A Pilot Integrative Analysis of Colonic Gene Expression, Gut Microbiota, and Immune Infiltration in Primary Sclerosing Cholangitis-Inflammatory Bowel Disease: Association of Disease With Bile Acid Pathways

Mohammed Nabil Quraishi, Animesh Acharjee, Andrew D Beggs, Richard Horniblow, Chris Tselepis, Georgios Gkoutos, Subrata Ghosh, A E Rossiter, Nicholas Loman, Willem van Schaik, David Withers, Julian R F Walters, Gideon M Hirschfield, Tariq H Iqbal, Mohammed Nabil Quraishi, Animesh Acharjee, Andrew D Beggs, Richard Horniblow, Chris Tselepis, Georgios Gkoutos, Subrata Ghosh, A E Rossiter, Nicholas Loman, Willem van Schaik, David Withers, Julian R F Walters, Gideon M Hirschfield, Tariq H Iqbal

Abstract

Background: Although a majority of patients with PSC have colitis [PSC-IBD; primary sclerosing cholangitis-inflammatory bowel disease], this is phenotypically different from ulcerative colitis [UC]. We sought to define further the pathophysiological differences between PSC-IBD and UC, by applying a comparative and integrative approach to colonic gene expression, gut microbiota and immune infiltration data.

Methods: Colonic biopsies were collected from patients with PSC-IBD [n = 10], UC [n = 10], and healthy controls [HC; n = 10]. Shotgun RNA-sequencing for differentially expressed colonic mucosal genes [DEGs], 16S rRNA analysis for microbial profiling, and immunophenotyping were performed followed by multi-omic integration.

Results: The colonic transcriptome differed significantly between groups [p = 0.01]. Colonic transcriptomes from HC were different from both UC [1343 DEGs] and PSC-IBD [4312 DEGs]. Of these genes, only 939 had shared differential gene expression in both UC and PSC-IBD compared with HC. Imputed pathways were predominantly associated with upregulation of immune response and microbial defense in both disease cohorts compared with HC. There were 1692 DEGs between PSC-IBD and UC. Bile acid signalling pathways were upregulated in PSC-IBD compared with UC [p = 0.02]. Microbiota profiles were different between the three groups [p = 0.01]; with inferred function in PSC-IBD also being consistent with dysregulation of bile acid metabolism. Th17 cells and IL17-producing CD4 cells were increased in both PSC-IBD and UC when compared with HC [p < 0.05]. Multi-omic integration revealed networks involved in bile acid homeostasis and cancer regulation in PSC-IBD.

Conclusions: Colonic transcriptomic and microbiota analysis in PSC-IBD point toward dysregulation of colonic bile acid homeostasis compared with UC. This highlights important mechanisms and suggests the possibility of novel approaches in treating PSC-IBD.

Keywords: Autoimmune liver disease; bioinformatics; colitis; dysbiosis.

© European Crohn’s and Colitis Organisation 2020.

Figures

Figure 1.
Figure 1.
Principal component analysis [PCA] score plot performed on the mucosal transcriptome datasets demonstrating clustering of subjects within, and variation between, cohorts. Dots represents samples and are coloured according to the subject cohort. Ellipse represents 95% confidence. Results are plotted according to the PC1 and PC2 scores, with the percent variation explained by the respective axis. [a] PSC-IBD versus HC; [b] UC versus HC; [c] PSC-UC versus UC; [d] PCA plots demonstrating variation between HC, UC, and PSC-UC samples. The three groups were significantly different from each other [p = 0.01]. PSC, primary sclerosing cholangitis; IBD, inflammatory bowel disease; UC, ulcerative colitis; HC, healthy controls.
Figure 2.
Figure 2.
Differential gene expression profiles and volcano plots highlighting differentially expressed genes. [a] PSC-IBD versus HC; [b] UC versus HC; [c] PSC-IBD versus UC. Differentially expressed genes with logFC >1 are shown; 939 genes had shared differential expression in PSC-IBD and UC when compared with healthy controls [588 upregulated; 351 downregulated]. PSC, primary sclerosing cholangitis; IBD, inflammatory bowel disease; UC, ulcerative colitis; HC, healthy controls.
Figure 3.
Figure 3.
Bioinformatic analysis identifies pathways including bile acid signalling as relevant to disease distinctions. [a] Detailed enrichment analysis of gene clusters in functionally grouped network. Gene networks and pathways involving key nuclear receptors involved in bile acid homeostasis such as FXR, PPAR, conjugation, binding, and transport are significantly upregulated in PSC-IBD compared with controls; [b] top 20 gene ontology biological processes PSC-IBD vs UC demonstrate metabolic pathways, many of which are involved in bile acid homeostasis are upregulated in PSC-IBD compared with UC, whereas immune activation and defense pathways are upregulated in UC compared with PSC-IBD. PSC, primary sclerosing cholangitis; IBD, inflammatory bowel disease; UC, ulcerative colitis; HC.
Figure 4.
Figure 4.
Distinct microbiota taxa in patients with PSC-IBD. [a] Phylum level differences in the three cohorts—UC and PSC-IBD are characterised by a relative expansion of Proteobacteria and Fusobacteria and reduction of Bacteroidetes compared with HC; [b] microbial taxa comparing PSC-IBD and UC. Association of specific microbiota taxa with PSC-UC and UC by linear discriminant analysis [LDA] effect size [LEfSe]. Red indicates taxa enriched in PSC-IBD and green indicates taxa enriched in UC; [c] Cladogram representation of the gut microbial taxa associated with PSC-IBD and UC. PSC-IBD is associated with an increased abundance of bacteria expressing bile salt hydrolase and hydroxysteroid dehydrogenases [such as Bacteroides fragilis, Roseburia spp, Shewanella spp.,and Clostridium ramosum] in comparison with UC. PSC, primary sclerosing cholangitis; IBD, inflammatory bowel disease; UC, ulcerative colitis.
Figure 5.
Figure 5.
Functional classification of the predicted metagenome content of the microbiota of PSC-IBD compared with UC. [a] MetaCyc pathways; and [b] KEGG pathways. There is significant enrichment of primary bile acid biosynthesis [associated with significantly higher expression of bile salt hydrolase and hydroxysteroid dehydrogenases], pentose and glucoronate interconversions, and galactose metabolism in PSC-IBD compared with UC. PSC, primary sclerosing cholangitis; IBD, inflammatory bowel disease; UC, ulcerative colitis.
Figure 6.
Figure 6.
Correlation networks between the mucosal transcriptome, 16S microbial profiles, and immunophenotype in patients with PSC-IBD. The mucosal transcriptome is in red, 16S microbial profiles purple. Genes were selected based on supervised and unsupervised dimension reduction as described in Methods. Microbial taxa were selected based on the loading values more than log2[3] values. Th17- and IL17-expressing cells were selected as they were significantly upregulated in PSC-IBD and UC compared with HC. Red line indicates negative correlation and green positive correlation. Thickness of the line shows strength of the correlation between each features. Clusters are defined based on a high inter-feature correlation [r = 0.8]. Two main clusters were identified—cluster A consists of genes involved in bile acid homeostasis; cluster B demonstrates genes associated with cancer regulatory pathways. PSC, primary sclerosing cholangitis; IBD, inflammatory bowel disease; UC, ulcerative colitis; HC, healthy controls.

References

    1. Hirschfield GM, Karlsen TH, Lindor KD, Adams DH. Primary sclerosing cholangitis. Lancet 2013;382:1587–99.
    1. Torres J, Palmela C, Brito H, et al. . The gut microbiota, bile acids and their correlation in primary sclerosing cholangitis associated with inflammatory bowel disease. United European Gastroenterol J 2018;6:112–22.
    1. Kummen M, Holm K, Anmarkrud JA, et al. . The gut microbial profile in patients with primary sclerosing cholangitis is distinct from patients with ulcerative colitis without biliary disease and healthy controls. Gut 2017;66:611–9.
    1. Quraishi MN, Sergeant M, Kay G, et al. . The gut-adherent microbiota of PSC-IBD is distinct to that of IBD. Gut 2017;66:386–8.
    1. Ji SG, Juran BD, Mucha S, et al. ; UK-PSC Consortium; International IBD Genetics Consortium; International PSC Study Group Genome-wide association study of primary sclerosing cholangitis identifies new risk loci and quantifies the genetic relationship with inflammatory bowel disease. Nat Genet 2017;49:269–73.
    1. Ellinghaus D, Folseraas T, Holm K, et al. . Genome-wide association analysis in primary sclerosing cholangitis and ulcerative colitis identifies risk loci at GPR35 and TCF4. Hepatology 2013;58:1074–83.
    1. Liu JZ, Hov JR, Folseraas T, et al. ; UK-PSCSC Consortium; International PSC Study Group; International IBD Genetics Consortium Dense genotyping of immune-related disease regions identifies nine new risk loci for primary sclerosing cholangitis. Nat Genet 2013;45:670–5.
    1. Andrews S. FastQC: a quality control tool for high throughput sequence data 2010.. Accessed December 12, 2018.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 2014;30:2114–20.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012;9:357–9.
    1. Dobin A, Davis CA, Schlesinger F, et al. . STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21.
    1. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 2014;30:923–30.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010;26:139–40.
    1. Wu D, Smyth GK. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res 2012;40:e133.
    1. Bindea G, Mlecnik B, Hackl H, et al. . ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 2009;25:1091–3.
    1. Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol 2017;18:220.
    1. Earth Microbiome Project. Illumina 16s PCR protocols . Accessed January 18, 2019.
    1. Bolyen E, Rideout JR, Dillon MR, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol 2019;37:852–857.
    1. Quast C, Pruesse E, Yilmaz P, et al. . The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res 2013;41:D590–6.
    1. Segata N, Izard J, Waldron L, et al. . Metagenomic biomarker discovery and explanation. Genome Biol 2011;12:R60.
    1. Langille MGI. Exploring linkages between taxonomic and functional profiles of the human microbiome. mSystems 2018;3.
    1. Parks DH, Tyson GW, Hugenholtz P, Beiko RG. STAMP: statistical analysis of taxonomic and functional profiles. Bioinformatics 2014;30:3123–4.
    1. Breiman L. Random forests. Machine Learning 2001;45:5–32.
    1. Epskamp S, Cramer AOJ, Waldorp LJ, Schmittmann VD, Borsboom D. qgraph: Network visualizations of relationships in psychometric data. J Stat Softw 2012;48:1–18.
    1. Chen ML, Takeda K, Sundrud MS. Emerging roles of bile acids in mucosal immunity and inflammation. Mucosal Immunol 2019;1 10.1038/s41385-019-0162-4.
    1. Zelcer N, Saeki T, Bot I, Kuil A, Borst P. Transport of bile acids in multidrug-resistance-protein 3-overexpressing cells co-transfected with the ileal Na+-dependent bile-acid transporter. Biochem J 2003;369:23–30.
    1. Zhou X, Cao L, Jiang C, et al. PPARα-UGT axis activation represses intestinal FXR-FGF15 feedback signalling and exacerbates experimental colitis. Nat Commun 2014;5:4573.
    1. Strassburg CP, Nguyen N, Manns MP, Tukey RH. UDP-glucuronosyltransferase activity in human liver and colon. Gastroenterology 1999;116:149–60.
    1. Hilgendorf C, Ahlin G, Seithel A, Artursson P, Ungell AL, Karlsson J. Expression of thirty-six drug transporter genes in human intestine, liver, kidney, and organotypic cell lines. Drug Metab Dispos 2007;35:1333–40.
    1. Becker-Pauly C, Barré O, Schilling O, et al. . Proteomic analyses reveal an acidic prime side specificity for the astacin metalloprotease family reflected by physiological substrates. Mol Cell Proteomics 2011;10:M111.009233.
    1. Pineda Torra I, Claudel T, Duval C, Kosykh V, Fruchart JC, Staels B. Bile acids induce the expression of the human peroxisome proliferator-activated receptor alpha gene via activation of the farnesoid X receptor. Mol Endocrinol 2003;17:259–72.
    1. Dawson PA. Roles of Ileal ASBT and OSTα-OSTβ in Regulating Bile Acid Signaling. Dig Dis 2017;35:261–6.
    1. Song Z, Cai Y, Lao X, et al. . Taxonomic profiling and populational patterns of bacterial bile salt hydrolase [BSH] genes based on worldwide human gut microbiome. Microbiome 2019;7:9.
    1. Fukiya S, Arata M, Kawashima H, et al. . Conversion of cholic acid and chenodeoxycholic acid into their 7-oxo derivatives by Bacteroides intestinalis AM-1 isolated from human feces. FEMS Microbiol Lett 2009;293:263–70.
    1. Dao A, Abidian M, Lestrange A, Mattar M, Rangnekar A, Charabaty A.. Oral Vancomycin Induces and Maintains Remission of Ulcerative Colitis in the Subset of Patients With Associated Primary Sclerosing Cholangitis | Inflammatory Bowel Diseases. Oxford, UK: Oxford Academic. 2019;25:90–e91.
    1. Trivedi PJ, Tickle J, Vesterhus MN, et al. . Vascular adhesion protein-1 is elevated in primary sclerosing cholangitis, is predictive of clinical outcome and facilitates recruitment of gut-tropic lymphocytes to liver in a substrate-dependent manner. Gut 2018;67:1135–45.
    1. Burkett PR, Meyer zu Horste G, Kuchroo VK. Pouring fuel on the fire: Th17 cells, the environment, and autoimmunity. J Clin Invest 2015;125:2211–9.
    1. Britton GJ, Contijoch EJ, Mogno I, et al. . Microbiotas from humans with inflammatory bowel disease alter the balance of gut Th17 and RORγt+ regulatory T Cells and exacerbate colitis in mice. Immunity 2019;50:212–24.e4.
    1. Hang S, Paik D, Yao L, et al. . Bile acid metabolites control TH17 and Treg cell differentiation. Nature 2019;576:143–8.
    1. Soroosh P, Wu J, Xue X, et al. . Oxysterols are agonist ligands of RORγt and drive Th17 cell differentiation. Proc Natl Acad Sci U S A 2014;111:12163–8.
    1. Beggs AD, James J, Caldwell G, et al. . Discovery and validation of methylation biomarkers for ulcerative colitis associated neoplasia. Inflamm Bowel Dis 2018;24:1503–9.
    1. Terjung B, Söhne J, Lechtenberg B, et al. . p-ANCAs in autoimmune liver disorders recognise human beta-tubulin isotype 5 and cross-react with microbial protein FtsZ. Gut 2010;59:808–16.
    1. Hu W, Yang Y, Li X, et al. . Multi-omics approach reveals distinct differences in left- and right-sided colon cancer. Mol Cancer Res 2018;16:476–85.
    1. Poplawski A, Binder H. Feasibility of sample size calculation for RNA-seq studies. Brief Bioinform 2018;19:713–20.
    1. Acharjee A, Ament Z, West JA, Stanley E, Griffin JL. Integration of metabolomics, lipidomics and clinical data using a machine learning method. BMC Bioinformatics 2016;17:440.
    1. Mueller M, Thorell A, Claudel T, et al. . Ursodeoxycholic acid exerts farnesoid X receptor-antagonistic effects on bile acid and lipid metabolism in morbid obesity. J Hepatol 2015;62:1398–404.

Source: PubMed

3
S'abonner