Alterations of the human gut microbiome in multiple sclerosis

Sushrut Jangi, Roopali Gandhi, Laura M Cox, Ning Li, Felipe von Glehn, Raymond Yan, Bonny Patel, Maria Antonietta Mazzola, Shirong Liu, Bonnie L Glanz, Sandra Cook, Stephanie Tankou, Fiona Stuart, Kirsy Melo, Parham Nejad, Kathleen Smith, Begüm D Topçuolu, James Holden, Pia Kivisäkk, Tanuja Chitnis, Philip L De Jager, Francisco J Quintana, Georg K Gerber, Lynn Bry, Howard L Weiner, Sushrut Jangi, Roopali Gandhi, Laura M Cox, Ning Li, Felipe von Glehn, Raymond Yan, Bonny Patel, Maria Antonietta Mazzola, Shirong Liu, Bonnie L Glanz, Sandra Cook, Stephanie Tankou, Fiona Stuart, Kirsy Melo, Parham Nejad, Kathleen Smith, Begüm D Topçuolu, James Holden, Pia Kivisäkk, Tanuja Chitnis, Philip L De Jager, Francisco J Quintana, Georg K Gerber, Lynn Bry, Howard L Weiner

Abstract

The gut microbiome plays an important role in immune function and has been implicated in several autoimmune disorders. Here we use 16S rRNA sequencing to investigate the gut microbiome in subjects with multiple sclerosis (MS, n=60) and healthy controls (n=43). Microbiome alterations in MS include increases in Methanobrevibacter and Akkermansia and decreases in Butyricimonas, and correlate with variations in the expression of genes involved in dendritic cell maturation, interferon signalling and NF-kB signalling pathways in circulating T cells and monocytes. Patients on disease-modifying treatment show increased abundances of Prevotella and Sutterella, and decreased Sarcina, compared with untreated patients. MS patients of a second cohort show elevated breath methane compared with controls, consistent with our observation of increased gut Methanobrevibacter in MS in the first cohort. Further study is required to assess whether the observed alterations in the gut microbiome play a role in, or are a consequence of, MS pathogenesis.

Figures

Figure 1. Study design.
Figure 1. Study design.
Faecal samples were collected from MS patients (n=60) and healthy subjects (n=43). Microbial DNA was extracted from frozen faecal samples and 16s rDNA sequencing was performed using Roche 454 and Illumina platforms. Gene expression profiling was performed on circulating monocytes and T cells from MS patients (n=18) and healthy subjects (n=18) using a Nanostring platform. Peripheral blood mononuclear cells were collected from MS patients (n=18) and healthy subjects (n=18) to conduct proliferation and cytokine assays in response to specific microbial stimulation. Sera from MS patients (n=45) and healthy subjects (n=16) was collected for ELISA-based techniques to capture serologic activity directed against specific microbes. Breath samples from MS patients (n=41) and healthy subjects (n=32) were collected from a second subject cohort to determine breath methane concentrations.
Figure 2. Compositional differences in faecal microbiota…
Figure 2. Compositional differences in faecal microbiota between MS patients and healthy subjects.
(a) Relative abundances of Euryarchaeota and Verrucomicrobia in the faecal microbiota of healthy controls (n=43, grey bar), all MS patients (n=60, red) and both untreated (n=28, orange) and treated MS patient (n=32, blue) subgroups as analysed by two independent sequencing technologies, 454 (top) or MiSeq (bottom). (b) Relative abundance of prevalent microbiota (>1% in any sample group) determined from MiSeq and 454 high-throughput sequencing. (c) Relative abundances of genera in the faecal microbiota that are significantly altered between healthy controls (n=43) and MS patients (n=60; MS-effect) or between untreated (n=28) and treated MS patients (n=32) (disease effect) as analysed by two independent sequencing technologies. Significance was determined by DESeq and Benjamini–Hochberg corrected P values <0.05 with a false discovery rate threshold of 0.1. Bars represent average, and error bars depict s.e.
Figure 3. Correlations between microbiota abundances and…
Figure 3. Correlations between microbiota abundances and immune gene expression.
(a) Gene expression was measured from circulating T cells and monocytes by the Nanostring Immunology panel in MS patients (n=18) and healthy controls (n=18). (a) canonical pathways significantly altered in MS patients and healthy subjects with an activation z-score>|1.5| in both T cells (black bars) and monocytes (grey bars) identified by Ingenuity Pathway Analysis. (b) Altered gut microbiota abundances correlate with immune gene expression in MS patients. Spearman's correlations (σ) between the relative abundance of significantly altered microbes in subject groups and the relative expression of genes from identified canonical pathways significantly altered between healthy controls and untreated MS patients. Colour and slope of ellipse indicate magnitude of correlation, with σ value superimposed on ellipse. Subject groups were either all MS patients and controls together (All), untreated MS patients alone (MS-U) or healthy controls alone (HC).
Figure 4. Measurement of breath methane production…
Figure 4. Measurement of breath methane production in MS patients (n=41) and controls (n=32).
Breath methane measured in each subject is represented on the y axis in parts-per-million on a logarithmic scale. The mean and s.e.m. are shown by the indicated horizontal lines. 28 of 41 MS patients and 24 of 32 controls had no detectable breath methane.

References

    1. Lozupone C. A., Stombaugh J. I., Gordon J. I., Jansson J. K. & Knight R. Diversity, stability and resilience of the human gut microbiota. Nature 489, 220–230 (2012).
    1. McDermott A. J. & Huffnagle G. B. The microbiome and regulation of mucosal immunity. Immunology 142, 24–31 (2014).
    1. Kostic A. D., Xavier R. J. & Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 146, 1489–1499 (2014).
    1. Alkanani A. K. et al. Alterations in intestinal microbiota correlate with susceptibility to type 1 diabetes. Diabetes 64, 3510–3520 (2015).
    1. Scher J. U. et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife 2, e01202 (2013).
    1. Berer K. et al. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479, 538–541 (2011).
    1. Ivanov II et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139, 485–498 (2009).
    1. Lee Y. K., Menezes J. S., Umesaki Y. & Mazmanian S. K. Proinflammatory T-Cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc. Natl. Acad. Sci. USA 108, (Suppl 1): 4615–4622 (2011).
    1. Round J. L. & Mazmanian S. K. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl. Acad. Sci. USA 107, 12204–12209 (2010).
    1. Ochoa-Reparaz J. et al. A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal Immunol. 3, 487–495 (2010).
    1. Miyake S. et al. Dysbiosis in the gut microbiota of patients with multiple sclerosis, with a striking depletion of species belonging to clostridia XIVa and IV clusters. PLoS ONE 10, e0137429 (2015).
    1. Machiels K. et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 63, 1275–1283 (2014).
    1. Varrin-Doyer M. et al. Aquaporin 4-specific T cells in neuromyelitis optica exhibit a Th17 bias and recognize Clostridium ABC transporter. Ann. Neurol. 72, 53–64 (2012).
    1. Jacobs B. C. et al. Campylobacter jejuni infections and anti-GM1 antibodies in Guillain-Barre syndrome. Ann. Neurol. 40, 181–187 (1996).
    1. Schloss P. D. et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 75, 7537–7541 (2009).
    1. Matsen F. A., Kodner R. B. & Armbrust E. V. pplacer: linear time maximum-likelihood and Bayesian phylogenetic placement of sequences onto a fixed reference tree. BMC Bioinformatics 11, 538 (2010).
    1. Bang C., Weidenbach K., Gutsmann T., Heine H. & Schmitz R. A. The intestinal archaea Methanosphaera stadtmanae and Methanobrevibacter smithii activate human dendritic cells. PLoS ONE 9, e99411 (2014).
    1. Cao Y. et al. Functional inflammatory profiles distinguish myelin-reactive T cells from patients with multiple sclerosis. Sci. Transl. Med. 7, 287ra274 (2015).
    1. Schneider A. et al. In active relapsing-remitting multiple sclerosis, effector T Cell Res.istance to adaptive T(regs) involves IL-6-mediated signaling. Sci. Transl. Med. 5, 170ra115 (2013).
    1. Miterski B. et al. Inhibitors in the NFkappaB cascade comprise prime candidate genes predisposing to multiple sclerosis, especially in selected combinations. Genes Immun. 3, 211–219 (2002).
    1. Gilli F. et al. Learning from nature: pregnancy changes the expression of inflammation-related genes in patients with multiple sclerosis. PLoS ONE 5, e8962 (2010).
    1. Miranda-Hernandez S. & Baxter A. G. Role of toll-like receptors in multiple sclerosis. Am. J. Clin. Exp. Immunol. 2, 75–93 (2013).
    1. Fernandez-Paredes L., de Diego R. P., de Andres C. & Sanchez-Ramon S. Close encounters of the first kind: innate sensors and multiple sclerosis. Mol. Neurobiol. doi:10.1007/s12035-015-9665-5 (2016).
    1. Blais Lecours P. et al. Immunogenic properties of archaeal species found in bioaerosols. PLoS ONE 6, e23326 (2011).
    1. McKay L. F., Eastwood M. A. & Brydon W. G. Methane excretion in man--a study of breath, flatus, and faeces. Gut 26, 69–74 (1985).
    1. Pimentel M. et al. Methane production during lactulose breath test is associated with gastrointestinal disease presentation. Dig. Dis. Sci. 48, 86–92 (2003).
    1. Haberman Y. et al. Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature. J. Clin. Invest. 124, 3617–3633 (2014).
    1. Dunne J. L. et al. The intestinal microbiome in type 1 diabetes. Clin. Exp. Immunol. 177, 30–37 (2014).
    1. Oksenberg J. R., Baranzini S. E., Sawcer S. & Hauser S. L. The genetics of multiple sclerosis: SNPs to pathways to pathogenesis. Nat. Rev. Genet. 9, 516–526 (2008).
    1. Verma R., Verma A. K., Ahuja V. & Paul J. Real-time analysis of mucosal flora in patients with inflammatory bowel disease in India. J. Clin. Microbiol. 48, 4279–4282 (2010).
    1. Yamabe K. et al. Distribution of Archaea in Japanese patients with periodontitis and humoral immune response to the components. FEMS Microbiol. Lett. 287, 69–75 (2008).
    1. Krishnan L., Sad S., Patel G. B. & Sprott G. D. The potent adjuvant activity of archaeosomes correlates to the recruitment and activation of macrophages and dendritic cells in vivo. J. Immunol. 166, 1885–1893 (2001).
    1. Samuel B. S. et al. Genomic and metabolic adaptations of Methanobrevibacter smithii to the human gut. Proc. Natl. Acad. Sci. USA 104, 10643–10648 (2007).
    1. Mbakwa C. A. et al. Gut colonization with Methanobrevibacter smithii is associated with childhood weight development. Obesity (Silver Spring) 23, 2508–2516 (2015).
    1. Tremlett H. et al. Gut microbiota composition and relapse risk in pediatric MS: A pilot study. J. Neurol. Sci. 363, 153–157 (2016).
    1. Cantarel B. L. et al. Gut microbiota in multiple sclerosis: possible influence of immunomodulators. J. Invest. Med. 63, 729–734 (2015).
    1. Scher J. U. et al. Decreased bacterial diversity characterizes the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease. Arthritis Rheumatol. 67, 128–139 (2015).
    1. Derrien M., Vaughan E. E., Plugge C. M. & de Vos W. M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 54, 1469–1476 (2004).
    1. Derrien M. et al. Modulation of mucosal immune response, tolerance, and proliferation in mice colonized by the mucin-degrader Akkermansia muciniphila. Front. Microbiol. 2, 166 (2011).
    1. Ganesh B. P., Klopfleisch R., Loh G. & Blaut M. Commensal Akkermansia muciniphila exacerbates gut inflammation in Salmonella typhimurium-infected gnotobiotic mice. PLoS ONE 8, e74963 (2013).
    1. Furusawa Y. et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504, 446–450 (2013).
    1. Wang W. et al. Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease. J. Clin. Microbiol. 52, 398–406 (2014).
    1. de Goffau M. C. et al. Aberrant gut microbiota composition at the onset of type 1 diabetes in young children. Diabetologia 57, 1569–1577 (2014).
    1. Joossens M. et al. Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives. Gut 60, 631–637 (2011).
    1. Wu G. D. et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 334, 105–108 (2011).
    1. Gevers D. et al. The treatment-naive microbiome in new-onset Crohn's disease. Cell Host Microbe 15, 382–392 (2014).
    1. De Angelis M. et al. Fecal microbiota and metabolome of children with autism and pervasive developmental disorder not otherwise specified. PLoS ONE 8, e76993 (2013).
    1. Levy M., Thaiss C. A. & Elinav E. Metagenomic cross-talk: the regulatory interplay between immunogenomics and the microbiome. Genome Med. 7, 120 (2015).
    1. Morgan X. C. et al. Associations between host gene expression, the mucosal microbiome, and clinical outcome in the pelvic pouch of patients with inflammatory bowel disease. Genome Biol. 16, 67 (2015).
    1. Achiron A., Gurevich M., Friedman N., Kaminski N. & Mandel M. Blood transcriptional signatures of multiple sclerosis: unique gene expression of disease activity. Ann. Neurol. 55, 410–417 (2004).
    1. Sheng W. et al. STAT5 programs a distinct subset of GM-CSF-producing T helper cells that is essential for autoimmune neuroinflammation. Cell Res. 24, 1387–1402 (2014).
    1. Kim G. et al. Methanobrevibacter smithii is the predominant methanogen in patients with constipation-predominant IBS and methane on breath. Dig. Dis. Sci. 57, 3213–3218 (2012).
    1. Riccio P., Rossano R. & Liuzzi G. M. May diet and dietary supplements improve the wellness of multiple sclerosis patients? A molecular approach. Autoimmune Dis. 2010, 249842 (2011).
    1. Giongo A. et al. Toward defining the autoimmune microbiome for type 1 diabetes. ISME J. 5, 82–91 (2011).
    1. Human Microbiome Project, C. A framework for human microbiome research. Nature 486, 215–221 (2012).
    1. Human Microbiome Project, C. Structure, function and diversity of the healthy human microbiome. Nature 486, 207–214 (2012).
    1. Caporaso J. G. et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J. 6, 1621–1624 (2012).
    1. Shannon C. E. W. W. A Mathematical Theory of Communication. Bell Syst. Tech. J. 27, 379–423 (1948).
    1. D. F. Bauer Constructing confidence sets using rank statistics. J. Am. Stat. Assoc. 67, 687–690 (1972).
    1. Lozupone C. & Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl. Environ. Microbiol. 71, 8228–8235 (2005).
    1. Excoffier L., Smouse P. E. & Quattro J. M. Analysis of molecular variance inferred from metric distances among DNA haplotypes: application to human mitochondrial DNA restriction data. Genetics 131, 479–491 (1992).
    1. Love M. I., Huber W. & Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
    1. McMurdie P.J. & Holmes S. Waste not, want not: why rarefying microbiome data is inadmissible. PLoS Comput. Biol. 10, (2014).
    1. Benjamini Y. & Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J.R. Stat. Soc. B 57, 289–300 (1995).
    1. Cole J. R. et al. Ribosomal Database Project: data and tools for high throughput rRNA analysis. Nucleic Acids Res. 42, D633–D642 (2014).
    1. Edgar R. C. MUSCLE: a multiple sequence alignment method with reduced time and space complexity. BMC Bioinformatics 5, 113 (2004).
    1. Price M. N., Dehal P. S. & Arkin A. P. FastTree: computing large minimum evolution trees with profiles instead of a distance matrix. Mol. Biol. Evol. 26, 1641–1650 (2009).
    1. Miller T. L., Wolin M. J., Conway de Macario E. & Macario A. J. Isolation of Methanobrevibacter smithii from human feces. Appl. Environ. Microbiol. 43, 227–232 (1982).
    1. Mazzola M. A. et al. Identification of a novel mechanism of action of fingolimod (FTY720) on human effector T cell function through TCF-1 upregulation. J. Neuroinflamm. 12, 245 (2015).
    1. R Development Core Team. R: a Language and Environment for Statistical Computing. (2015).
    1. Murdoch D. & Chow E. D. ellipse: Functions for Drawing Ellipses and Ellipse-Like Confidence Regions. Cran Repository. Version 0.3-8 (2013).

Source: PubMed

3
구독하다