Skip to main content
Top

18-10-2018 | Microbiome | Editorial | Article

Why every rheumatologist should understand the microbiome

Authors: James T Rosenbaum, Mark J Asquith

Learning objectives

Introduction

Akkermansia muciniphila. Prevotella copri. Enterococcus gallinarum. Porphyromonas gingivalis. These bacteria were rarely the subject of lectures in medical schools prior to 2018, but might well revolutionize the practice of rheumatology in the next decade or two. In this editorial, we will explain why rheumatologists might need to learn a bit more about the microbiome, the collection of micro-organisms that cohabit in, and on, the human body.

How the gut microbiome interacts with the immune system

By some parameters the gut, home to trillions of bacteria, is the largest organ in the immune system. The immune system interacts with these bacteria and decides which bacteria are symbionts, and which could potentially cause disease. Lymphocytes that interact with bacteria in the intestine distribute themselves widely throughout the body [1–3]. Many gut bacteria influence the production of cytokines and various T-cell subsets (reviewed in [4]), and bacterial products can be detected in the blood [5], the joints [6], and elsewhere throughout the body.

Since the immune system is educated by intestinal bacteria, and since rheumatologic diseases are usually immune-mediated, it stands to reason that gut bacteria may have an influence on rheumatic diseases. Indeed, gut bacteria are altered in patients with rheumatoid arthritis [7], systemic lupus erythematosus [8], ankylosing spondylitis [9], reactive arthritis [10], psoriatic arthritis [11], juvenile onset arthritis [12], gout [13], and scleroderma [14]. Bacteria in the intestine have a profound effect on diseases such as collagen-induced arthritis [15], murine lupus [16], and spondyloarthritis in either rats [17] or mice [18].

How the microbiome might induce rheumatic diseases

Specific bacteria are sometimes implicated in an immune-mediated disease. Beta hemolytic Streptococcus in rheumatic fever is a classic example. Additional examples include Campylobacter as a trigger for Guillain-Barré syndrome (reviewed in [19]) and Salmonella, Shigella, Yersinia or Campylobacter as triggers for reactive arthritis [20,21]. Some evidence suggests that Staphylococcus aureus could contribute to the causation of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis [22–24]. But what if research fails to implicate a specific bacterium as the cause of, say, rheumatoid arthritis? Can the microbiome still have a therapeutic implication?

Altering the microbiome with antibiotics and fecal transplants

Antibiotics can dramatically alter the microbiome, and broad spectrum antibiotics can ameliorate disease in animal models of immune-mediated inflammation [25]. Antibiotics have also demonstrated some benefit in selected patients with inflammatory bowel disease [26]. However, the benefit from antibiotics is transient, and bacteria will ultimately develop resistance. While fecal transplants have shown therapeutic efficacy in pseudomembranous colitis [27] and in ulcerative colitis [28,29], we doubt that the ultimate strategy to exploit the microbiome will be via a technique as crude as fecal transplantation, which carries the risk of transplanting a pathogen along with therapeutic bacteria.

The effect of diet on the microbiome

The bacteria which coexist within the human body are dependent on the food substrates provided to them. Thus, diet has a huge influence on the microbiome [30]. For many years, we and others had assumed that most claims of dietary benefit in a disease such as rheumatoid arthritis represented a placebo effect. After all, many more people claim to be gluten sensitive compared with the number of people who have documented celiac disease [31]. But what if eliminating gluten alters the microbiome, resulting in a therapeutic effect even in those who do not have gluten sensitivity? Physicians have an impoverished understanding of the role of diet in health and disease because diet is immensely complex, and thus a major challenge to study. We predict that well-controlled studies on diet will eventually reveal strategies that could profoundly affect outcomes in immune-mediated diseases.

Altering the microbiome with medications

While diet and possibly probiotics could change the intestinal ecosystem, it might also be possible to alter the dynamic between gut bacteria and the immune system without knowing anything about the detailed populations in that ecosystem. For example, intestinal mucus is a major barrier that limits the interaction between intestinal bacteria and both immune and non-immune cells in the gut (reviewed in [32]. A medication that increases mucus production in the intestine could therefore theoretically be used to treat rheumatic disease.

Sulfasalazine has some efficacy in treating the peripheral arthritis associated with spondyloarthritis [33]. A portion of its benefit might be its ability to reduce the number of bacteria in the gut by 100-fold [34]. Its efficacy might also relate to its ability to reduce gut permeability [35] such that bacterial products are less likely to distribute throughout the body from the intestine. Increased intestinal permeability is a feature of several rheumatic diseases including ankylosing spondylitis [36]. Other medications which limit the permeability of the intestine are also under study [37]. Assuming that the dispersion of bacterial products from the gut contributes to rheumatic disease, medications that affect the receptors, such as Toll-like receptors, which recognize these bacterial products could have a therapeutic niche in the rheumatologic armamentarium [38].

Conclusion

Many rheumatologists are old enough to have begun practicing medicine well before the emergence of biologics that could target a specific cytokine. The therapy of rheumatic diseases has been revolutionized by this approach. If we find that E. gallinarum is the cause of autoimmune hepatitis as is seen with lupus [16], or that P. gingivalis is causally related to rheumatoid arthritis (reviewed in [39–41]), we might advance to using vaccination against a specific organism; and thus we could experience a similar revolution. The ideal is to target a specific micro-organism, but even in the absence of this understanding of human disease, we anticipate that the microbiome will become a therapeutic target for many diseases that are mediated by the immune response.

Acknowledgements

Literature
  1. Morton AM, Sefik E, Upadhyay R et al. Endoscopic photoconversion reveals unexpectedly broad leukocyte trafficking to and from the gut. Proc Natl Acad Sci U S A 2014; 111: 6696–6701.
  2. Nakamura YK, Janowitz C, Metea C et al. Short chain fatty acids ameliorate immune-mediated uveitis partially by altering migration of lymphocytes from the intestine. Sci Rep 2017; 7: 11745.
  3. Hegazy AN, West NR, Stubbington MJT et al. Circulating and tissue-resident CD4+ T cells with reactivity to intestinal microbiota are abundant in healthy individuals and function is altered during inflammation. Gastroenterology 2017; 153: 1320–1337.e16.
  4. Belkaid Y, Harrison OJ. Homeostatic Immunity and the Microbiota. Immunity 2017; 46: 562–576.
  5. Thaiss CA, Levy M, Grosheva I et al. Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science 2018; 359: 1376–1383.
  6. Schrijver IA, Melief MJ, Tak PP, Hazenberg MP, Laman JD. Antigen-presenting cells containing bacterial peptidoglycan in synovial tissues of rheumatoid arthritis patients coexpress costimulatory molecules and cytokines. Arthritis Rheum 2000; 43: 2160–2168.
  7. Zhang X, Zhang D, Jia H et al. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med 2015; 21: 895–905.
  8. Hevia A, Milani C, López P et al. Intestinal dysbiosis associated with systemic lupus erythematosus. MBio 2014; 5: e01548-14.
  9. Costello ME, Ciccia F, Willner D et al. Brief report: intestinal dysbiosis in ankylosing spondylitis. Arthritis Rheumatol 2015; 67: 686–691.
  10. Manasson J, Shen N, Garcia Ferrer HR et al. Gut microbiota perturbations in reactive arthritis and postinfectious spondyloarthritis. Arthritis Rheumatol 2018; 70: 242–254.
  11. Scher JU, Ubeda C, Artacho A et al. Decreased bacterial diversity characterizes the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease. Arthritis Rheumatol 2015; 67: 128–139.
  12. Stoll ML, Kumar R, Morrow CD et al. Altered microbiota associated with abnormal humoral immune responses to commensal organisms in enthesitis-related arthritis. Arthritis Res Ther 2014; 16: 486.
  13. Guo Z, Zhang J, Wang Z et al. Intestinal microbiota distinguish gout patients from healthy humans. Sci Rep 2016; 6: 20602.
  14. Andréasson K, Alrawi Z, Persson A, Jönsson G, Marsal J. Intestinal dysbiosis is common in systemic sclerosis and associated with gastrointestinal and extraintestinal features of disease. Arthritis Res Ther 2016; 18: 278.
  15. Liu X, Zeng B, Zhang J et al. Role of the gut microbiome in modulating arthritis progression in mice. Sci Rep 2016; 6: 30594.
  16. Manfredo Viera S, Hiltensperger M, Kumar V et al. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science 2018; 359: 1156–1161.
  17. Asquith MJ, Stauffer P, Davin S et al. Perturbed mucosal immunity and dysbiosis accompany clinical disease in a rat model of spondyloarthritis. Arthritis Rheumatol 2016; 68: 2151–2162.
  18. Rehaume LM, Mondot S, Aguirre de de Cárcer D et al. ZAP-70 genotype disrupts the relationship between microbiota and host, leading to spondyloarthritis and ileitis in SKG mice. Arthritis Rheumatol 2014; 66: 2780–2792.
  19. Nachamkin I, Allos BM, Ho T. Campylobacter species and Guillain-Barré syndrome. Clin Microbiol Rev 1998; 11: 555–567.
  20. Granfors K, Jalkanen S, von Essen R et al. Yersinia antigens in synovial-fluid cells from patients with reactive arthritis. N Engl J Med 1989; 320: 216–221.
  21. Ajene AN, Fischer Walker CL, Black RE. Enteric pathogens and reactive arthritis: a systematic review of Campylobacter, Salmonella and Shigella-associated Reactive Arthritis. J Health Popul Nutr 2013; 31: 299–307.
  22. Stegeman CA, Tervaert JW, Sluiter WJ et al. Association of chronic nasal carriage of  Staphylococcus aureus and higher relapse rates in Wegener granulomatosis. Ann Intern Med 1994; 120: 12–17.
  23. Popa ER, Stegeman CA, Bos NA, Kallenberg CG, Tervaert JW. Staphylococcal superantigens and T cell expansions in Wegener's granulomatosis. Clin Exp Immunol 2003; 132: 496–504.
  24. Glasner C, de Goffau MC, van Timmeren et al. Genetic loci of Staphylococcus aureus associated with anti-neutrophil cytoplasmic autoantibody (ANCA)-associated vasculitides. Sci Rep 2017; 7: 12211.
  25. Nakamura YK, Metea C, Karstens L et al. Gut microbial alterations associated with protection from autoimmune uveitis. Invest Ophthalmol Vis Sci 2016; 57: 3747–3758.
  26. Khan KJ, Ullman TA, Ford AC et al. Antibiotic therapy in inflammatory bowel disease: a systematic review and meta-analysis. Am J Gastroenterol 2011; 106; 661–673.
  27. van Nood E, Vrieze A, Nieuwdorp M et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med 2013; 368: 407–415.
  28. Paramsothy S, Kamm MA, Kaakoush NO et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet 2017; 389: 1218–1228.
  29. Moayyedi P, Surette MG, Kim PT et al. Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial. Gastroenterology 2015; 149: 102–109.e6.
  30. David LA, Maurice CF, Carmody RN et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014; 505: 559–563.
  31. Aziz I. The global phenomenon of self-reported wheat sensitivity. Am J Gastroenterol 2018; 113: 945–948.
  32. Hansson GC. Role of mucus layers in gut infection and inflammation. Curr Opin Microbiol 2012; 15: 57–62.
  33. Clegg DO, Reda DJ, Abdellatif M. Comparison of sulfasalazine and placebo for the treatment of axial and peripheral articular manifestations of the seronegative spondylarthropathies: a Department of Veterans Affairs cooperative study. Arthritis Rheum 1999; 42: 2325–2329.
  34. West B, Lendrum R, Hill MJ, Walker G. Effects of sulphasalazine (Salazopyrin) on faecal flora in patients with inflammatory bowel disease. Gut 1974; 15: 960–965.
  35. Wang F, Graham WV, Wang T et al. Interferon-gamma and tumor necrosis factor-alpha synergize to induce intestinal epithelial barrier dysfunction by up-regulating myosin light chain kinase expression. Am J Pathol 2005; 166: 409–419.
  36. Mielants H, De Vos M, Goemaere S et al. Intestinal mucosal permeability in inflammatory rheumatic diseases. II. Role of disease. J Rheumatol 1991; 18: 394–400.
  37. Monteleone G, Neurath MF, Ardizzone S et al. Mongersen, an oral SMAD7 antisense oligonucleotide, and Crohn's disease. N Engl J Med 2015; 372: 1104–1113.
  38. Abdollahi-Roodsaz S, Joosten LA, Roelofs MF et al. Inhibition of Toll-like receptor 4 breaks the inflammatory loop in autoimmune destructive arthritis. Arthritis Rheum. 2007; 56: 2957–2967.
  39. Payne JB, Golub LM, Thiele GM, Mikuls TR. The link between periodontitis and rheumatoid arthritis: a periodontist's perspective. Curr Oral Health Rep 2015; 2: 20–29.
  40. Koziel J, Mydel P, Potempa J. The link between periodontal disease and rheumatoid arthritis: an updated review. Curr Rheumatol Rep 2014; 16; 408.
  41. Leech MT, Bartold PM. The association between rheumatoid arthritis and periodontitis. Best Pract Res Clin Rheumatol 2015; 29; 189–201.