate levels but did not generate butyrate or propionate (Fig S6E-G). Fecal acetate levels were similar between water and lactulose-treated mice at day 0, but on day 10 lactulose augmented acetate production (Fig S6E). On day 10 after B. longum colonization, fecal bile acid profiling demonstrated increased primary bile acids but undetectable secondary BA (Fig S6H-J).
To test whether B. longum inhibits VRE growth and to determine the impact of lactulose administration, we performed co-culture assays (Fig 5, C-D). While simultaneous inoculation of B. longum and VRE into media did not reduce VRE growth (Fig 5C-D), pre-culture of B. longum for 24h or 48h fully inhibited VRE growth in the presence of lactulose and only partially inhibited VRE growth in the absence of lactulose (Fig 5C-D). Serial dilutions of B. longum culture supernatant with fresh media sequentially reduced the inhibitory effect on VRE growth (Fig S7A, B), and alkalization of B. longum supernatant pH from 5.8 up to 7.2 restored VRE’s initial log phase growth (Fig S7C) but did not enable VRE to reach the maximal OD600 obtained in fresh, neutral pH media (Fig S7C). VRE growth is inhibited by addition of short chain fatty acids to the media, and inhibition is augmented at lower media pH (Fig S7D). Taken together, these data suggest that B. longum inhibits VRE growth through parallel mechanisms that include media acidification, SCFA production, and, to a lesser extent, nutrient deprivation. B. longum-mediated VRE inhibition is significantly augmented by lactulose, potentially explaining the expansion of VRE seen in fecal samples lacking expanded Bifidobacteria populations despite lactulose administration. Consistent with VRE expansion seen in patients treated with lactulose without Bifidobacteria expansion, VRE growth is also augmented by lactulose (Fig S7E,F).
Bifidobacteria expansion and associated metabolite production correlates with reduced incidence of infections in advanced liver disease
To determine whether lactulose-mediated Bifidobacteria expansion and associated metabolite changes are associated with clinical benefits, we correlated the gut microbiome metagenomic and metabolic data with development of common infections in cirrhosis, including spontaneous bacterial peritonitis (SBP) and bacteremia (Figs 6, S8, and S9).
We identified 111 ascites samples with near concurrent fecal samples for analysis, among which there were 19 diagnoses of SBP (Fig 6A, B, S8A, Table S1). LEfSe analysis demonstrated that Bifidobacteria was strongly associated with resistance to SBP, while presence of the mucus-degrading Akkermansia was associated with SBP (Fig 6A). We used a frequency cutoff of 10% to distinguish patients with high Bifidobacteria from those with low Bifidobacteria frequencies. We found that 1 of 29 patients (3.4%) with ≥10% Bifidobacteria frequency was diagnosed with SBP while 18 of 63 patients (28.6%) with less than 10% Bifidobacteria developed SBP (Fig 6B; Odds Ratio 0.12, 95% CI 0.01 – 0.79, p value 0.021). Qualitative metabolomics demonstrated that glycholithocholic acid and serotonin were significantly associated with being SBP-free (Fig S8B). The Bifidobacteria-generated SCFA acetate was also relatively increased in SBP-free samples, albeit to a lesser extent (p < 0.05, log2 fold-change 0.9, Fig S8B). Quantitative metabolomics demonstrated that acetate was significantly higher in SBP-free fecal samples and the conjugated primary bile acid taurocholic acid was significantly higher in samples with SBP (Fig 6C). Consistent with a role for Bifidobacteria in protection from SBP, samples from patients without SBP had a significantly greater conversion of conjugated to unconjugated primary BA (CA:GCA ratio) but similar conversion of primary to secondary BA (Fig S8D).
To correlate fecal microbiome compositions and metabolite concentrations with development of bacteremia, we paired 328 blood cultures with fecal samples for analysis. Blood culturing identified 23 distinct diagnoses of bacteremia (Fig 6D, E, S9A, Table S2) and LEfSe analysis demonstrated that Bifidobacteria was associated with remaining bacteremia-free (Fig 6D). There were 2 diagnoses of bacteremia in 57 patients (3.5%) with ≥10% Bifidobacteria and 21 of 148 patients (14.2%) with <10% Bifidobacteria (Fig 6E; Odds Ratio 0.247, 95% CI 0.056 – 0.996, p value 0.047). Qualitative metabolomics demonstrated that conjugated primary BA (glycocholic, taurocholic, and glycochenodeoxycholic acid) and the primary BA cholic acid were all significantly increased in bacteremia-associated samples (Fig S9B). Targeted quantitative metabolomics confirmed that samples from bacteremic patients had reduced capacity to hydrolyze conjugated bile acids (Fig 6F, S9D). These data suggest that lactulose-mediated Bifidobacteria expansion with resultant SCFA production and bile salt hydrolase activity may play a protective role against some of the most common and devastating infectious complications of advanced liver disease.
Lactulose-mediated gut Bifidobacteria expansion is associated with improved transplant-free survival
Finally, we determined whether initial stool characteristics can identify patients at increased risk of adverse outcomes and thus might serve as targets for microbiome-based therapies. To do so, we assessed 90-day overall survival in patients stratified by lactulose exposure and Bifidobacteria expansion. Compared to patients who received lactulose but did not expand Bifidobacteria, patients with lactulose-associated Bifidobacteria expansion had significantly improved overall 90-day survival (Fig 6G) with similar rates of liver transplantation (Fig S10). While many patient characteristics between the groups were similar (including age, race, BMI, cause of liver disease, and presence of clinically significant portal hypertension), patients who received lactulose but did not expand Bifidobacteria had clinical parameters that correlate with a poor prognosis (including higher MELD-Na scores and higher incidence of additional organ failures present upon hospitalization) (Table 3). This suggests that Bifidobacteria expansion and associated metabolite profiles can serve as a biomarker for healthier patient cohort, which may be due to the gut microbiome differences along with other disease-specific factors.
DISCUSSION
Lactulose has been used to treat hepatic encephalopathy for over 50 years66. Despite widespread use and guideline recommendations for HE treatment from medical societies, its mechanism of action is incompletely understood14,67. While lactulose is postulated to provide benefits by decreasing bowel transit time and gut lumen acidification, thereby decreasing ammonia absorption, its role in altering the microbiome has remained largely undefined. Herein, we demonstrate that in patients with liver disease, lactulose administration, in the absence of broad-spectrum antibiotic administration, leads to expansion of Bifidobacteria species. This expansion results in a distinct gut microbiome taxonomic and metabolic profile and is associated with exclusion of antibiotic-resistant pathobionts. Bifidobacteria inhibit growth of antibiotic-resistant Enterococcal and Enterobacterales species, effects that are augmented by lactulose. Our findings suggest a protective role of lactulose-mediated Bifidobacteria expansion in patients with liver disease. Consistent with this, Bifidobacteria expansion and associated metabolites are associated with reduced incidence of SBP and bacteremia as well as improved 90-day survival.
In addition to treating precipitating factors, such as systemic infection, lactulose has largely replaced dietary modification, antibiotics, and laxatives as the first line treatment for hepatic encephalopathy. All HE therapies are aimed at reducing gut ammonia production and absorption, and the initial reports of lactulose use for HE concluded that this is due to luminal acidification66,68. It has also been assumed that, similar to healthy subjects and experimental models 12,69,70, lactulose exhibits a prebiotic effect, but this has not been shown in patients with liver disease7,10,66,71. The authors of the initial trial of lactulose were unable to detect changes in fecal bacteria66; however, they were limited by culture-based techniques. More recent studies characterizing the microbiomes of cirrhotic patients did not reveal Bifidobacteria expansion in patients treated with lactulose 9, most likely because 16S rRNA sequencing platforms often do not amplify Bifidobacterium 16S rRNA genes72. Along similar lines, lactulose withdrawal demonstrated only slight decreases in Faecalibacterium species using sequencing techniques10. Our study avoided 16S rRNA amplification bias by metagenomically sequencing fecal samples and demonstrates marked expansion of Bifidobacteria in a large subset of cirrhotic patients treated with lactulose.
Bifidobacterium species are dominant fecal microbes in breast fed infants and are commonly thought of as prototypical health-promoting bacteria73. Bifidobacterium species imprint the human immune system74, are associated with decreased development of atopy and autoimmune diseases75, and administration decreases rates of necrotizing enterocolitis (NEC) in preterm infants76. In adults, fecal Bifidobacteria are thought to modulate anti-tumor immunity and enhance immunotherapy efficacy in humans with melanoma and synergize with immunotherapies to reduce melanoma growth in mouse models77,78. Bifidobacteria also produce acetate and lactic acid, which antagonize pathogens to reduce the incidence of enteric infections in infants. While Bifidobacteria does not provide colonization resistance to pathogenic E. coli in a murine model, specific acetate-producing Bifidobacteria strains limit systemic disease by improving mucosal integrity via acetate promotion of epithelial integrity and from heightened immune surveillance64.Gut acidification, mucosal barrier enhancement, and immunomodulatory effects of Bifidobacteria may all benefit patients with liver disease, who are immunocompromised and at high risk of enteric pathogen colonization, growth and dissemination.
While Bifidobacteria attenuate liver disease progression in rodent models79,80, our study suggests that Bifidobacteria may reduce the complications of severe liver disease and thus may enhance approaches for supportive care. We report an association between Bifidobacteria expansion and reduced incidence of systemic infection and prolonged patient survival. This beneficial association has multiple plausible explanations that may be driven by the unique metabolite profile including increased acetate levels and enhanced hydrolysis of conjugated primary bile acids. Similar metabolite profiles appear to be protective against both SBP and bacteremia. Short chain fatty acid production has been linked to gut colonization resistance of common gram-negative enteric pathogens81. Similarly, Bifidobacteria expansion was associated with reduced abundance of the common gram-positive pathogen, VRE in our patients, and inhibited VRE growth in vitro, likely via both media acidification and SCFA production. Bifidobacteria-mediated VRE growth inhibition was significantly enhanced by lactulose, which also significantly increased SCFA production and acidification in vitro and acetate production in both mice and patients. While colonization and expansion are important initial steps in infection development, SCFAs are also known to promote intestinal barrier function by enhancing both epithelial integrity and mucosal immune function, thereby limiting pathogen translocation 28,29,64,82. Moreover, Bifidobacteria are associated with decreased intestinal permeability in patients with alcohol dependence, which is extremely common in our study population83. In our study, Bifidobacteria-expanded samples also had significantly higher levels of indole-3-carboxaldehyde, which activates the aryl hydrocarbon receptor to increase mucosal IL-22 production and maintain mucosal reactivity towards pathogens.
Hospitalized patients with liver disease are commonly treated with broad spectrum antibiotics with accompanying marked microbiome derangements, and our findings will aid in identifying patients who may benefit from targeted microbiome therapy. Success of a biotherapeutic approach to supportive care likely depends on both the abundance of Bifidobacteria in the patient’s gut at the time of treatment initiation and co-administration of a prebiotic, such as lactulose65. In subjects without liver disease, both prebiotic and probiotic approaches result in Bifidobacteria expansion, and a synbiotic approach prolongs functional Bifidobacteria engraftment12,84,85. In outpatients with liver disease, synbiotic approaches with multiple lactic acid bacteria and fermentable fiber sources reduce the blood ammonia levels, endotoxinemia, and the incidence of hepatic encephalopathy86,87. These studies do not assess the durability or metabolic function of the administered probiotic or correlate use with other outcomes in liver disease. Another important consideration that has not been well-studied even in healthy adults is the functional (i.e. metabolomic) change that marked Bifidobacteria expansion has on the gut microbiome.
While associated with improved outcomes in our population, a Bifidobacteria-expanded microbiome is not reflective of a normal, healthy adult microbiome with regards to microbial composition or metabolite profiles. While acetate has been shown to prevent gut pathogen dissemination in a murine model, butyrate is better studied with regard to epithelial barrier function and mucosal immune function28,29,31,64. Secondary bile acids are not only often considered markers of a “healthy” gut microbiome but have also been implicated in prevention of enteric infections40 and reducing intestinal inflammation88. This will also be an important consideration in rational probiotic therapy design, and supplementation with multiple bacterial species with complementary metabolite production is likely the preferred approach. Conversely, Bifidobacteria are an important component of the developing human gut microbiome. Lactulose-mediated Bifidobacteria expansion may therefore be a stepping-stone to allow for expansion and engraftment of additional beneficial commensal organisms and restoration of a microbiome more similar to healthy adults. A recent paper studying the effect of active alcohol consumption on the murine gut microbiome concluded that acetate produced during alcohol metabolism results in increased Bacteroidetes89, and it is plausible to think that Bifidobacteria-derived metabolites may also allow for commensal expansion.
In conclusion, we report here that lactulose-mediated gut Bifidobacteria expansion is associated with a unique fecal metagenomic and metabolic profile, reduced clinically-relevant antibiotic-resistant pathobiont burden, and improved clinical outcomes in hospitalized patients with liver disease, including reduced incidence of SBP and bacteremia and prolonged survival. Our study provides novel insights into the mechanism of action of a liberally prescribed medication and rationale for future studies designed to precisely target the gut microbiome to prevent complications of advanced liver disease.
Data Availability
In addition to the repositories specified below, all raw data included in the manuscript is publicly available at: https://github.com/yingeddi2008/DFILiverDiseaseMicrobiome. 1. Metagenomic information is publicly available on NCBI under BioProject ID PRJNA912122. 2. Quantitative fecal metabolomic information paired to fecal metagenomic information is publicly available on NCBI under BioProject ID PRJNA912122. 3. Raw data files are publicly available on MetaboLights project ID MTBLS7046.
GRAPHICAL ABSTRACT
SUPPLEMENTAL FIGURE LEGENDS
REFERENCES
- 1.↵
- 2.
- 3.
- 4.↵
- 5.↵
- 6.↵
- 7.↵
- 8.↵
- 9.↵
- 10.↵
- 11.
- 12.↵
- 13.↵
- 14.↵
- 15.↵
- 16.↵
- 17.↵
- 18.↵
- 19.
- 20.
- 21.↵
- 22.↵
- 23.↵
- 24.↵
- 25.
- 26.↵
- 27.↵
- 28.↵
- 29.↵
- 30.↵
- 31.↵
- 32.↵
- 33.
- 34.↵
- 35.↵
- 36.
- 37.↵
- 38.↵
- 39.↵
- 40.↵
- 41.↵
- 42.↵
- 43.↵
- 44.↵
- 45.↵
- 46.
- 47.
- 48.↵
- 49.↵
- 50.
- 51.
- 52.
- 53.↵
- 54.↵
- 55.↵
- 56.↵
- 57.↵
- 58.↵
- 59.↵
- 60.↵
- 61.↵
- 62.↵
- 63.↵
- 64.↵
- 65.↵
- 66.↵
- 67.↵
- 68.↵
- 69.↵
- 70.↵
- 71.↵
- 72.↵
- 73.↵
- 74.↵
- 75.↵
- 76.↵
- 77.↵
- 78.↵
- 79.↵
- 80.↵
- 81.↵
- 82.↵
- 83.↵
- 84.↵
- 85.↵
- 86.↵
- 87.↵
- 88.↵
- 89.↵