Altered Mucosal Microbiome Diversity and Disease Severity in Sjögren Syndrome

Cintia S de Paiva, Dan B Jones, Michael E Stern, Fang Bian, Quianta L Moore, Shani Corbiere, Charles F Streckfus, Diane S Hutchinson, Nadim J Ajami, Joseph F Petrosino, Stephen C Pflugfelder, Cintia S de Paiva, Dan B Jones, Michael E Stern, Fang Bian, Quianta L Moore, Shani Corbiere, Charles F Streckfus, Diane S Hutchinson, Nadim J Ajami, Joseph F Petrosino, Stephen C Pflugfelder

Abstract

There is mounting evidence that the microbiome has potent immunoregulatory functions. We assessed the effects of intestinal dysbiosis in a model of Sjögren syndrome (SS) by subjecting mice to desiccating stress (DS) and antibiotics (ABX). We characterized the conjunctival, tongue and fecal microbiome profiles of patients with SS. Severity of ocular surface and systemic disease was graded. 16S ribosomal RNA gene sequencing characterized the microbiota. ABX + DS mice had a significantly worse dry eye phenotype compared to controls, a decrease in Clostridium and an increase in Enterobacter, Escherichia/Shigella, and Pseudomonas in stool after ABX + DS for 10 days. Goblet cell density was significantly lower in ABX treated groups compared to controls. Stool from SS subjects had greater relative abundances of Pseudobutyrivibrio, Escherichia/Shigella, Blautia, and Streptococcus, while relative abundance of Bacteroides, Parabacteroides, Faecalibacterium, and Prevotella was reduced compared to controls. The severity of SS ocular and systemic disease was inversely correlated with microbial diversity. These findings suggest that SS is marked by a dysbiotic intestinal microbiome driven by low relative abundance of commensal bacteria and high relative abundance of potentially pathogenic genera that is associated with worse ocular mucosal disease in a mouse model of SS and in SS patients.

Figures

Figure 1. Decreased diversity after antibiotic regimen…
Figure 1. Decreased diversity after antibiotic regimen in mice subjected to desiccating stress.
(A) Schematic of experimental design. Mice were left non-stressed (NS) or subjected to desiccating stress (DS) for 5 or 10 days (DS5 and DS10) while drinking regular water. A separate group of mice received oral antibiotics (ABX: Ampicillin, Gentamicin, Metronidazole, Neomycin, Vancomycin) in water 14 days prior (baseline) and later were randomized to remain non-stressed (ABX + NS) or to be subjected to desiccating stress for 5 or days while still on ABX water (ABX + DS5 and ABX + DS10, respectively). Thick black arrow indicates duration of water treatment. (B) Number of observed operational taxonomic units (OTUs) and Shannon Diversity Index scores in non-stressed mice (NS, baseline) prior to exposure to desiccating stress (DS) with antibiotic cocktail (ABX) for 5 or 10 days (DS5 and DS10, respectively). ****P < 0.0001 compared to baseline group (Kruskall-Wallis test with FDR correction). (C) Principal coordinate analysis (PCoA) plot of unweighted UniFrac distances. Each symbol represents an individual sample from baseline and ABX mice subjected to DS for 5 and 10 days. PERMANOVA test, R2 = coefficient of determination. (D) Comparison of significant relative abundance of different genera among groups. Dotted line divides significant genera that decrease (left) or increase (right) in ABX + DS5 and ABX + DS10 compared to baseline. (Mean ± SEM) *P < 0.05; ****P < 0.0001 (Kruskall-Wallis test with FDR correction).
Figure 2. Gut dysbiosis worsens response to…
Figure 2. Gut dysbiosis worsens response to desiccating stress and increases production of T-cell related cytokines in conjunctival epithelium.
(A–C) Goblet cell density (A), CD4+T cell infiltration (B) and corneal barrier function measured by uptake of Oregon-Green Dextran (OGD, C) in mice prior to (baseline, NS) and after exposure to desiccating stress with and without antibiotic cocktail (ABX) for 5 or 10 days (DS5 and DS10, respectively). *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001 water vs. ABX comparisons; Kruskall-Wallis followed by Sidak’s multi-comparisons test. (D). Relative fold of expression of IL-17, IFN-γ, IL-13, Foxa2, IL-13/IFN-γ ratio and Integrin alpha 2 (Itga2, CD49b) in conjunctiva from mice prior to (non-stressed, NS) and after exposure to desiccating stress with and without antibiotic cocktail (ABX) for 5 or 10 days (DS5 and DS10, respectively). Data are presented as mean ± SEM of a representative experiment containing 3-4 individual samples/group. Experiment was repeated once with similar results. *p < 0.05; **p < 0.01, ***p < 0.001; ****p < 0.0001 water vs. ABX at each time point, calculated by Kruskall-Wallis followed by Sidak’s multi-comparisons test.
Figure 3. Human stool microbiome in SS.
Figure 3. Human stool microbiome in SS.
(A) Comparison of all significant shifts in the abundance of genera between control subjects and SS patients (Mean ± SEM). Dotted line divides significant genera that decrease (top) or increase (bottom) in SS compared to controls. (B) Inverse Pearson’s correlation of combined severity index and number of intestinal OTUs, R = coefficient of correlation (C). Ocular severity graded (0–4) Dry Eye Workshop (DEWS) criteria, systemic severity graded (0–33) using unweighted 12-domain ESSDAI (European League against rheumatism (EULAR) Sjögren syndrome disease activity index) and combined ocular and systemic severity (sum of ocular and systemic severity scores). NS = non-significant. *p < 0.05; **p < 0.01, ***p < 0.001; ****p < 0.0001 Mann-Whitney test. NP: not performed.

References

    1. Haugen A. J. et al.. Estimation of the prevalence of primary Sjogren’s syndrome in two age-different community-based populations using two sets of classification criteria: the Hordaland Health Study. Scand. J. Rheumatol. 37, 30–34 (2008).
    1. Christodoulou M. I., Kapsogeorgou E. K. & Moutsopoulos H. M. Characteristics of the minor salivary gland infiltrates in Sjogren’s syndrome. J. Autoimmun. 34, 400–407 (2010).
    1. Nguyen C. Q. & Peck A. B. Unraveling the pathophysiology of Sjogren syndrome-associated dry eye disease. Ocul. Surf. 7, 11–27 (2009).
    1. Pflugfelder S. C. et al.. Aqueous Tear Deficiency Increases Conjunctival Interferon-gamma (IFN-gamma) Expression and Goblet Cell Loss. Invest. Ophthalmol. Vis. Sci. 56, 7545–7550 (2015).
    1. Pflugfelder S. C. et al.. Conjunctival cytologic features of primary Sjogren’s syndrome. Ophthalmology 97, 985–991 (1990).
    1. Pflugfelder S. C., Jones D., Ji Z., Afonso A. & Monroy D. Altered cytokine balance in the tear fluid and conjunctiva of patients with Sjogren’s syndrome keratoconjunctivitis sicca. Curr.Eye Res. 19, 201–211 (1999).
    1. MacFarlane T. W. & Mason D. K. Changes in the oral flora in Sjogren’s syndrome. J. Clin. Pathol. 27, 416–419 (1974).
    1. Carsons S. A review and update of Sjogren’s syndrome: manifestations, diagnosis, and treatment. Am. J. Manag. Care 7, S433–S443 (2001).
    1. Buchholz P. et al.. Utility assessment to measure the impact of dry eye disease. Ocul.Surf. 4, 155–161 (2006).
    1. Bielory L. & Syed B. A. Pharmacoeconomics of anterior ocular inflammatory disease. Curr. Opin. Allergy Clin. Immunol. 13, 537–542 (2013).
    1. Stern M. E., Schaumburg C. S. & Pflugfelder S. C. Dry eye as a mucosal autoimmune disease. Int.Rev.Immunol. 32, 19–41 (2013).
    1. de Paiva C. S. et al.. Dry Eye-Induced Conjunctival Epithelial Squamous Metaplasia Is Modulated by Interferon-{gamma}. Invest Ophthalmol. Vis. Sci. 48, 2553–2560 (2007).
    1. Zhang X. et al.. Topical interferon-gamma neutralization prevents conjunctival goblet cell loss in experimental murine dry eye. Exp. Eye Res. 118, 117–124 (2014).
    1. Zhang X. et al.. Interferon-gamma exacerbates dry eye-induced apoptosis in conjunctiva through dual apoptotic pathways. Invest Ophthalmol. Vis. Sci. 52, 6279–6285 (2011).
    1. de Paiva C. S. et al.. IL-17 disrupts corneal barrier following desiccating stress. Mucosal. Immunol. 2, 243–253 (2009).
    1. Chauhan S. K. et al.. A novel pro-lymphangiogenic function for Th17/IL-17. Blood 118, 4630–4634 (2011).
    1. Tzioufas A. G., Kapsogeorgou E. K. & Moutsopoulos H. M. Pathogenesis of Sjogren’s syndrome: what we know and what we should learn. J. Autoimmun. 39, 4–8 (2012).
    1. Tzioufas A. G. et al.. Clinical, immunological, and immunogenetic aspects of autoantibody production against Ro/SSA, La/SSB and their linear epitopes in primary Sjogren’s syndrome (pSS): a European multicentre study. Ann. Rheum. Dis. 61, 398–404 (2002).
    1. Zhang X. Y. et al.. [The study on correlativity between HLA-DQ gene polymorphism and primary Sjogren’s syndrome of the Han nationality in Shanxi province]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 27, 182–185 (2011).
    1. Gong Y. Z. et al.. Differentiation of follicular helper T cells by salivary gland epithelial cells in primary Sjogren’s syndrome. J. Autoimmun. 51, 57–66 (2014).
    1. Anaya J. M., Delgado-Vega A. M. & Castiblanco J. Genetic basis of Sjogren’s syndrome. How strong is the evidence? Clin. Dev. Immunol. 13, 209–222 (2006).
    1. Furusawa Y. et al.. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504, 446–450 (2013).
    1. Arpaia N. et al.. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504, 451–455 (2013).
    1. Nagalingam N. A., Kao J. Y. & Young V. B. Microbial ecology of the murine gut associated with the development of dextran sodium sulfate-induced colitis. Inflamm. Bowel Dis. 17, 917–926 (2011).
    1. Maslowski K. M. et al.. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461, 1282–1286 (2009).
    1. Shaw S. Y., Blanchard J. F. & Bernstein C. N. Association between the use of antibiotics in the first year of life and pediatric inflammatory bowel disease. Am. J. Gastroenterol. 105, 2687–2692 (2010).
    1. Sokol H. et al.. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA. 105, 16731–16736 (2008).
    1. Chotikavanich S. et al.. Production and Activity of Matrix Metalloproteinase-9 on the Ocular Surface Increase in Dysfunctional Tear Syndrome. Invest. Ophthalmol. Vis. Sci. 50, 3203–3209 (2009).
    1. Corrales R. M. et al.. Entrapment of conjunctival goblet cells by desiccation-induced cornification. Invest. Ophthalmol. Vis. Sci. 52, 3492–3499 (2011).
    1. de Paiva C. S. et al.. Corticosteroid and doxycycline suppress MMP-9 and inflammatory cytokine expression, MAPK activation in the corneal epithelium in experimental dry eye. Exp. Eye Res. 83, 526–535 (2006).
    1. de Paiva C. S. et al.. Apical corneal barrier disruption in experimental murine dry eye is abrogated by methylprednisolone and doxycycline. Invest. Ophthalmol. Vis. Sci. 47, 2847–2856 (2006).
    1. de Paiva C. S., Schwartz C. E., Gjorstrup P. & Pflugfelder S. C. Resolvin E1 (RX-10001) reduces corneal epithelial barrier disruption and protects against goblet cell loss in a murine model of dry eye. Cornea 31, 1299–1303 (2012).
    1. Beardsley R. M., de Paiva C. S., Power D. F. & Pflugfelder S. C. Desiccating stress decreases apical corneal epithelial cell size–modulation by the metalloproteinase inhibitor doxycycline. Cornea 27, 935–940 (2008).
    1. Dohlman T. H. et al.. The CCR6/CCL20 Axis Mediates Th17 Cell Migration to the Ocular Surface in Dry Eye Disease. Invest Ophthalmol. Vis. Sci. 54, 4081–4091 (2013).
    1. Ecoiffier T., El A. J., Rashid S., Schaumberg D. & Dana R. Modulation of integrin alpha4beta1 (VLA-4) in dry eye disease. Arch.Ophthalmol. 126, 1695–1699 (2008).
    1. Goyal S., Chauhan S. K., Zhang Q. & Dana R. Amelioration of murine dry eye disease by topical antagonist to chemokine receptor 2. Arch.Ophthalmol. 127, 882–887 (2009).
    1. Lee H. S., Chauhan S. K., Okanobo A., Nallasamy N. & Dana R. Therapeutic Efficacy of Topical Epigallocatechin Gallate in Murine Dry Eye. Cornea 30 1465–1472 (2011).
    1. van ‘t Hof W., Veerman E. C., Nieuw Amerongen A. V. & Ligtenberg A. J. Antimicrobial defense systems in saliva. Monogr. Oral Sci. 24, 40–51 (2014).
    1. Haynes R. J., Tighe P. J. & Dua H. S. Antimicrobial defensin peptides of the human ocular surface. Br.J Ophthalmol. 83, 737–741 (1999).
    1. Hill D. A. et al.. Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic inflammation. Nat. Med. 18, 538–546 (2012).
    1. de Paiva C. S. et al.. Homeostatic control of conjunctival mucosal goblet cells by NKT-derived IL-13. Mucosal. Immunol. 4, 397–408 (2011).
    1. Zhang X. et al.. Topical interferon-gamma neutralization prevents conjunctival goblet cell loss in experimental murine dry eye. Exp Eye Res. 118, 117–124 (2014).
    1. Tukler Henriksson J., Coursey T. G., Corry D. B., De Paiva C. S. & Pflugfelder S. C. IL-13 Stimulates Proliferation and Expression of Mucin and Immunomodulatory Genes in Cultured Conjunctival Goblet Cells. Invest. Ophthalmol. Vis. Sci. 56, 4186–4197 (2015).
    1. Krimi R. B. et al.. Resistin-like molecule beta regulates intestinal mucous secretion and curtails TNBS-induced colitis in mice. Inflamm. Bowel Dis. 14, 931–941 (2008).
    1. Nair M. G. et al.. Goblet cell-derived resistin-like molecule beta augments CD4 + T cell production of IFN-gamma and infection-induced intestinal inflammation. J. Immunol. 181, 4709–4715 (2008).
    1. Human Microbiome Project, C. Structure, function and diversity of the healthy human microbiome. Nature 486, 207–214 (2012).
    1. Aagaard K. et al.. The Human Microbiome Project strategy for comprehensive sampling of the human microbiome and why it matters. FASEB J. 27, 1012–1022 (2013).
    1. Human Microbiome Project, C. A framework for human microbiome research. Nature 486, 215–221 (2012).
    1. Miquel S. et al.. Faecalibacterium prausnitzii and human intestinal health. Curr. Opin. Microbiol. 16, 255–261 (2013).
    1. Blumberg R. & Powrie F. Microbiota, disease, and back to health: a metastable journey. Sci. Transl. Med. 4, 137rv137 (2012).
    1. Markle J. G. et al.. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science 339, 1084–1088 (2013).
    1. Wen L. et al.. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature 455, 1109–1113 (2008).
    1. Horai R. et al.. Microbiota-Dependent Activation of an Autoreactive T Cell Receptor Provokes Autoimmunity in an Immunologically Privileged Site. Immunity 43, 343–353 (2015).
    1. Berer K. et al.. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479, 538–541 (2011).
    1. Keeney K. M., Yurist-Doutsch S., Arrieta M. C. & Finlay B. B. Effects of antibiotics on human microbiota and subsequent disease. Annu. Rev. Microbiol. 68, 217–235 (2014).
    1. Atarashi K. et al.. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337–341 (2011).
    1. Coursey T. G., Tukler Henriksson J., Chen M., de Paiva C. S. & Pflugfelder S. C. IFN-γ Induced Unfolded Protein Response in Conjunctival Goblet Cells as Cause of Mucin Deficiency in Sjögren’s Syndrome Am. J. Pathol. in press (2016).
    1. Zegans M. E. & Van Gelder R. N. Considerations in understanding the ocular surface microbiome. Am. J. Ophthalmol. 158, 420–422 (2014).
    1. Shiboski S. C. et al.. American College of Rheumatology classification criteria for Sjogren’s syndrome: a data-driven, expert consensus approach in the Sjogren’s International Collaborative Clinical Alliance cohort. Arthritis Care Res. (Hoboken) 64, 475–487 (2012).
    1. Foulks G. N. et al.. Clinical Guidelines for Management of Dry Eye Associated with Sjogren Disease. Ocul Surf 13, 118–132 (2015).
    1. Seror R. et al.. EULAR Sjogren’s syndrome disease activity index: development of a consensus systemic disease activity index for primary Sjogren’s syndrome. Ann. Rheum. Dis. 69, 1103–1109 (2010).
    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. Quast C. et al.. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 41, D590–596 (2013).
    1. Edgar R. C. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods 10, 996–998 (2013).
    1. Lozupone C. & Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl. Environ. Microbiol. 71, 8228–8235 (2005).

Source: PubMed

3
Subscribe