Systemic sclerosis is associated with specific alterations in gastrointestinal microbiota in two independent cohorts

Elizabeth R Volkmann, Anna-Maria Hoffmann-Vold, Yu-Ling Chang, Jonathan P Jacobs, Kirsten Tillisch, Emeran A Mayer, Philip J Clements, Johannes R Hov, Martin Kummen, Øyvind Midtvedt, Venu Lagishetty, Lin Chang, Jennifer S Labus, Øyvind Molberg, Jonathan Braun, Elizabeth R Volkmann, Anna-Maria Hoffmann-Vold, Yu-Ling Chang, Jonathan P Jacobs, Kirsten Tillisch, Emeran A Mayer, Philip J Clements, Johannes R Hov, Martin Kummen, Øyvind Midtvedt, Venu Lagishetty, Lin Chang, Jennifer S Labus, Øyvind Molberg, Jonathan Braun

Abstract

Objective: To compare faecal microbial composition in patients with systemic sclerosis (SSc) from 2 independent cohorts with controls and to determine whether certain genera are associated with SSc-gastrointestinal tract (GIT) symptoms.

Design: Adult patients with SSc from the University of California, Los Angeles (UCLA) and Oslo University Hospital (OUH) and healthy controls participated in this study (1:1:1). All participants provided stool specimens for 16S rRNA sequencing. Linear discriminant analysis effect size demonstrated genera with differential expression in SSc. Differential expression analysis for sequence count data identified specific genera associated with GIT symptoms as assessed by the GIT 2.0 questionnaire.

Results: The UCLA-SSc and OUH-SSc cohorts were similar in age (52.1 and 60.5 years, respectively), disease duration (median (IQR): 6.6 (2.5-16.4) and 7.0 (1.0-19.2) years, respectively), gender distribution (88% and 71%, respectively), and GIT symptoms (mean (SD) total GIT 2.0 scores of 0.7 (0.6) and 0.6 (0.5), respectively). Principal coordinate analysis illustrated significant microbial community differences between SSc and controls (UCLA: p=0.001; OUH: p=0.002). Patients with SSc had significantly lower levels of commensal genera deemed to protect against inflammation, such as Bacteroides (UCLA and OUH), Faecalibacterium (UCLA), Clostridium (OUH); and significantly higher levels of pathobiont genera, such as Fusobacterium (UCLA), compared with controls. Increased abundance of Clostridium was associated with less severe GIT symptoms in both cohorts.

Conclusions: The present analysis detected specific aberrations in the lower GIT microbiota of patients with SSc from 2 geographically and ethnically distinct cohorts. These findings suggest that GIT dysbiosis may be a pathological feature of the SSc disease state.

Keywords: AUTOIMMUNE DISEASE; INTESTINAL MICROBIOLOGY; SYSTEMIC SCLEROSIS.

Conflict of interest statement

Competing interests: None declared.

Figures

Figure 1
Figure 1
Significant differences in the β diversity of the SSc and healthy samples as demonstrated by principal coordinate analysis plots of the weighted UniFrac distance. Each dot represents a sample from a UCLA-SSc cohort patient (open circle) or a healthy control (closed circle). Each star represents a sample from a OUH-SSc cohort patient. The p values provided were calculated by analysis of variance using distance matrices. OUH, Oslo University Hospital; PCoA, principle coordinate analysis; SSc, systemic sclerosis; UCLA, University of California, Los Angeles.
Figure 2
Figure 2
Microbial composition at the phylum level in UCLA-SSc samples (top left), OUH-SSc samples (top right) and healthy samples (bottom left). Legend provides colour coding specific to each phylum. OUH, Oslo University Hospital; SSc, systemic sclerosis; UCLA, University of California, Los Angeles.
Figure 3
Figure 3
Genus-level taxa associated with UCLA-SSc cohort patients versus healthy particiapnts. LefSe multivariate analysis was used to identify significant associations (q2.5 were included in this figure. LDA, linear discriminant analysis; LefSe, linear discriminant analysis effect size; SSc, systemic sclerosis; UCLA, University of California, Los Angeles.
Figure 4
Figure 4
Genus-level taxa associated with OUH-SSc cohort patients versus healthy participants. LefSe multivariate analysis was used to identify significant associations (q2.5 were included in this figure. LDA, linear discriminant analysis; LefSe, linear discriminant analysis effect size; OUH, Oslo University Hospital; SSc, systemic sclerosis.
Figure 5
Figure 5
Bacterial taxa associated with GIT disease score and domains. Patients were dichotomised into low (none-to-mild) or high (moderate to severe) disease severity groups for the total GIT 2.0 score and its individual domains (constipation, diarrhoea or distension/bloating). DESeq2 multivariate analysis was used to identify microbial taxa significantly associated with low versus high groups (online supplementary table S1), and calculate the fold change between groups. Negative fold change scores (log2) denote organisms decreased in high disease severity groups; whereas, positive fold change scores denote organisms increased in high disease severity groups. Legend provides colour code of bacterial taxa at the phylum level; ‘f” denotes family-level taxa. The size of the coloured dots represents the square root of the absolute mean counts of the OTUs at the genus level. DESeq2, differential expression analysis for sequence count data; GIT, gastrointestinal tract; OUT, operational taxonomic unit.

References

    1. Lock G, Holstege A, Lang B, et al. . Gastrointestinal manifestations of progressive systemic sclerosis. Am J Gastroenterol 1997;92:763–71.
    1. Sallam H, McNearney TA, Chen JD. Systematic review: pathophysiology and management of gastrointestinal dysmotility in systemic sclerosis (scleroderma). Aliment Pharmacol Ther 2006;23:691–712.
    1. Marie I, Ducrotté P, Denis P, et al. . Small intestinal bacterial overgrowth in systemic sclerosis. Rheumatology 2009;48:1314–19.
    1. Franck-Larsson K, Graf W, Rönnblom A. Lower gastrointestinal symptoms and quality of life in patients with systemic sclerosis: a population-based study. Eur J Gastroenterol Hepatol 2009;21:176–82.
    1. Bodukam V, Hays RD, Maranian P, et al. . Association of gastrointestinal involvement and depressive symptoms in patients with systemic sclerosis. Rheumatology (Oxford) 2011;50:330–4.
    1. Frank DN, Amand ALS, Feldman RA, et al. . Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci USA 2007;104:13780–5.
    1. Fava F, Danese S. Intestinal microbiota in inflammatory bowel disease: friend of foe? World J Gastroenterol 2011;17:557–66.
    1. Presley LL, Ye J, Li X, et al. . Host–microbe relationships in inflammatory bowel disease detected by bacterial and metaproteomic analysis of the mucosal–luminal interface. Inflamm Bowel Dis 2012;18:409–17.
    1. McHardy IH, Goudarzi M, Tong M, et al. . Integrative analysis of the microbiome and metabolome of the human intestinal mucosal surface reveals exquisite inter-relationships. Microbiome 2013;1:17
    1. Volkmann ER, Chang YL, Barroso N, et al. . Association of systemic sclerosis with a unique colonic microbial consortium. Microbiome 2016;68:1483–92.
    1. van den Hoogen F, Khanna D, Fransen J, et al. . 2013 classification criteria for systemic sclerosis: an American College of Rheumatology/European League against Rheumatism collaborative initiative. Arthritis Rheum 2013;65:2737–47.
    1. Tsuda A, Suda W, Morita H, et al. . Influence of proton-pump inhibitors on the luminal microbiota in the gastrointestinal tract. Clin Transl Gastroenterol 2015;6:e89
    1. Tong M, Jacobs JP, McHardy IH, et al. . Sampling of intestinal microbiota and targeted amplification of bacterial 16S rRNA genes for microbial ecologic analysis. Curr Protoc Immunol 2014;107:7, 41.1–11.
    1. Khanna D, Hays RD, Maranian P, et al. . Reliability and validity of the University of California, Los Angeles scleroderma clinical trial consortium gastrointestinal tract instrument. Arthritis Care Res 2009;61:1257–63.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B 1995;57:289–300.
    1. Goh NS, Desai SR, Veeraraghavan S, et al. . Interstitial lung disease in systemic sclerosis: a simple staging system. Am J Respir Crit Care Med 2008;177:1248–54.
    1. Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 2014;146:1489–99.
    1. Ley RE, Turnbaugh PJ, Klein S, et al. . Microbial ecology: human gut microbes associated with obesity. Nature 2006;444:1022–3.
    1. Mariat D, Firmesse O, Levenez F, et al. . The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol 2009;9:123
    1. Schaffler H, Herlemann DP, Alberts C, et al. . Mucosa-attached bacterial community in Crohn's disease coheres with the clinical disease activity index. Environ Microbiol Rep 2016. doi: 10.1111/1758-2229.12411 [Epub: 26 May 2016].
    1. Sokol H, Pigneur B, Watterlot L, et al. . Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci 2008;105:16731–6.
    1. Varela E, Manichanh C, Gallart M, et al. . Colonisation by Faecalibacterium prausnitzii and maintenance of clinical remission in patients with ulcerative colitis. Aliment Pharmacol Ther 2013;38:151–61.
    1. Prosberg M, Bendtsen F, Vind I, et al. . The association between the gut microbiota and the inflammatory bowel disease activity: a systematic review and meta-analysis. Scand J Gastroenterol 2016;51:1407–15.
    1. Atarashi K, Tanoue T, Oshima K, et al. . Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013;500:232–6.
    1. Slobodin G, Rimar D. Regulatory T cells in systemic sclerosis: a comprehensive review. Clin Rev Allergy Immunol 2017;52:194–201.
    1. Maeda Y, Kurakawa T, Umemoto E, et al. . Dysbiosis contributes to arthritis development via activation of autoreactive T cells in the intestine. Arthritis Rheumatol 2016;68:2646–61.
    1. Benjamin JL, Hedin CR, Koutsoumpas A, et al. . Smokers with active Crohn's disease have a clinically relevant dysbiosis of the gastrointestinal microbiota. Inflamm Bowel Dis 2012;18:1092–100.
    1. De Cruz P, Kang S, Wagner J, et al. . Association between specific mucosa-associated microbiota in Crohn's disease at the time of resection and subsequent disease recurrrence: a pilot study. J Gastroenterol Hepatol 2015;30:268–78.
    1. Scher JU, Sczesnak A, Longman RS, et al. . Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife 2013;2:e01202
    1. Gevers D, Kugathasan S, Denson LA, et al. . The treatment-naive microbiome in new-onset Crohn's disease. Cell Host Microbe 2014;15:382–92.
    1. Strauss J, Kaplan GG, Beck PL, et al. . Invasive potential of gut mucosa-derived Fusobacterium nucleatum positively correlates with IBD status of the host. Inflamm Bowel Dis 2011;17:1971–8.
    1. Andréasson K, Alrawi Z, Persson A, et al. . Intestinal dysbiosis is common in systemic sclerosis and associated with gastrointestinal and extraintestinal features of disease. Arthritis Res Ther 2016;18:278
    1. McNulty NP, Yatsunenko T, Hsiao A, et al. . The impact of a consortium of fermented milk strains on the gut microbiome in gnotobiotic mice and monozygotic twins. Sci Transl Med 2011;3:106ra106
    1. Walter J. Ecological role of lactobacilli in the gastrointestinal tract: implications for fundamental and biomedical research. Appl Environ Microbiol 2008;74:4985–96.
    1. Tillisch K, Labus J, Kilpatrick L, et al. . Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 2013;144:1394–401.
    1. Samuelson DR, Welsh DA, Shellito JE. Regulation of lung immunity and host defense by the intestinal microbiota. Front Microbiol 2015;6:1085
    1. Mar JS, LaMere BJ, Lin DL, et al. . Disease severity and immune activity relate to distinct interkingdom gut microbiome states in ethnically distinct ulcerative colitis patients. MBio 2016;7:e01072–16.
    1. Marie I, Leori AM, Menard JF, et al. . Fecal calprotectin in systemic sclerosis and review of the literature. Autoimmun Rev 2015;14:547–54.

Source: PubMed

3
Abonner