Microbiome and Probiotics: Link to Arthritis

Medscape.com
Mohamed K. Bedaiwi, Robert D. Inman
Curr Opin Rheumatol. 2014;26(4):410-415.

Abstract

Purpose of review

The gut microbiome plays an integral role in the development and maintenance of the host immune system. Expanding knowledge about this microbial microenvironment has raised the possibility of new treatments based on this knowledge. In this review, we describe the recent evidence of the impact of the gut microbiome on arthritis and possible novel therapeutic approaches to alter the gut flora.

Recent findings

Recent studies support the growing evidence of microbiome as a causative agent underlying certain rheumatic diseases like ankylosing spondylitis and rheumatoid arthritis. There is intriguing yet still inconclusive evidence to support the use of probiotics as a treatment for these diseases.

Summary

There is recently a new level of understanding how the microbiome interacts with the immune system. Gene–environment interaction is another important element that sets the stage for initiation of autoimmune disease, which calls for further investigation. Probiotics could be an appealing therapeutic strategy, but further interventional studies exploring the dynamic interaction of microbiome and probiotics are still needed.


Introduction

Although most rheumatic diseases have a significant heritable component, predisposing genetic factors may require some environmental trigger to initiate the immunopathological events responsible for disease manifestation. A variety of investigations implicate a relationship between the gut microbiome and the development of certain rheumatic diseases. It is known that the microorganisms which populate the gastrointestinal tract profoundly influence the host immune system. However, despite intensive study of these organisms, there is as yet no definitive proof that bacteria play a causal role in rheumatoid arthritis (RA) or ankylosing spondylitis (AS). Yet, many studies of the microbiome have provided new understanding of the complexities of the host commensal microbiota.[1]

This article reviews the recent evidence for the potential role of microbiota in the pathogenesis of RA and AS, and addresses the possibility of therapeutic modulation of the gut microbiome.

Biology of Human Microbiota

Although many organs have their distinctive local microbial flora, the gastrointestinal tract has the largest proportion.[2] In 1676, Van Leeuwenhoek reported his discovery of microorganisms when he described ‘animalcules’ recovered from his mouth. It is now recognized that gut microorganisms may constitute up to 3 lbs of body weight. Microbial colonization has an important impact on the immune system development earlier in infant life. Organisms may play a protective role against certain autoimmune diseases, as exemplified by nondiabetic mice with an overrepresentation of Bacteroidetes in the microbiota.[3] In contrast, evidence has provided some links between the gut microbiome and the development of some rheumatic diseases. In addition to impacting the immune system, endogenous gut microbes may regulate the body weight by influencing the host’s metabolic and neuroendocrine homeostasis.[4]

Microbiome in Rheumatoid Arthritis

RA is a chronic systemic autoimmune inflammatory disease, with complex genetic and autoimmune factors contributing to it.[5] In the late 1970s, the importance of the histocompatability complex (HLA) region to the pathogenesis of RA was recognized, with HLA-Dw4 being more common among RA patients compared with healthy individuals.[6,7] This suggested the possibility of arthritogenic peptides being presented by proteins encoded by HLA-DRB1 genes. Subsequently, a large number of studies have examined and expanded the genetic basis of RA.

Yet, the genetic predisposition to RA does not predict disease development accurately. Twin and sibling studies have shed light on the genetic factors in RA. Monozygotic twins have a higher concordance than dizygotic twins for the development of RA. In two studies[8,9] of twins, the concordance for monozygotic twins was 13 versus 3.5% for dizygotic twins. This low percentage of concordance has suggested possible interaction between gene and environmental triggering factors.

Periodontal disease has been strongly linked to RA, with patients demonstrating a higher prevalence and severity of periodontal disease. Periodontal disease further shows correlation with disease activity, as patients with periodontitis have higher disease activity scores.[10] Furthermore, treatment of periodontitis has been shown in some studies to reduce disease activity as measured by Disease activity score28, Erythrocyte sedimentation rate, and serum TNF-α.[11] In a cross-sectional study of RA patients, recovery of Prevotella intermedia and Porphyromonas gingivalis in subgingival dental plaque as well as in synovial fluid supports a microbiome role in initiating or maintaining the chronic inflammation of RA.[12–14]

Studies of synovial fluid with PCR probes specific for the 16S rRNA demonstrated the presence of P. gingivalis DNA with greater frequency in RA patients than controls (15.7 versus 3.5%, P = 0.045), and RA patients in this study[15] demonstrated a correlation between dental health and burden of disease imposed by RA.

These recent findings have supported the recommendations by rheumatologist to encourage RA patients to obtain early, intensive dental care. In one study,[16] more advanced forms of periodontitis were found in early RA patients compared with controls, and RA patients had more missing teeth and greater periodontal friability on probing despite comparable oral hygiene. The characteristic oral organisms in this study were Tannerella forsythia subgingivally and Streptococcus anginosus supragingivally. The investigators concluded that in early RA, there is a greater risk of periodontal and alveolar bone disease.[16]

Breaking tolerance RA could occur in reaction to periodontal pathogens. Antibody reactivity to peptidylarginine deiminase (PADI) was higher in RA patients compared with individuals with periodontitis and controls.[17] Other than a potent citrullinating enzyme which might contribute to anti-CCP antibodies in RA, this report presents PADI as a possible therapeutic target as well.

Other than gut microbiota, lung organisms may have their own unique contribution to the host immune system. Earlier reports using spirometry and high-resolution computed tomography lung imaging found lung inflammation in autoantibody-positive individuals without inflammatory arthritis, and findings were similar to airway abnormalities seen in patients with early RA.[18]

That report has been followed with a recently published data to support the concept of a pathogenic role for lung microbes. Preclinical RA (i.e. anti-CCP-positive but with no clinical arthritis) is characterized by differences in lung microbiota in comparison to healthy control individuals.[19]

Further studies to define a potential role for Prevotella copri in the pathogenesis of RA utilized stool samples from RA patients and controls, and found that the presence of P. copri was strongly correlated with disease in recent-onset, untreated RA patients. Increases in Prevotella load correlated with a reduction in Bacteroides and a loss of reportedly beneficial microbes in these RA patients. In addition, unique Prevotella genes were found to be correlated with the activity of disease.[20]

Microbiome in Spondyloarthritis

The term ‘spondyloarthritis’ (SpA) refers to a group of disorders that includes AS, undifferentiated SpA, reactive arthritis, and the arthritis accompanying psoriasis and inflammatory bowel diseases. For the SpA group as a whole, there is a strong genetic relation with HLA-B27, especially for AS. However, the strongest genetic association (HLA-B27) accounts for only 30% of the heritability, thus a large part of the genetic susceptibility remains unknown and this has led to an intense effort to identify additional predisposing factors.[21]

In studying a possible role for microbiota in the pathogenesis of AS, it has been observed that active AS patients have a higher serum IgA Klebsiella pneumoniae in comparison with RA and healthy controls, whereas inactive AS patients had no such antibody elevation,[22] with possible flare of anterior uveitis in relation to Klebsiella colonization in bowel flora.[23] In these studies, it was noted that serum antibodies could be detected against pullulanase-D peptide, which contains a sequence having homology with HLA-B27.[22] However, subsequent studies did not provide confirmation of the specificity of anti-Klebsiella antibodies in AS.[24]

HLA-B27 may indirectly alter the bacterial flora. It has been demonstrated that transfected monocytes with HLAB-27 showed reduced proliferative response to endotoxin.[25,26]

Initial studies[27,28] of animal models which might link the genetic component of SpA to environmental factors found no spontaneous development of inflammatory process in HLA-B27 transgenic mice. On the other hand, rats transgenic for human HLA-B27 and β2-microglobulin spontaneously develop an inflammatory response in the form of nail changes, hair loss, and arthritis, thus mimicking the spectrum of clinical manifestations seen in SpA. These studies indicated the need for gut colonization with conventional bacterial flora in concert with genetic predisposition to initiate the inflammatory response.[29] Recently, an RA-like arthritis accompanied by a positive autoantibody profile has been developed in SKG mice.[30] SKG mice raised in a specific pathogen-free environment stayed healthy until exposed to curdlan (a β-1,3-glucan derived from the cell wall of yeast, fungi, and bacteria). Thereafter, the mice developed peripheral and axial arthritis associated with extra-articular manifestations similar to SpA.[1]

Inflammatory bowel disease (IBD) shares a wide spectrum of manifestations with HLA-B27-related SpA. There is now mounting evidence that certain bowel flora play a causative agent in IBD,[31,32] in concert with numerous genetic factors like NOD2 and CARD9 to initiate the immune response.[33,34] Many observational studies have supported the notion of subclinical gut inflammation commonly occurring in SpA, with as many as two-thirds of SpA patients demonstrating this phenomenon. In a recent study,[35] the absence of CD14+ macrophages and a large increase in CD163+ (M2) macrophages were observed in AS, supporting the concept of subclinical ileal inflammation in the AS patient. It has been observed that the eventual development of symptomatic IBD occurs in approximately 6.5% of SpA patients presenting without symptoms of IBD.[36]

The prevalence of gut inflammation in nonradiographic axial SpA and AS was comparable in one study[37] which addressed this question, although other parameters including male sex, high disease activity, restricted spinal mobility, and younger age were independently associated with gut involvement.

In a recently published observational study,[38] the results showed higher antiflagellin antibody-positivity rates and acute-phase reactants in AS patients compared with mechanical back pain patients.

In one recent report, a cohort of 68 patients with AS had an ileocolonoscopy and MRI of the sacroiliac joints, and the result showed higher radiological inflammatory MRI scores in axial SpA patients with chronic gut inflammation compared with axial SpA patients showing normal gut histology. These findings indicated that greater Sacroiliac joint bone marrow edema was correlated with chronic gut inflammation.[39]

Although there have been numerous studies suggesting a link between microbiome and SpA, the precise mechanism by which those environmental factors initiate the immune response still needs further investigation.

Probiotics as a Therapeutic Modulation of the Microbiota

Probiotics are defined as ‘live microorganisms which when administered in adequate amounts confer a health benefit on the host’.[40] Evidence suggests that probiotic bacteria modulate both innate and adaptive immunity in the host and may have therapeutic applications of chronic inflammatory diseases. Lactic acid bacteria (LAB) and bifidobacteria are the most known types of bacteria used as probiotics, but certain yeasts and bacilli have also been used.

Probiotics as a modality of treatment will require extensive study to understand their anti-inflammatory effect, long-term efficacy, and the mechanism of action by which they exert their therapeutic role.

Bifidobacterium animalis subspecies lactis IPLA R1 and Bifidobacterium longum IPLA E44 strains have been tested for their safety and their ability to modulate the intestinal microbiota in vivo. The oral administration of B. animalis IPLA R1 and B. longum E44 is considered nontoxic, and has the capacity to modulate the intestinal microbiota of rats by influencing short-chain fatty acids and the bifidobacterial population levels.[41] Oral administration of Lactobacillus casei suppresses the type II collagen-reactive effector function of Th1-type cellular and humoral immune responses.[42] When L. casei was tested in rat collagen-induced arthritis (CIA), arthritis scores and proinflammatory cytokine levels were observed to be lower compared with control rats and with CIA treated with indomethacin.[43] Previous evidence had showed the ability of L. casei to suppress the type II collagen-reactive effector function of Th1-type cellular and humoral immune responses in rat arthritis.[42] HLA-B27 transgenic rats with Bacteroides vulgatus-induced colitis have been treated with antibiotics to prevent and treat colitis, Lactobacillus rhamnosus GG prevented the relapse of colitis with significantly reduced histologic scores.[44]

But there are few studies on the efficacy of probiotics in human arthritis. The effect of L. casei was recently tested in RA patients and showed significantly lower serum proinflammatory cytokines (TNF-α, IL-6, and IL-12) in the probiotic-treated group, with higher level regulatory cytokine (IL-10) as well; clinically, the disease activity score was significantly decreased with L. casei 01 supplementation.[45] Studying the possible mechanisms by which L. casei protects against RA progression showed the effect of oral administration of L. casei to suppress the type II collagen-reactive effector function of Th1-type cellular and humoral immune responses in arthritic inflammation.[42]

One pilot study[46] assessed the effect of probiotics (Lactobacillus acidophilus and Lactobacillus salivarius) in patients with quiescent ulcerative colitis and active SpA, and there was a significant reduction of Bath AS Disease Activity Index and Visual analogue scale score. Despite a theoretical rationale for this therapy in AS patients, another randomized controlled trial of a 12-week course of an oral probiotic in active AS showed no significant benefit over placebo in any of the core outcome domains.[47]

As there are increasingly sophisticated tools for characterizing the gut microbiome, the complex interrelationships between gut flora, immune response, and arthritis will gradually be resolved.

Conclusion

Microbiota have a substantive impact on the immune system development and it remains an attractive, but unproven, theory that predisposing genetic factors may interact with microbiota to trigger peripheral or axial arthritis. Table 1 presents a list of pathogens that have been linked to arthritis.

Our review summarizes the recent evidence of oral and lung organisms in relation to RA. Regarding SpA, the recent finding of a correlation of axial inflammation, as detected by MRI, with subclinical gut inflammation may support the previous finding of gut–joint interactions in HLA-B27 transgenic rats. More evidence is needed to confirm the impact of the role of microbiota in arthritis, and further studies are needed to evaluate the role of probiotics as a novel therapeutic modality for arthritis.

References

Yeoh N, Burton JP, Suppiah P, et al. The role of the microbiome in rheumatic diseases. Curr Rheumatol Rep 2013; 15:903–907.

Simon GL, Gorbach SL. Intestinal flora in health and disease. Gastroenterology 1984; 86:174–193.

Maslowski KM, Mackay CR. Diet gut microbiota and immune responses. Nat Immunol 2011; 12:5–9.

SanzF Y. Santacruz A, Gauffin P. Gut microbiota in obesity and metabolic disorders. Proc Nutr Soc 2010; 69:434–441.

Klareskog L, Catrina AI, Paget S. Rheumatoid arthritis. Lancet 2009; 373:659–672.

McMichael AJ, Sasazuki T, McDevitt HO, et al. Increased frequency of HLA-Cw3 and HLA-Dw4 in rheumatoid arthritis. Arthritis Rheum 1977; 20:1037.

Stastny P. Association of the B-cell alloantigen DRw4 with rheumatoid arthritis. N Engl J Med 1978; 298:869.

Aho K, KoskenvuoM, Tuominen J, et al. Occurrence of rheumatoid arthritis in a nationwide series of twins. J Rheumatol 1986; 13:899–902.

Silman AJ, MacGregor AJ, Thomson W, et al. Twin concordance rates for rheumatoid arthritis: results from a nationwide study. Br J Rheumatol 1993; 32:903–907.

Jayatilake JAMS, Rajapakse S, Weerasinghe IE, et al. Oral hygiene and periodontal status in a group of patients with rheumatoid arthritis. Indian J Rheumatol 2011; 6:111–115.

Ortiz P, Bissada N, Palomo L, et al. Periodontal therapy reduces the severity of active rheumatoid arthritis in patients treated with or without tumor necrosis factor inhibitors. J Periodontol 2009; 80:535–540.

Martinez-Martinez RE, Abud-Mendoza C, Patiño-Marin N, et al. Detection of periodontal bacterial DNA in serum and synovial fluid in refractory rheumatoid arthritis patients. J Clin Periodontol 2009; 36:1004–1010.

Moen K, Brun J, Valen M, et al. Synovial inflammation in active rheumatoid arthritis and psoriatic arthritis facilitates trapping of a variety of oral bacterial DNAs. Clin Exp Rheumatol 2006; 24:656–663.

Moen K, Brun JG, Eribe ERK, et al. Oral bacterial DNAs in synovial fluids of arthritis patients. Microb Ecol Health Dis 2005; 17:2–8.

Reichert S, Haffner M, Keyßer G, et al. Detection of oral bacterial DNA in synovial fluid. J Clin Periodontol 2013; 40:591–598. doi: 10.1111/jcpe.12102.

* DNA of P. gingivalis can be found in synovial fluid, and oral bacteria may play a role in the pathogenesis of arthritis with a correlation between dental health and burden of disease imposed by RA.

Wolff B, Berger T, Frese C, et al. Oral status in patients with early rheumatoid arthritis: a prospective, case–control study. Rheumatology (Oxford) 2014; 53:526–531.

* A prospective case–control study showed increased loss of periodontal attachment and alveolar bone in patients with early RA; patients should be encouraged to obtain early intensive dental care.

Quirke AM, Lugli EB, Wegner N, et al. Heightened immune response to autocitrullinated Porphyromonas gingivalis peptidylarginine deiminase: a potential mechanism for breaching immunologic tolerance in rheumatoid arthritis. Ann Rheum Dis 2014; 73:263–269. doi: 10.1136/annrheumdis-2012-202726.

Demoruelle MK, Weisman MH, Simonian PL, et al. Brief report: airways abnormalities and rheumatoid arthritis-related autoantibodies in subjects without arthritis: early injury or initiating site of autoimmunity? Arthritis Rheum 2012; 64:1756–1761. doi: 10.1002/art.34344.

Demoruelle MK, Norris JM, Holers VM, et al. The lung microbiome differs in asymptomatic subjects at elevated risk of future rheumatoid arthritis compared with healthy control subjects. Ann Am Thorac Soc 2014; 11 (Suppl. 1):S74.

* This study showed the differences of lung microbiota in preclinical RA (anti-CCPpositive with no clinical arthritis) compared with the healthy controls, considering the lung as a possible source of pathogenic microbiota.

Bernard N J. Rheumatoid arthritis: Prevotella copri associated with new-onset untreated RA. Nat Rev Rheumatol 2014; 10:2. doi: 10.1038/nrrheum.2013.187.

* This study reported Prevotella copri as strongly correlated with new-onset untreated rheumatoid arthritis and identified Prevotella genes correlated with the disease.

Breban M. Genetics of spondyloarthritis. Best Pract Res Clin Rheumatol 2006; 20:593–599.

Tani Y, Tiwana H, Hukuda S, et al. Antibodies to Klebsiella, Proteus, and HLAB27 peptides in Japanese patients with ankylosing spondylitis and rheumatoid arthritis. J Rheumatol 1997; 24:109–114.

Ebringer R. Acute anterior uveitis and faecal carriage of Gram negative bacteria. Br J Rheumatol 1988; 27 (Suppl. 2):42–45.

Stone MA, Payne U, Schentag C, et al. Comparative immune responses to candidate arthritogenic bacteria do not confirm a dominant role for Klebsiella pneumonia in the pathogenesis of familial ankylosing spondylitis. Rheumatology (Oxford) 2004; 43:148–155.

Penttinen MA, Holmberg CI, Sistonen L, et al. HLA-B27 modulates nuclear factor kappaB activation in human monocytic cells exposed to lipopolysaccharide. Arthritis Rheum 2002; 46:2172–2180.

Rosenbaum JT, Davey MP. Time for a gut check: evidence for the hypothesis that HLA-B27 predisposes to ankylosing spondylitis by altering the microbiome. Arthritis Rheum 2011; 63:3195–3198.

Taurog JD, Richardson JA, Croft JT, et al. The germfree state prevents development of gut and joint inflammatory disease in HLA-B27 transgenic rats. J Exp Med 1994; 180:2359–2364.

Nickerson CL, Hanson J, David CS. Expression of HLA-B27 in transgenic mice is dependent on the mouse H-2D genes. J Exp Med 1990; 172:1255–1261.

Khare SD, Luthra HS, David CS. Spontaneous inflammatory arthritis in HLAB27 transgenic mice lacking beta 2-microglobulin: a model of human spondyloarthropathies. J Exp Med 1995; 182:1153–1158.

Sakaguchi S, Takahashi T, Hata H, et al. SKG mice, a new genetic model of rheumatoid arthritis. Arthritis Res Ther 2003; 5 (Suppl. 3):10.

Friswell M, Campbell B, Rhodes J. The role of bacteria in the pathogenesis of inflammatory bowel disease. Gut Liver 2010; 4:295–306.

Tannock GW. The bowel microbiota and inflammatory bowel diseases. Int J Inflamm 2010; 2010:954051.

Zhernakova A, Festen EM, Franke L, et al. Genetic analysis of innate immunity in Crohn’s disease and ulcerative colitis identifies two susceptibility loci harboring CARD9 and IL18RAP. Am J Hum Genet 2008; 82:1202–1210.

Waterman M, Xu W, Stempak JM, et al. Distinct and overlapping genetic loci in Crohn’s disease and ulcerative colitis: correlations with pathogenesis. Inflamm Bowel Dis 2011; 17:1936–1942.

Ciccia F, Alessandro R, Rizzo A, et al. Macrophage phenotype in the subclinical gut inflammation of patients with ankylosing spondylitis. Rheumatology 2014; 53:104–113.

* This study supports the subclinical gut inflammation in ankylosing spondylitis by the absence of CD14(+) macrophages together with the expansion of resolution phase and M2 macrophages.

De Vos M, Mielants H, Cuvelier C, et al. Long-term evolution of gut inflammation in patients with spondyloarthropathy. Gastroenterology 1996; 110:1696–1703.

Van Praet L, Van den Bosch FE, Jacques P, et al. Microscopic gut inflammation in axial spondyloarthritis: a multiparametric predictive model. Ann Rheum Dis 2013; 72:414–417.

Wallis D, Asaduzzaman A, Weisman M, et al. Elevated serum antiflagellin antibodies implicate subclinical bowel inflammation in ankylosing spondylitis. Arthritis Res Ther 2013; 15:R166.

* This observational study supports the theory of mucosal dysregulation as a mechanism underlying the pathophysiology of AS; patients with AS have higher antiflagellin antibodies and median antibody levels compared with those with mechanical back pain.

Van Praet L, Jans L, Carron P, et al. Degree of bone marrow oedema in sacroiliac joints of patients with axial spondyloarthritis is linked to gut inflammation and male sex: results from the GIANT cohort. Ann Rheum Dis 2014; 73:1186–1189. doi: 10.1136/annrheumdis-2013-203854.

* This GIANT cohort supported the links between chronic gut inflammation and axial spondyloarthritis. A higher bone marrow edema of the sacroiliac joints was observed in axial spondyloarthritis patients showing chronic gut inflammation compared with axial spondyloarthritis patients with normal gut histology.

Evaluation of health and nutritional properties of powder milk and live lactic acid bacteria. Food and Agriculture Organization of the United Nations and World Health Organization Expert Consultation Report; 1–4 October 2001; Cordova, Argentina.

Salazar N, Binetti A, Gueimonde M, et al. Safety and intestinal microbiota modulation by the exopolysaccharide-producing strains Bifidobacterium animalis IPLA R1 and Bifidobacterium longum IPLA E44 orally administered to Wistar rats. Int J Food Microbiol 2011; 144:342–351.

So JS, Kwon HK, Lee CG, et al. Lactobacillus casei suppresses experimental arthritis by downregulating T helper 1 effector functions. Mol Immunol 2008; 45:2690–2699.

Amdekar S, Singh V, Singh R, et al. Lactobacillus casei reduces the inflammatory joint damage associated with collagen-induced arthritis (CIA) by reducing the proinflammatory cytokines: Lactobacillus casei: COX-2 inhibitor. J Clin Immunol 2011; 31:147–154.

Dieleman LA, Goerres MS, Arends A, et al. Lactobacillus GG prevents recurrence of colitis in HLA-B27 transgenic rats after antibiotic treatment. Gut 2003; 52:370–376.

Vaghef-Mehrabany E, Alipour B, Homayouni-Rad A, et al. Probiotic supplementation improves inflammatory status in patients with rheumatoid arthritis. Nutrition 2014; 30:430–435. pii: S0899-9007(13)00439-5.

* In this study, Rheumatoid Arthritis Activity Score was significantly decreased with probiotic supplementation (Lactobacillus casei 01); this presents the probiotics as a modality of treatment for rheumatoid arthritis.

Sanges M, Valente G, Rea M, et al. Probiotics in spondyloarthropathy associated with ulcerative colitis: a pilot study. Eur Rev Med Pharmacol Sci 2009; 13:233–234.

Jenks K, Stebbings S, Burton J, et al. Probiotic therapy for the treatment of spondyloarthritis: a randomized controlled trial. J Rheumatol 2010; 37:2118–2125. doi: 10.3899/jrheum.100193.

Story Source

Comments Are Closed