Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Dec 11.
Published in final edited form as: Gut. 2014 Nov 21;64(2):332–341. doi: 10.1136/gutjnl-2014-308514

Does the microbiota play a role in the pathogenesis of autoimmune diseases?

Mairi H McLean 1, Dario Dieguez Jr 2, Lindsey M Miller 2, Howard A Young 1
PMCID: PMC6288812  NIHMSID: NIHMS999071  PMID: 25416067

Abstract

The microbiota of the human metaorganism is not a mere bystander. These microbes have coevolved with us and are pivotal to normal development and homoeostasis. Dysbiosis of the GI microbiota is associated with many disease susceptibilities, including obesity, malignancy, liver disease and GI pathology such as IBD. It is clear that there is direct and indirect crosstalk between this microbial community and host immune response. However, the precise mechanism of this microbial influence in disease pathogenesis remains elusive and is now a major research focus. There is emerging literature on the role of the microbiota in the pathogenesis of autoimmune disease, with clear and increasing evidence that changes in the microbiota are associated with some of these diseases. Examples include type 1 diabetes, coeliac disease and rheumatoid arthritis, and these contribute significantly to global morbidity and mortality. Understanding the role of the microbiota in autoimmune diseases may offer novel insight into factors that initiate and drive disease progression, stratify patient risk for complications and ultimately deliver new therapeutic strategies. This review summarises the current status on the role of the microbiota in autoimmune diseases.

INTRODUCTION

The concept of the human metaorganism arose with the realisation that we harbour many trillions of microbes on and within the human body.1 These microbes are located at the host-environmental interface, such as the skin, the GI tract, the genital tract and respiratory mucosal barrier. All genes of our microbial cohabitants constitute the micro-biome, and this microbiome outweighs the genetic contribution of the host by 10-fold.1 Our personal microbial world is rich in diversity and many thousands of species survive and thrive within us. The sheer enormity of this microbial community has become apparent over the last decade as technology, such as sophisticated sequencing techniques and high throughput technology, has allowed for the identification of the microbial community and analysis of its function. It is clear that our micro-biota is not a mere bystander; they have coevolved with us and are pivotal to normal development and homoeostasis, from a metabolic, trophic and protective capacity.2,3

The intestinal microbiota interacts with the adjacent mucosal environment directly, impacts intestinal permeability, and influences local and systemic inflammatory activity.4 There is also an indirect crosstalk between the microbial community and the host via their metabolites; for example, digestion of plant polysaccharides by gut bacteria yields short chain fatty acids and these in turn modulate host mucosal immune response by various mechanisms, including promotion of regulatory T cells.5 The composition of our microbiota is not static, but changes with age, geography and is influenced by many external factors, such as diet and medication.68

Large global consortia such as the US Human Microbiome Project seek to provide knowledge on microbial composition in health and disease and it is clear that an appreciable interindividual and intraindividual variation exists, influenced by many external factors.9 It is now known that alteration in the balance of intestinal microbial species leading to a dysbiosis is associated with many disease susceptibilities. Examples of this include obesity,4 multiple sclerosis (MS),10 malignancy,1113 liver disease,14,15 and GI pathology such as IBD,16 but the precise mechanism of this microbial influence in disease pathogenesis remains elusive and is now becoming a major research focus.

Within this exploration of the relationship between the gut microbiota and disease, there has been interest in the role of the microbiota in the pathogenesis of systemic and organ targeted auto-immune disease. Autoimmune diseases are characterised by serological evidence of autoantibodies, pronouncing lack of tolerance and self-directed immune response. Several autoimmune diseases such as type 1 diabetes (T1D) and rheumatoid arthritis (RA) contribute significantly to global morbidity and mortality. Understanding the role of the microbiota in these diseases may offer novel insight into factors that initiate and drive disease progression, stratify patient risk for complications and ultimately could deliver new therapeutic strategies.

Reflecting the importance of this topic, the role of the microbiota in autoimmunity was the subject of a 2014 National Institutes of Health (NIH) symposium, cosponsored by the Society for Women’s Health Research. This review aims to summarise current data on the role of the microbiota on auto-immunity, and concludes by summarising points raised within the closing discussion at the NIH symposium.

TYPE 1 DIABETES

T1D is a chronic, proinflammatory autoimmune disorder characterised by immune-mediated destruction of the pancreatic β cells, resulting in insulin deficiency and hyperglycaemia. Clinical manifestation of T1D usually presents in childhood and adolescence and incidence of this disease continues to increase globally.17,18 There is clear evidence of a genetic susceptibility to T1D.1921 However, given the 50% concordance rate in monozygotic twins22 and the fact that immigrants exhibit risk associated with place of residence rather than origin23 there clearly is a central role for environmental factors in T1D pathogenesis. This has been explored extensively and the main aspects of environmental risk focus on diet, including neonatal exposure to bovine derived milk products, age at weaning and early exposure to gluten (reviewed in ref.24) as well as a potential infective component such as childhood viral infection, particularly Enterovirus given the seasonal timing of clinical presentation.25

In light of the emerging evidence that the gut microbiome has a strong and broad impact on health and disease, the question of whether the gut microflora could impact T1D has arisen. Indeed, a growing evidence base from animal and human studies suggests that changes in the gut microbiome may precede the onset of T1D and are associated with progression from detectable autoantibody levels in high-risk asymptomatic individuals through to those with clinical disease and this has been reviewed elsewhere.2630

Several animal models of T1D exist.31 Of these, the non-obese diabetic (NOD) mouse and the biobreeding diabetes-prone rodent model exhibit similar genetic predisposition and pathological disease progression to human T1D and have been used to explore the relationship between the gut microbiome and T1D.

Manipulation of the gut microbiota by different approaches, such as treatment with antibiotics,3235 exposure to acidified water,36,37 exposure to pathogenic and non-pathogenic bacterial strains,36,3840 housing in germ-free conditions,32,41 along with temporal analysis of faecal microbiota preceding and during disease development has revealed the importance of the colonic microbial community in the pathogenesis of T1D. However, many conflicting reports exist in the literature and highlight the complexity of this association. It is clear from the evidence that the gut microbial community influences host immunity and this could ultimately aid emergence of disease. For example, Wen et al32 revealed the importance of crosstalk between host and gut microbiota in T1D pathogenesis. MyD88 (myeloid differentiation primary response protein 88) is pivotal to bacterial sensing and downstream signalling host innate immune response, and genetic silencing of this bacterial sensor in specific pathogen-free NOD mice interrupted development of T1D as compared with wild type controls. In contrast, the same genetically altered mice, either raised in germ-free conditions or treated with antibiotics to disrupt gut commensal bacteria, developed disease. This indicates protection was mediated by a constituent of their gut microbial community although the underlying mechanisms are not fully understood. Certainly, changes in the intestinal microbiota composition were demonstrated in conjunction with loss of the ability to sense microbes via MyD88. A follow-on study showed that faecal bacterial transplant from the MyD88 null protected mice conferred protection to the wild type diabetes-prone strain, and altered mucosal immunity and faecal microbial composition of the recipient.40 The importance of innate pattern recognition receptors in microbial sensing of the gut microbiome in directing the downstream host immune response and development of T1D has been validated elsewhere.34,42 Alkanani et al34 identified a crucial role of an additional innate microbial sensor upstream of MyD88, Toll like receptor (TLR)3, but not TLR9 in this capacity to modulate the emergence of T1D.

The pH of drinking water, particularly low acidity, influences the composition of the gut microbiome and incidence of T1D.36,37 Exposure of female NOD mice to acidified water resulted in a differential induction speed and severity of insulitis and hyperglycaemia, associated with intestinal dysbiosis, along with increased gut and systemic proinflammatory status.36 Conversely, Wolf et al37 reported an increased incidence of T1D in mice exposed to neutral water, with a protective role seen along with exposure to acidified water. The differences in study design suggest that the timing of microbial disruption, in this case through exposure to acidified water, is a key consideration.37 Alteration of the microbiome at a very early age, such as in the newborn period when the gut microbiota is being established and thus long before onset of disease, may impact subsequent disease induction in genetically susceptible individuals.

Exposure to vancomycin, an antibiotic to specifically target Gram-positive bacteria, in infant and adult NOD mice was associated with a decreased incidence of T1D, and lower levels of blood glucose and insulitis scores, respectively.33 Notably, a single species, namely Akkermansia muciniphila emerged as a dominant potentially protective species in this context. This protective effect of antibiotic treatment in diabetes-prone rodents has been long recognised.43,44

Autoimmune disease tends to be prevalent in female mice preferentially. Could the gut microbiota influence this gender-specific preponderance? Markle et al39 reported that microbial exposures early in life impacts sex hormone levels and alters progression to T1D in the NOD mouse. Indeed, transplant of faeces from adult NOD mice into immature female mice led to increase in testosterone and protection from T1D.

How does this data from preclinical models translate to human disease? There have been several human studies that have confirmed the association between the gut microbiota and risk of T1D.45 The main outcomes from these studies are presented in table 1. Children with T1D have a low abundance of butyrate-producing bacteria.46,47 Gut bacterial diversity lacks stability, confers differential changes over time in islet autoantibody-positive children as compared with nonautoimmune matched controls and children with T1D have more variation between individuals.48 As such, children with propensity to autoimmune diabetes yield an increase in faecal Bacteriodetes and reduction in Firmicutes over time from their early childhood years, potentially even before T1D clinically develops, representing a composition that is opposite to that seen in control subjects.46,48,49

Table 1.

The main outcomes from studies assessing the human gut microbiome in T1D (faecal stream analysis)

Association with T1D compared with control Reference
↓ Butyrate producing bacteria 46, 47
↓ Mucin degrading bacteria (Akkermansia sp) 47
Bacterial metabolic functional capabilities differ 47
Bacterial diversity lacks stability 48
↑ Bacteroidetes, ↓ Firmicutes preceding disease onset 46, 48, 49, 51
Interactions between bacteria distorted 50
Change in microbiome may revert with glucose normalizing treatment 51
No single causative agent identified

TID, type 1 diabetes.

Interestingly, akin to that found in the preclinical models, Brown et al reported a reduction in Akkermansia spp in those with early disease.47 As such, a greater proportion of these mucin-degrading bacteria species, as well as butyrate secreting bacteria, were observed among healthy controls when compared with a small number of T1D cases at time of clinical presentation. In comparison, bacteria capable of producing non-butyrate short chain fatty acids were higher among T1D cases.47 From a mechanistic perspective, metabolic focused gene expression analysis of the microbiome revealed that gut bacteria display different metabolic functional capabilities between the two groups.

However, this association is far from clear as a recent prospective stool collection study on young children did not confirm these findings of change in microbial diversity in those who developed anti-islet autoimmunity. However, despite this, the interactions between gut microbes were distorted in this group.50 In another study, although newly diagnosed children were identified as carrying an increase in Bacteroides, this dysbiosis had returned to the status of control subjects in those who had received 2 years of glucose normalising treatment.51

Overall, most would agree that T1D is associated with a change in gut microbial composition. Certainly, it seems that no single species from the gut microbial community has emerged as a causative agent. Rather, in genetically susceptible individuals, there is emerging evidence that dysbiosis within the gut micro-biota and interruption of microbial colonisation in early life, maybe even as early as birth or the neonatal period, is associated with emergence and progression of T1D. In line with this, babies born by caesarean section have a >20% increased risk of developing T1D.52,53 It has been reported that birth mode impacts infant intestinal colonisation.54 The speculation is that caesarean delivery is associated with a lack of exposure to maternal microbiota and impacts infant intestinal colonisation conferring risk of future autoimmunity in genetically susceptible individuals. In fact, it may be that environmental influences on the intestinal microbiota can even extend to in utero exposure. NOD mice given a broad spectrum antibiotic cocktail during gestation bore offspring with a lower gut microbial diversity and a modulation of T cell phenotype in the mesenteric lymph nodes (increased CD3+CD8+ T cells) and Peyer’s patches of the intestine (reduced CD4+CD25+, but not Foxp3+ Treg subgroup). However, this only impacted emergence of hyperglycaemia to a minor level at 20 weeks of age and this risk did not persist into later life.35

The next consideration is whether a particular gut microbial community is linked to cause or effect in disease pathogenesis. Is the gut microbial dysbiosis an initiator of T1D, a perpetrator of increasing progression or a consequence of other pathological features? This remains unanswered. The immune mechanisms involved in islet cell destruction have been extensively studied and include pathogenic T cells, shift in B cell phenotype, features of antigen presentation, and distorted immunoregulatory mechanisms.55 All the human studies reporting an association between altered gut microbiota and T1D have not explored this aspect of disease pathogenesis, likely a consequence of easy access to faecal sampling, offset against the invasive nature of mucosal biopsy that would not normally be pursued as part of diagnosis and management per se. From preclinical animal models, it is clear that changes in gut micro-biota or GI microbial exposures are associated with differential host immune response, including change in splenic or GI mucosa T cell phenotype,3638,40,56 for example, modulation of T helper cell (Th)17 response. Whether this Th17 association is pathogenic or protective, remains under debate.57 Additionally, it has been shown that breakdown of the GI epithelial barrier integrity is present in T1D, with increased gut permeability.5861

It is inherently difficult to assess causality in human studies for several reasons; T1D is an early onset disease, with clinical presentation after destruction of islets has occurred. The preclinical phase of early islet autoimmunity is asymptomatic, and there is no biomarker that will predict disease in the general population. The aetiology is multifactorial and it is difficult to fully account for confounding factors. In addition, all human studies to date have used faecal samples for analysis and the question of whether this, as opposed to mucosal biopsy derived analysis, reflects the true status of the microbiome in T1D remains unanswered.

Several large global collection consortia are ongoing that will yield powerful data from prospectively collected data. There are several of these, for example, The Environmental Determinants of Diabetes in the Young study, and Diabetes Prediction and prevention Project study. The ultimate question is whether the colonic microbiota can be manipulated to therapeutic advantage for T1D. More likely, this strategy may be of greater benefit to prevent the onset of T1D in high-risk individuals, such as those receiving antibiotics or other treatments in the neonatal period that may alter gut microbial acquisition and increase risk of T1D in later life.

COELIAC DISEASE

Coeliac disease, like other autoimmune conditions, requires genetic susceptibility and environmental influences.62,63 This autoimmune disease is unique in that the main environmental factor is known, well characterised and therapeutically targeted. This environmental trigger in question is dietary gluten, derived from wheat and other related grains. Gluten, composed of gliadin peptides and glutenin, evokes a predominantly T cell mediated mucosal response in the proximal small bowel,64 with the cytokine interleukin (IL) 15 playing a pivotal role in the immunopathogenesis.65 This results in the characteristic pathological characteristics of progressive villous atrophy, distorted crypt architecture and increase in intraepithelial cells, leading to a reduction in absorptive capacity and emergence of GI and extraintestinal symptoms.62,63,66 However, there is often a lag of many years after gluten exposure until disease manifests serologically or clinically. Indeed, adult onset coeliac is not uncommon, and therefore this suggests that additional environmental influences are required in coeliac disease pathogenesis.

Concordance rates amongst monozygotic twins are high at more than 80%, compared with 10% in dizygotic twins,67,68 highlighting the importance of genetic susceptibility in this disease pathogenesis. HLA class II haplotype DQ2 or DQ8 are the most characterised genetic determinant.62,63 Carriage of these haplotypes plays a pivotal role in the presentation of the gliadin peptides to CD4+ T cells. This is not the whole story, with genome wide association studies and high throughput technology identifying many other susceptibility genes.6971

Coeliac disease is thought to affect 1% of the global population, and has been increasing in prevalence at a striking rate; doubling over 20 years in a Finnish population72 and increasing fourfold in a US population.73

There are well-characterised autoantibodies available for serological diagnosis and screening, namely tissue transglutaminase IgA antibody and anti-endomysial IgA antibody. Both of these display high specificity and sensitivity.62,63,66

As a disease of the GI tract, there has been florid interest over several years as to whether the gut microbiota could be implicated in the pathogenesis of coeliac disease.62,74 Given the proximal location of pathology in coeliac disease, the microbiota of the duodenum has been the focus of investigation in this context. Despite the hostile conditions of the proximal small bowel with fluctuating pH, digestive enzymes and bile, and robust peristalsis, a distinctive collection of bacteria appear to survive in this environment, dominated by Streptococci, Bacteroidetes, Proteobacteria and clusters of Clostridium sp.7578 Initial analysis employed conventional culturing or limited molecular techniques and reported differences in duodenal mucosal biopsy or faecal stream bacteria associated with coeliac disease.74,7984 There has been no unifying pattern to identify a distinct bacterial composition or diversity that marks presence of coeliac disease, nor successful treatment. Overall, there appears to be a trend for abundance in Firmicutes and Bacteroidetes over several studies, in adults and children, respectively. However, there are other reports with opposing observations. The inconsistency of the findings in these studies is a reflection of several issues; geographical difference with undoubted impact on diet (dictated by culture) and genetic susceptibilities, differences in experimental methodology including culture versus culture-independent, the low number of patients included in analysis, and the origin of the material to be tested, that is, faecal versus mucosal biopsy.

With the advent of increasingly sophisticated technology, most recent analyses have used sequencing and other high-throughput molecular analysis and yielded conflicting results with no dramatic or distinctive dysbiosis of the duodenal micro-biota at either the phylum or genus level in children with coeliac disease compared with healthy controls.7678

Therefore the debate on whether the microbiota is associated with disease pathogenesis is ongoing. If a component of the intestinal microbiota was a driving causative factor for initiation and progression of coeliac disease, one may expect an obvious candidate to emerge from analysis on adults and children, and to revert to that seen in an individual without coeliac disease, with successful treatment. This has not as yet emerged to date, but this is still an active field and therefore the debate goes on.

Nevertheless, there have been some published reports of how changes in the intestinal microbiome may influence underlying mucosal immune response. Sanchez et al85 used an in vitro system to show that exposure of Caco-2 cells to digested gliadin and specific Bacteriodes sp resulted in increased proinflammatory cytokine profile and disruption of permeability. Exposure of dendritic cells to intestinal bacterial species, such as Enterobacteria or Bifidobacteria, led to altered phenotype and function. When these cells were subsequently cocultured with Caco-2 epithelial cells, an altered expression of proteins involved in intestinal permeability was identified.86 When considering animal models of coeliac disease, there is no spontaneous model in small rodents. An induced rat model has been widely used (germ-free Wistar rats exposed to gliadin immediately after birth), along with transgenic mice exhibiting HLA genetic susceptibilities akin to human disease.87 These in vivo models have been employed to understand the underlying immune activity in coeliac disease initiation and progression. With regard to the role of the microbiota in this process, exposure to specific bacterial strains in vivo does impact epithelial permeability and underlying mucosal immunity. For example, administration of the intestinal commensal Bifidobacterium longum to the induced rat model is protective to emergence of disease and associated with increased mucosal anti-inflammatory activity such as increased IL-10.88,89 However, this strategy has been directed at exploring the effect of targeted exposure of single agents, rather than an assessment of total micro-biome composition dysbiosis in its entirety. There are additional models of coeliac disease emerging in the literature;87 for example transfer of gliadin presensitised CD4+CD25−CD45RBlow T cells into a Rag-deficient murine host,90 and these novel models may be able to shed some new light on the role of the microbiota in coeliac pathogenesis. To date, the vast majority of studies assessing the microbiota in coeliac disease has used human faecal and/or biopsy tissue specimens, as discussed below, rather than employ animal models. The main findings from the assessment of the microbiota in coeliac disease are presented in table 2.

Table 2.

The main outcomes from studies assessing the microbiome in coeliac disease (upper small bowl mucosal biopsies and faecal stream analysis)

Association with coeliac disease compared with control Reference
Dysbiosis and ↑ diversity in adults and children 76, 8184
No unifying pattern or distinct composition or diversity
↑ Firmicutes in adults 76, 83
↑ Bacteroidetes in children 80, 81, 83, 84
Bifidobacterium in children 82, 86
No difference in diversity by microarray HITChip/16S sequencing 77, 78
High genetic risk—↑ Firmicutes 91
High genetic risk—↓ Bacteroidetes 92
High genetic risk—altered diversity 91, 92
No single causative agent identified
Altered oral microbiome 96

Olivares et al91 provided evidence that underlying genetic status can influence composition of the developing microbiota. Faecal stream pyrosequencing analysis from infants deemed high or low genetic risk of coeliac disease (HLA-DQ2 carriers or non-HLA-DQ2/8, respectively) was assessed for intestinal microbial composition. Interestingly, HLA status was associated with differential faecal bacterial composition, with those deemed high risk carrying increased Firmicutes and Proteobacteria, and reduced Actinobacteria, suggesting genetic status may impact the composition of the evolving intestinal microbiota. Whether this association leads to emergence of disease remains unclear and is under investigation. Similarly, Sellitto et al92 performed dynamic stool sequencing analysis from birth to age 2 years, in infants genetically at high risk of coeliac disease. They reported the temporal evolution of the intestinal microbiota in this cohort, and asked whether this changed in accordance with timing of dietary gluten exposure. As expected, faecal bacterial composition changed over time. However, regardless of timing to gluten exposure, the microbiota did not reach that expected of a healthy adult by 24 months. In particular, this high-risk group carried much less Bacteroidetes. Close monitoring of these children for a longer term may give clues as to whether this dysbiosis in infancy and early childhood impacts disease emergence.

Nistal et al76 report a change in bacterial richness between adults and children with coeliac disease, and provide evidence of a dysbiosis between treated and untreated adults, especially when considering unknown bacterial composites. Similarly, Schippa et al83 assessed the duodenal microbiota in children before and after introduction of a gluten-free diet in the same individuals and identified around 65% similarity, with increased diversity in the active state compared with after treatment. It has been suggested that these observations may indicate that the duodenal microbiome can be modulated by exposure to dietary gluten. An alternative explanation is that it may be modulated by differences in mucosal inflammatory activity with withdrawal of the dietary stimulant.

Coeliac disease can present as a variety of symptoms, including classical GI or extraintestinal symptoms, such as the characteristic skin lesion, dermatitis herpetiformis. The factors that dictate how an individual will manifest their disease clinically are unknown. Could the microbiota be involved in this process? Wacklin et al93 assessed this and found that patients presenting with GI symptoms or anaemia clustered separately on principle coordinate analysis than those with skin presentation, had a reduced duodenal mucosal bacterial diversity and differential bacterial population characterised by an increase in Proteobacteria, and a reduction in Bacteroidetes and Firmicutes.

It has recently been shown that bacteria with enzymatic ability to degrade gluten-derived peptides are present in the oral cavity of healthy individuals.94 It is unknown whether there is an altered abundance or functional ability of these bacteria in those with coeliac disease. Treatment strategy currently rests on adherence to a gluten-free diet, but this can be difficult to rigorously achieve, and exclusivity is a challenge. Therefore alternative and adjunct therapies are under development. One of these adjunct strategies uses oral recombinant glutenase and has reported a successful outcome in a Phase II trial.95 It may be that lessons from endogenous oral bacteria can assist this effort. There has been some interest in whether the oral microbiome differs in those with coeliac disease. Francavilla et al96 showed that children treated for coeliac disease do have an altered oral microbiome, characterised by reduction in diversity and a change in abundance of various bacterial species. Specifically, there was an increase in Bacteroidetes and a reduction in Actinobacteria with representative changes in the oral metabo-lome. The authors suggest that this parameter could in turn be developed as a non-invasive screening tool for coeliac disease in the future.

RHEUMATOID ARTHRITIS

RA is a chronic, systemic, polyarthritic disease characterised by synovial inflammation and erosion of bone and cartilage, progressing to functional disability.97,98 Longitudinal studies indicate that autoimmune aspects of RA are initiated years before clinical manifestations of the disease are evident,99 with circulating anticyclic citrullinated peptide antibodies and rheumatoid factor (RF) evident up to a decade prior to emergence of clinical disease.100 ACPAs are specific biomarkers for RA, present in 70–80% of patients with RA, and are typically associated with worse outcomes.97 RA affects up to 1% of adults worldwide97,98 and is multifactorial in aetiology, requiring interaction between genetic and environmental factors for its onset.9799

As with T1D, genetic factors are important97,101 but account for only a proportion of risk susceptibility for RA, and genetic predisposition does not guarantee the development of RA.99 Although twin studies show a higher concordance in monozygotic twins (12–15%) than in dizygotic twins (3.5%), the overall concordance is low and indicative of a pivotal role for environmental influences.102,103

There is ongoing debate on whether RA may be initiated by an infectious microorganism,98,104 and many bacteria have been proposed in this capacity, such as Mycoplasma fermentans,105 Escherichia coli106 and Proteus mirabilis.107,108 This idea of ‘molecular mimicry’ has existed for at least a century, but has never been definitively proven.104 As part of this assessment, the oral microbiota has been explored in RA pathogenesis. Belief in the so-called ‘oral sepsis hypothesis’ resulted in tooth extraction as a common treatment for RA—a practice that dates back to the early 1900s,109 which continued for several decades. Current literature continues to support associations between RA and the microbiota. The main findings from this assessment are presented in table 3. The periodontal microbiota has been a particular focus. Animal models indicate that the periodontal pathogens Porphyromonas gingivalis and Porphyromonas nigres-cans significantly aggravate the severity of collagen-induced arthritis (CIA), with bacterially induced IL-17 directly correlated with intensity of arthritic bone erosion.110 Moreover, in humans, patients with new-onset RA have a higher prevalence of severe periodontitis at RA disease onset despite their young age and paucity of smoking history and normal oral hygiene routine.111 Patients with RA have more tooth loss and greater periodontal friability despite oral hygiene comparable to that in healthy controls112 and the severity of periodontal disease is correlated with RA disease activity.113 After controlling for a variety of confounding factors, including RA status, age, gender, education,114 smoking,111,115 alcohol consumption and body mass index (BMI), only RA status and age predict periodontal disease.114 In addition, patients with RA who receive treatment for periodontal disease show improvements in RA with concomitant decreases in APCAs, anti-P. gingivalis antibodies,116 and proinflammatory cytokines such as TNF-α.117

Table 3.

The main outcomes from studies assessing the microbiome in RA

Microbiome site Association with RA compared with control Reference
Oral Porphyromonas Gingivalis and Porphyromonas nigrescans aggravate animal models of arthritis 110, 121
↑ Prevalence of periodontitis in patients with RA 111115
Evidence of periodontal pathogens in synovial fluid of patients with RA 111, 119, 120
Intestinal Animal models of arthritis exacerbated or rescued by changes in gut microbiome 131, 132, 134
Humans—↓ Bacteroidetes and Bifidobacterium 135, 136

RA, Rheumatoid arthritis.

The presence of antibodies to P. gingivalis is associated with the presence of RA-related autoantibodies in patients with RA,118 as well as individuals at risk for, but who have not yet developed, RA.115 Levels of antibodies to P. gingivalis correlate with levels of APCAs and RF, which are indicative of RA disease activity.115 The question is whether this association between the oral microbiota and RA directly impacts pathogenesis. DNA of P. gingivalis119 and P. nigrescens120 are found in serum and synovial fluid of patients with RA. Similarly, P. gingivalis111 and P. nigrescens119,120 are present in subgingival dental plaque and synovial fluid of patients with RA. Thus, it has been speculated that a particular species of Porphyromonas, perhaps working in concert with oral bacteria from other genera (including Anaeroglobus, Prevotella and Leptotrichia) may potentially serve as an environmental trigger for RA in genetically susceptible individuals.111 However, it remains to be definitely determined whether local periodontal disease precedes the development of RA, or whether periodontitis could be an extra-articular feature of RA, in which case periodontal tissue and joints are preferential targets of the same auto-immune processes.111 To explore this relationship, Marchesan et al121 infected a CIA mouse model with P. gingivalis, and reported increased severity of joint disease, associated with systemic proinflammatory cytokine profiles representative of activation of the Th17 pathway.

P. gingivalis is the only known prokaryote carrying a gene capable of expressing the endogenous peptidylarginine deiminase enzyme, required for the conversion of arginine residues to citrulline. Thus, P. gingivalis could be involved in the pathogenesis of autoimmunity by facilitating the generation of citrullinated proteins that can foster loss of immune tolerance and production of APCAs. It has been hypothesised that individuals who possess a genetic predisposition (or other susceptibility factors) together with P. gingivalis within their oral microbiota are more likely to develop immune responses to citrullinated antigens. As an example, patients with RA can be positive for antibodies to citrullinated α-enolase peptide-1 (CEP-1) that cross react with bacterial enolase and there is a correlation between the presence of APCA and CEP-1, perhaps due to a shared epitope.122

Recently, there has been interest in the role of the respiratory tract microbiota in RA. It is suggested that, by virtue of their constant exposure to bacterial antigens, the lungs may be a potential site of early events that facilitate the initiation and, or progression of RA. While there have not been any studies that directly examined the role of the lungs and their microbiota in patients with RA, several studies suggest that the lungs may be susceptible to proinflammatory microbiota originating from periodontal tissue.123 First off, the respiratory mucosa houses their own unique set of microbiota that can be come perturbed in disease states.124,125 In addition, the lungs are a site of local citrullination, which can be accelerated by smoking in the absence of RA.126 Moreover, patients with early RA and at-risk, seropositive individuals without RA, show signs of inflammatory associated airway injury, such as bronchial wall thickening, and air trapping.127

Could the intestinal microbiota play a pivotal role in the pathogenesis of RA?128130 Animal models of RA can be rescued or exacerbated by elimination or exposure to gut-residing bacteria, respectively.131,132 For example, the K/BxN T cell receptor transgenic mouse model of spontaneous inflammatory arthritis is attenuated by germ-free rearing or modulation of gut micro-biota with antibiotic treatment. In contrast, segmented filamentous bacteria related to Clostridium can induce proinflammatory small bowel lamina propria responses in this mouse model of arthritis, via an increase in Th17 cells and subsequent exacerbation in arthritic pathology.132 Another spontaneous murine model of inflammatory arthritis, namely the IL-1 receptor antagonist-knockout mouse (IL-1RA−/−), also showed no development of arthritis in germ-free conditions.131 Crucially, exposure of these germ-free mice to Lactobacillus bifidus, a Gram-positive anaerobic commensal of the GI tract, exacerbated disease. Elegant use of further TLR genetic knockout in this model revealed that TLR signalling is intimately linked to arthritis pathogenesis; IL-1RA−/−TLR2−/− mice displayed exacerbated arthritis through reduction in regulatory T cell response. In comparison, IL-1RA−/−TLR4−/− mice were protected from arthritis through reduction in Th17 T cell response. These results suggest that innate receptor sensing, potentially of gut microbiota, may be a crucial step in disease pathogenesis and provides insight into a gut to joint mechanism in disease pathogenesis.

Mice carrying arthritis susceptibility genes (HLA DRB*0401) have a different composition of gut microbiota compared with genetically resistant counterparts (HLA DRB*0402), rich in Clostridium-like bacteria. This was associated with differential Th17 gene transcripts in the gut, altered mucosal immune function and increased gut permeability.133 Dorożyńska et al134 showed that modulation of the gut flora with antibiotic treatment reduced disease severity in the CIA animal model of RA, along with differential cytokine response in mesenteric lymph nodes.

How does this translate to human disease? Vaahtovuo et al135 identified a dysbiosis of faecal microbiota in patients with newly diagnosed RA compared with fibromyalgic controls, characterised by a decrease in Bifidobacteria and Bacteroidetes. Similarly, faecal 16sRNA sequencing has shown that patients with new onset RA carry a distinctive enterotype of gut micro-biota characterised by an abundance of Prevotella copri and a relative lack of Bacteroides.136 P. copri robustly correlates with disease severity in patients with new-onset RA although whether this impacts the initiation or progression of autoimmunity is unclear. However, this species of bacteria is capable of expanding to dominate the commensal microbiota and exacerbates experimental colitis when delivered to mice by gavage.136

IS THERE ANY EVIDENCE FOR THE ROLE OF THE MICROBIOME IN OTHER AUTOIMMUNE DISEASES?

Autoimmune diseases can occur at any site in the body, and indeed there is a long list of diagnoses in this category. Again these occur due to a prescribed genetic susceptibility and largely unknown environmental influences, and manifest serologically with evidence of autoantibody production. Examples include autoimmune thyroiditis, autoimmune pancreatitis, Sjogren’s syndrome, systemic lupus erythematosus (SLE) and autoimmune liver diseases such as autoimmune hepatitis and primary biliary cirrhosis. Given the evidence for the role of the microbiota in the pathogenesis of T1D, RA and coeliac disease as discussed previously, there is an emerging interest in whether the micro-biota may be implicated in other, often rarer autoimmune conditions. This is certainly in its infancy, but there are a few publications appearing in the literature to this effect.

Zhou et al137 recently analysed the faecal microbiota from patients with hyperthyroidism compared with healthy controls. Denaturing gradient gel electrophoresis analysis revealed an increased bacterial diversity in those with hyperthyroidism, with a reduction in Lactobacillus and Bifidobacteria. To date, this appears to be the only study of this nature in thyroid disease. However, a clue that the intestinal microbiota may be an important environmental factor appeared over a decade ago in the literature; disease susceptibility of a rat model of auto-immune thyroiditis could be affected through modulation of the gut microbiota.138 Animals raised in specific pathogen-free conditions were less susceptible to disease. In contrast, treatment with oral antibiotics and stool transplantation from conventionally reared animals into specific pathogen-free rats, resulted in exacerbated disease. Furthermore, this effect was seen in off-spring when this modulation was given to mothers during gestation.

Sjogren’s syndrome and SLE are characterised by the emergence of anti-Ro66/Sjögren’s syndrome antigen A antibodies, but the initiating event leading to this is unclear. As loss of tolerance by T cells is known to be necessary in this process, Szymula et al139 explored whether these T cells could be activated through recognition of gut derived bacterial antigens. They created Ro60 reactive T cell hybidomas from mice transgenic for the genetic susceptibility for Sjogren’s syndrome and SLE, and tested their ability to react to different bacteria-derived peptides. They found reactivity to three peptides derived from oral commensal bacteria, and also four peptides from gut-derived commensal bacteria; three of the latter belonged to Bacteroides spp. This suggests that autoreactive T cells responsible for these autoimmune disorders may be primed in the gut by exposure to commensal microbiota. However, as yet, there are no reports of intestinal microbiota analysis in SLE and Sjogren’s syndrome. Indeed, from animal studies using the NZB mouse model that spontaneously develops autoimmune features likened to human SLE, the potential role of the microbiota is less clear, in that there is little difference in disease emergence and autoantibody formation between germ-free and conventionally raised litters.140 However, disease characteristics can be modified by dietary change. The mechanism of this is unclear, but it has been hypothesised that this may reflect modulation of the gut commensal bacteria.140

Ankylosing spondylitis (AS) is associated with a clear genetic susceptibility of HLA-B27 positivity and is characterised clinically by spinal and large joint arthropathy, enthesopathy and other systemic manifestations. There is a strong association between AS and microscopic or overt IBD.141,142 Serological evidence of anti-cBir antibodies in patients with AS have implicated flagellated bacteria in this disease.143 Animal models of AS with HLA-B27 genotype do not develop disease in germ-free facilities.144 These findings have fuelled interest in whether the gut microbiota could be involved in this disease pathogenesis. A small human study in 2002 using denaturing gradient gel electrophoresis techniques assessed this question and did not find any clear dysbiosis.145 The same group went on to show that circulating T cells from patients with AS evoked a diminished IL-10 cytokine response after exposure to autologous faecal Bacteriodes sp.146 Recently, Lin et al147 have shown that carriage of the human transgene HLA-B27 in rats itself alters the caecal microbiota, although the mechanism by which this then crosstalks to the host immune system and impacts phenotype remains unclear. To date, there are no additional reports of human studies assessing the gut microbiota in AS compared with controls that employ high sensitivity sequencing techniques.

MS is a chronic demyelinating disorder of the central nervous system (CNS), mediated by a predominant T cell driven myelin directed autoimmunity. The initiating factor is unknown, although genetic and environmental factors play an important role. The concept of the microbiota-gut-brain axis has emerged given that the enteric microbial community has the ability to crosstalk with our nervous system,148 for example, gut microbes can secrete various molecules that can directly impact enteric neuronal signalling, such as serotonin, melatonin or acetylcho-line, and enteric neurons express TLRs and so are able to sense and react to the microbial community directly.149 There is a growing body of evidence to show that the intestinal microbiota may be implicated in the pathogenesis of this disease (reviewed in refs.10,149,150). The quintessential animal model of MS is the experimental allergic encephalomyelitis (EAE) mouse; progressive demyelinating neurological disease is precipitated by pathogenic autoreactive T cells induced by simultaneous injection of a myelin antigen and bacterial adjuvants. In this model, manipulation of the gut microbiota by germ-free rearing151 or antibiotics152154 confers resistance to disease onset and diminishes severity. Re-establishing intestinal colonisation in germ-free resistant mice, for example monocolonisation with segmented filamentous bacteria reinstates the disease susceptibility.151 This protection is mediated by an altered adaptive immune response, characterised by an increase in regulatory T cell151,153 and IL-10 producing regulatory B cell populations,154 and reduction in proinflammatory Th1 and Th17 cells.151,152 Similarly, a spontaneous murine model of CNS demyelination (SJL/J mice expressing T cell receptor towards myelin peptide antigen) is also protected by germ-free rearing, with emergence of disease with gut microbial recolonisation,155 and disease pathogenesis implicating autoreactive pathogenic T cells and autoantibody producing B cells. Restitution of germ-free EAE mice with intact Bacteroides fragilis conferred protection, dependant on capsular polysaccharide A, whose presence attenuated disease from a therapeutic and preventative strategy, through promotion of IL-10 producing regulatory T cells via TLR2 signalling.156159 As yet, there is no reported assessment of the gut microbiota in human patients with MS and this data is eagerly awaited and may yield novel therapeutic targets.

CONCLUSION

It is clear that the human microbiota holds a pivotal position in health and disease. The enormity of this relationship is just beginning to become apparent as technology allows more in-depth analysis of the microbial community we harbour, in composition and functionality. In autoimmune disease, there is a clear strong genetic predisposition. The role of environmental influences is appreciated but not fully understood for many of these diseases and the initiating factor often remains elusive. Autoimmune disease is a complex interplay between genetics, environmental exposures and immune function, and there are several ongoing unanswered questions; what dictates the spectrum of disease severity? What dictates why some individuals with appropriate genotype remain disease-free lifelong, while others harbour latent disease or overt clinical pathology? Why do autoimmune diseases present in patients of differing ages despite the same environmental exposures? Could the micro-biota be responsible for this disparity?

There is clear and increasing evidence that changes in the microbiota are associated with some autoimmune diseases as discussed in this review. This dysbiosis in the microbiota is associated with several autoimmune diseases, involving the GI mucosa that lies in close contact with luminal contents as exemplified by coeliac disease, and also autoimmunity targeted towards distant sites, such as the pancreas in T1D and joints in RA. However, for now and for the most part, the relationship between the microbiota and autoimmune diseases remains an association. The question of ‘cause or effect?’ retains prominent status. Is dysbiosis of the microbiota an initiator of autoimmune disease, a perpetrator of increasing progression or a consequence of other pathological features? This remains unanswered. It appears that the large, global, longitudinal, prospective consortium efforts that are now in place, aim to address this point and this is certainly a Herculean task. A strength of these efforts is the detail of the design and use of cutting edge technology to maximise and thoroughly analyse the data generated. This approach has the power to revolutionise our understanding of these diseases and ultimately offer insight into novel preventative or therapeutic strategies.

Acknowledgments

Funding This research was supported (in part) by the Intramural Research Program of the NIH, National Cancer Institute. MHMcL—fellowship funded by Crohn’s and Colitis Foundation of America.

Footnotes

Correction notice The second author’s name has been corrected since published Online First.

Competing interests None.

Provenance and peer review Not commissioned; externally peer reviewed.

REFERENCES

  • 1.Turnbaugh PJ, Ley RE, Hamady M, et al. The human microbiome project: exploring the microbial part of ourselves in a changing world. Nature 2007;449:804–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Guarner F, Malagelada JR. Gut flora in health and disease. Lancet 2003;361:512–19. [DOI] [PubMed] [Google Scholar]
  • 3.Hollister EB, Gao C, Versalovic J. Compositional and functional features of the gastrointestinal microbiome and their effects on human health. Gastroenterology 2014;146:1449–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Cox AJ, West NP, Cripps AW. Obesity, inflammation, and the gut microbiota. Lancet Diabetes Endocrinol Published Online First: 21 Jul 2014. doi:10.1016/S2213-8587(14)70134-2 [DOI] [PubMed] [Google Scholar]
  • 5.Shapiro H, Thaiss CA, Levy M, et al. The cross talk between microbiota and the immune system: metabolites take center stage. Curr Opin Immunol 2014;30:54–62. [DOI] [PubMed] [Google Scholar]
  • 6.Lozupone CA, Stombaugh JI, Gordon JI, et al. Diversity, stability and resilience of the human gut microbiota. Nature 2012;489:220–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yatsunenko T, Rey FE, Manary MJ, et al. Human gut microbiome viewed across age and geography. Nature 2012;486:222–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zeissig S, Blumberg RS. Life at the beginning: perturbation of the microbiota by antibiotics in early life and its role in health and disease. Nat Immunol 2014;15:307–10. [DOI] [PubMed] [Google Scholar]
  • 9.Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012;486:207–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Joscelyn J, Kasper LH. Digesting the emerging role for the gut microbiome in central nervous system demyelination. Mult Scler 20:1553–9. [DOI] [PubMed] [Google Scholar]
  • 11.Schwabe RF, Jobin C. The microbiome and cancer. Nat Rev Cancer 2013;13:800–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Abreu MT, Peek RM Jr. Gastrointestinal malignancy and the microbiome. Gastroenterology 2014;146:1534–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bultman SJ. Emerging roles of the microbiome in cancer. Carcinogenesis 2014;35:249–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Roderburg C, Luedde T. The role of the gut microbiome in the development and progression of liver cirrhosis and hepatocellular carcinoma. Gut Microbes 2014;5:441–5. [DOI] [PubMed] [Google Scholar]
  • 15.Schnabl B, Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology 2014;146:1513–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 2014;146:1489–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Dabelea D The accelerating epidemic of childhood diabetes. Lancet 2009;373:1999–2000. [DOI] [PubMed] [Google Scholar]
  • 18.Vehik K, Ajami NJ, Hadley D, et al. The changing landscape of type 1 diabetes: recent developments and future frontiers. Curr Diab Rep 2013;13:642–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Hakonarson H, Grant SF. GWAS and its impact on elucidating the etiology of diabetes. Diabetes Metab Res Rev 2011;27:685–96. [DOI] [PubMed] [Google Scholar]
  • 20.Steck AK, Rewers MJ. Genetics of type 1 diabetes. Clin Chem 2011;57:176–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bakay M, Pandey R, Hakonarson H. Genes involved in type 1 diabetes. Genes (Basel) 2013;4:499–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Alper CA, Husain Z, Larsen CE, et al. Incomplete penetrance of susceptibility genes for MHC-determined immunoglobulin deficiencies in monozygotic twins discordant for type 1 diabetes. J Autoimmun 2006;27:89–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Oilinki T, Otonkoski T, Ilonen J, et al. Prevalence and characteristics of diabetes among Somali children and adolescents living in Helsinki, Finland. Pediatr Diabetes 2012;13:176–80. [DOI] [PubMed] [Google Scholar]
  • 24.Nielsen DS, Krych L, Buschard K, et al. Beyond genetics. Influence of dietary factors on gut microbiota on type 1 diabetes. FEBS Lett 2014;588:4234–43. [DOI] [PubMed] [Google Scholar]
  • 25.Kondrashova A, Hyöty H. Role of viruses and other microbes in the pathogenesis of type 1 diabetes. Int Rev Immunol 2014;33:284–95. [DOI] [PubMed] [Google Scholar]
  • 26.Boerner BP, Sarvetnick NE. Type 1 diabetes: role of intestinal microbiome in humans and mice. Ann N Y Acad Sci 2011;1243:103–18. [DOI] [PubMed] [Google Scholar]
  • 27.Atkinson MA, Chervonsky A. Does the gut microbiota have a role in type 1 diabetes? Early evidence from humans and animal models of the disease. Diabetologia 2012;55:2868–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hara N, Alkanani AK, Ir D, et al. The role of the intestinal microbiota in type 1 diabetes. Clin Immunol 2013;146:112–19. [DOI] [PubMed] [Google Scholar]
  • 29.Zipris D The interplay between the gut microbiota and the immune system in the mechanism of type 1 diabetes. Curr Opin Endocrinol Diabetes Obes 2013;20:265–70. [DOI] [PubMed] [Google Scholar]
  • 30.Dunne JL, Triplett EW, Geyers D, et al. The intestinal microbiome in type 1 diabetes. Clin Exp Immunol 2014;177:30–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Brehm MA, Powers AC, Shultz LD, et al. Advancing animal models of human type 1 diabetes by engraftment of functional human tissues in immunodeficient mice. Cold Spring Harb Perspect Med 2012;2:a007757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wen L, Ley RE, Volchkov PY, et al. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature 2008;455:1109–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hansen CH, Krych L, Nielsen DS, et al. Early life treatment with vancomycin propagates Akkermansia muciniphila and reduces diabetes incidence in the NOD mouse. Diabetologia 2012;55:2285–94. [DOI] [PubMed] [Google Scholar]
  • 34.Alkanani AK, Hara N, Lien E, et al. Induction of diabetes in the RIP-B7.1 mouse model is critically dependent on TLR3 and MyD88 pathways and is associated with alterations in the intestinal microbiome. Diabetes 2014;63:619–31. [DOI] [PubMed] [Google Scholar]
  • 35.Tormo-Badia N, Hakansson A, Vasudevan K, et al. Antibiotic treatment of pregnant non-obese diabetic (NOD) mice leads to altered gut microbiota and intestinal immunological changes in the offspring. Scand J Immunol 2014;80:250–60. [DOI] [PubMed] [Google Scholar]
  • 36.Sofi MH, Gudi R, Karumuthil-Melethil S, et al. pH of drinking water influences the composition of gut microbiome and type 1 diabetes incidence. Diabetes 2014;63:632–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wolf KJ, Daft JG, Tanner SM, et al. Consumption of acidic water alters the gut microbiome and decreases the risk of diabetes in NOD mice. J Histochem Cytochem 2014;62:237–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kriegel MA, Sefik E, Hill JA, et al. Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice. Proc Natl Acad Sci USA 2011;108:11548–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Markle JG, Frank DN, Adeli K, et al. Microbiome manipulation modifies sex-specific risk for autoimmunity. Gut Microbes 2014;5:485–93. [DOI] [PubMed] [Google Scholar]
  • 40.Peng J, Narasimhan S, Marchesi JR, et al. Long term effect of gut microbiota transfer on diabetes development. Journal of Autoimmunity 2014;53:85–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.King C, Sarvetnick N. The incidence of type-1 diabetes in NOD mice is modulated by restricted flora not germ-free conditions. PLoS ONE 2011;6:e17049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zipris D Toll-like receptors and type 1 diabetes. Adv Exp Med Biol 2010;654:585–610. [DOI] [PubMed] [Google Scholar]
  • 43.Brugman S, Klatter FA, Visser JT, et al. Antibiotic treatment partially protects against type 1 diabetes in the Bio-Breeding diabetes prone rat. Is the gut flora involved in the development of type 1 diabetes? Diabetologia 2006;49:2105–8. [DOI] [PubMed] [Google Scholar]
  • 44.Schwartz RF, Neu J, Schatz D, et al. Comment on: Brugman S et al. (2006) Antibiotic treatment partially protects against type 1 diabetes in the Bio-Breeding diabetes prone rat. Is the gut flora involved in the development of type 1 diabetes? Diabetologia 49:2105–2108. Diabetologia 2007;50:220–1. [DOI] [PubMed] [Google Scholar]
  • 45.Vaarala O Human intestinal microbiota and type 1 diabetes. Curr Diab Rep 2013;13:601–7. [DOI] [PubMed] [Google Scholar]
  • 46.de Goffau MC, Fuentes S, van den Bogert B, et al. Aberrant gut microbiota composition at the onset of type 1 diabetes in young children. Diabetologia 2014;57:1569–77. [DOI] [PubMed] [Google Scholar]
  • 47.Brown CT, Davis-Richardson AG, Giongo A, et al. Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS ONE 2011;610:e25792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Giongo A, Gano KA, Crabb DB, et al. Toward defining the autoimmune microbiome for type 1 diabetes. ISME J 2011;5:82–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Murri M, Leiva I, Gomez-Zumaquero JM, et al. Gut microbiota in children with type 1 diabetes differs from that in healthy children: a case-control study. BMC Med 2013;11:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Endesfelder D, zu Castell W, Ardissone A, et al. Compromised gut microbiota networks in children with anti-islet cell autoimmunity. Diabetes 2014;63:2006–14. [DOI] [PubMed] [Google Scholar]
  • 51.Mejía-León ME, Petrosino JF, Ajami NJ, et al. Fecal microbiota imbalance in Mexican children with type 1 diabetes. Sci Rep 2014;4:3814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Cardwell CR, Stene LC, Joner G, et al. Caesarean section is associated with an increased risk of childhood onset type 1 diabetes: a meta-analysis of observational studies. Diabetologia 2008;51:726–35. [DOI] [PubMed] [Google Scholar]
  • 53.Phillips J, Gill N, Sikdar K, et al. History of cesarean section associated with childhood onset of T1DM in Newfoundland and Labrador, Canada. J Environ Public Health 2012;2012:635097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Dominguez-Bello MG, Costello EK, Contreras M, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci USA 2010;107:11971–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wallberg M, Cooke A. Immune mechanisms in type 1 diabetes. Trends Immunol 2013;34:583–91. [DOI] [PubMed] [Google Scholar]
  • 56.Lau K, Benitez P, Ardissone A, et al. Inhibition of type 1 diabetes correlated to a Lactobacillus johnsonii N6.2-mediated Th17 bias. J Immunol 2011;186:3538–46. [DOI] [PubMed] [Google Scholar]
  • 57.Bedoya SK, Lam B, Lau K, et al. Th17 cells in immunity and autoimmunity. Clin Dev Immunol 2013;2013:986789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bosi E, Molteni L, Radaelli MG, et al. Increased intestinal permeability precedes clinical onset of type 1 diabetes. Diabetologia 2006;49:2824–7. [DOI] [PubMed] [Google Scholar]
  • 59.Secondulfo M, Iafusco D, Carratù R, et al. Ultrastructural mucosal alterations and increased intestinal permeability in non-celiac, type 1 diabetic patients. Dig Liver Dis 2004;36:35–45. [DOI] [PubMed] [Google Scholar]
  • 60.Vaarala O Leaking gut in type 1 diabetes. Curr Opin Gastroenterol 2008;24:701–6. [DOI] [PubMed] [Google Scholar]
  • 61.Visser J, Rozing J, Sapone A, et al. Tight junctions, intestinal permeability, and autoimmunity: celiac disease and type 1 diabetes paradigms. Ann N Y Acad Sci 2009;1165:195–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kupfer SS, Jabri B. Pathophysiology of celiac disease. Gastrointest Endosc Clin N Am 2012;22:639–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Guandalini S, Assiri A. Celiac disease: a review. JAMA Pediatr 2014;168:272–8. [DOI] [PubMed] [Google Scholar]
  • 64.Meresse B, Malamut G, Cerf-Bensussan N. Celiac disease: an immunological jigsaw. Immunity 2012;36:907–19. [DOI] [PubMed] [Google Scholar]
  • 65.Abadie V, Jabri B. IL-15: a central regulator of celiac disease immunopathology. Immunol Rev 2014;260:221–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mooney PD, Hadjivassiliou M, Sanders DS. Coeliac disease. BMJ 2014;348:g156. [DOI] [PubMed] [Google Scholar]
  • 67.Greco L, Romino R, Coto I, et al. The first large population based twin study of coeliac disease. Gut 2002;50:624–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Nisticò L, Fagnani C, Coto I, et al. Concordance, disease progression, and heritability of coeliac disease in Italian twins. Gut 2006;55:803–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Dubois PC, Trynka G, Franke L, et al. Multiple common variants for celiac disease influencing immune gene expression. Nat Genet 2010;42:295–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Trynka G, Hunt KA, Bockett NA, et al. Dense genotyping identifies and localizes multiple common and rare variant association signals in celiac disease. Nat Genet 2011;43:1193–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Wijmenga C, Gutierrez-Achury J. Celiac disease genetics: past, present and future challenges. J Pediatr Gastroenterol Nutr 2014;59(Suppl 1):S4–7. [DOI] [PubMed] [Google Scholar]
  • 72.Lohi S, Mustalahti K, Kaukinen K, et al. Increasing prevalence of coeliac disease over time. Aliment Pharmacol Ther 2007;26:1217–25. [DOI] [PubMed] [Google Scholar]
  • 73.Rubio-Tapia A, Kyle RA, Kaplan EL, et al. Increased prevalence and mortality in undiagnosed celiac disease. Gastroenterology 2009;137:88–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.de Sousa Moraes LF, Grzeskowiak LM, de Sales Teixeira TF, et al. Intestinal microbiota and probiotics in celiac disease. Clin Microbiol Rev 2014;27:482–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ou G, Hedberg M, Hörstedt P, et al. Proximal small intestinal microbiota and identification of rod-shaped bacteria associated with childhood celiac disease. Am J Gastroenterol 2009;104:3058–67. [DOI] [PubMed] [Google Scholar]
  • 76.Nistal E, Caminero A, Herran AR, et al. Differences of small intestinal bacteria populations in adults and children with/without celiac disease: effect of age, gluten diet, and disease. Inflamm Bowel Dis 2012;18:649–56. [DOI] [PubMed] [Google Scholar]
  • 77.Cheng J, Kalliomäki M, Heilig HG, et al. Duodenal microbiota composition and mucosal homeostasis in pediatric celiac disease. BMC Gastroenterol 2013;13:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.de Meij TG, Budding AE, Grasman ME, et al. Composition and diversity of the duodenal mucosa-associated microbiome in children with untreated coeliac disease. Scand J Gastroenterol 2013;48:530–6. [DOI] [PubMed] [Google Scholar]
  • 79.Collado MC, Calabuig M, Sanz Y. Differences between the fecal microbiota of coeliac infants and healthy controls. Curr Issues Intest Microbiol 2007;8:9–14. [PubMed] [Google Scholar]
  • 80.Nadal I, Donat E, Ribes-Koninckx C, et al. Imbalance in the composition of the duodenal microbiota of children with coeliac disease. J Med Microbiol 2007;56:1669–74. [DOI] [PubMed] [Google Scholar]
  • 81.Sanz Y, Sanchez E, Marzotto M, et al. Differences in faecal bacterial communities in coeliac and healthy children as detected by PCR and denaturing gradient gel electrophoresis. FEMS Immunol Med Microbiol 2007;51:562–8. [DOI] [PubMed] [Google Scholar]
  • 82.Collado MC, Donat E, Ribes-Koninckx C, et al. Specific duodenal and faecal bacterial groups associated with paediatric coeliac disease. J Clin Pathol 2009;62:264–9. [DOI] [PubMed] [Google Scholar]
  • 83.Schippa S, Iebba V, Barbato M, et al. A distinctive ‘microbial signature’ in celiac pediatric patients. BMC Microbiol 2010;10:175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Di Cagno R, De Angelis M, De Pasquale I, et al. Duodenal and faecal microbiota of celiac children: molecular, phenotype and metabolome characterization. BMC Microbiol 2011;11:219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sanchez E, Laparra JM, Sanz Y. Discerning the role of Bacteroides fragilis in celiac disease pathogenesis. Appl Environ Microbiol 2012;78:6507–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.De Palma G, Kamanova J, Cinova J, et al. Modulation of phenotypic and functional maturation of dendritic cells by intestinal bacteria and gliadin: relevance for celiac disease. J Leukoc Biol 2012;92:1043–54. [DOI] [PubMed] [Google Scholar]
  • 87.Stoven S, Murray JA, Marietta EV. Latest in vitro and in vivo models of celiac disease. Expert Opin Drug Discov 2013;8:445–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Laparra JM, Olivares M, Gallina O, et al. Bifidobacterium longum CECT 7347 modulates immune responses in a gliadin-induced enteropathy animal model. PLoS ONE 2012;7:e30744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Olivares M, Laparra M, Sanz Y. Oral administration of Bifidobacterium longum CECT 7347 modulates jejuna proteome in an in vivo gliadin induced enteropathy animal model. J Proteomics 2012;77:310–20. [DOI] [PubMed] [Google Scholar]
  • 90.Freitag TL, Rietdijk S, Junker Y, et al. Gliadin-primed CD4 +CD45RBlowCD25-T cells drive gluten-dependent small intestinal damage after adoptive transfer into lymphopenic mice. Gut 2009;58:1597–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Olivares M, Neef A, Castillejo G, et al. The HLA-DQ2 genotype selects for early intestinal microbiota composition in infants at high risk of developing coeliac disease. Gut Published Online First: 17 Jun 2014. doi:10.1136/gutjnl-2014-306931. [DOI] [PubMed] [Google Scholar]
  • 92.Sellitto M, Bai G, Serena G, et al. Proof of concept of microbiome-metabolome analysis and delayed gluten exposure on celiac disease autoimmunity in genetically at-risk infants. PLoS ONE 2012;7:e33387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wacklin P, Kaukinen K, Tuovinen E, et al. The duodenal microbiota composition of adult celiac disease patients is associated with the clinical manifestation of the disease. Inflamm Bowel Dis 2013;19:934–41. [DOI] [PubMed] [Google Scholar]
  • 94.Fernandez-Feo M, Wei G, Blumenkranz G, et al. The cultivable human oral gluten-degrading microbiome and its potential implications in coeliac disease and gluten sensitivity. Clin Microbiol Infect 2013;19:E386–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lähdeaho ML, Kaukinen K, Laurila K, et al. Glutenase ALV003 attenuates gluten-induced mucosal injury in patients with celiac disease. Gastroenterology 2014;146:1649–58. [DOI] [PubMed] [Google Scholar]
  • 96.Francavilla R, Ercolini D, Piccolo M, et al. Salivary microbiota and metabolome associated with celiac disease. Appl Environ Microbiol 2014;80:3416–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med 2011;365:2205–19. [DOI] [PubMed] [Google Scholar]
  • 98.Scher JU, Abramson SB. The microbiome and rheumatoid arthritis. Nat Rev Rheumatol 2011;7:569–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Luckey D, Gomez A, Murray J, et al. Bugs & us: the role of the gut in autoimmunity. Indian J Med Res 2013;138:732–43. [PMC free article] [PubMed] [Google Scholar]
  • 100.Nielen MM, van Schaardenburg D, Reesink HW, et al. Increased levels of C-reactive protein in serum from blood donors before the onset of rheumatoid arthritis. Arthritis Rheum 2004;50:2423–7. [DOI] [PubMed] [Google Scholar]
  • 101.Kochi Y, Suzuki A, Yamamoto K. Genetic basis of rheumatoid arthritis: A current review. Biochem Biophys Res Commun 2014;452:254–62. [DOI] [PubMed] [Google Scholar]
  • 102.MacGregor AJ, Snieder H, Rigby AS, et al. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum 2000;43:30–7. [DOI] [PubMed] [Google Scholar]
  • 103.Bogdanos DP, Smyk DS, Rigopoulou EI, et al. Twin studies in autoimmune disease: genetics, gender and environment. J Autoimmun 2012;38:J156–69. [DOI] [PubMed] [Google Scholar]
  • 104.Paget SA. The microbiome, autoimmunity, and arthritis: cause and effect: an historical perspective. Trans Am Clin Climatol Assoc 2012;123:257–66. [PMC free article] [PubMed] [Google Scholar]
  • 105.Sato N, Oizumi T, Kinbara M, et al. Promotion of arthritis and allergy in mice by aminoglycoglycerophospholipid, a membrane antigen specific to Mycoplasma fermentans. FEMS Immunol Med Microbiol 2010;59:33–41. [DOI] [PubMed] [Google Scholar]
  • 106.Newkirk MM, Zbar A, Baron M, et al. Distinct bacterial colonization patterns of Escherichia coli subtypes associate with rheumatoid factor status in early inflammatory arthritis. Rheumatology (Oxford) 2010;49:1311–16. [DOI] [PubMed] [Google Scholar]
  • 107.Ebringer A, Ptaszynska T, Corbett M, et al. Antibodies to proteus in rheumatoid arthritis. Lancet 1985;2:305–7. [DOI] [PubMed] [Google Scholar]
  • 108.Ebringer A, Rashid T, Wilson C. Rheumatoid arthritis, Proteus, anti-CCP antibodies and Karl Popper. Autoimmun Rev 2010;9:216–23. [DOI] [PubMed] [Google Scholar]
  • 109.Hunter W Oral sepsis as a cause of disease. Br Med J 1900;2:215–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.de Aquino SG, Abdollahi-Roodsaz S, Koenders MI, et al. Periodontal pathogens directly promote autoimmune experimental arthritis by inducing a TLR2- and IL-1-driven Th17 response. J Immunol 2014;192:4103–11. [DOI] [PubMed] [Google Scholar]
  • 111.Scher JU, Ubeda C, Equinda M, et al. Periodontal disease and the oral microbiota in new-onset rheumatoid arthritis. Arthritis Rheum 2012;64:3083–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Wolff B, Berger T, Frese C, et al. Oral status in patients with early rheumatoid arthritis: a prospective, case-control study. Rheumatology (Oxford) 2014;53:526–31. [DOI] [PubMed] [Google Scholar]
  • 113.Al-Katma MK, Bissada NF, Bordeaux JM, et al. Control of periodontal infection reduces the severity of active rheumatoid arthritis. J Clin Rheumatol 2007;13:134–7. [DOI] [PubMed] [Google Scholar]
  • 114.Pischon N, Pischon T, Kröger J, et al. Association among rheumatoid arthritis, oral hygiene, and periodontitis. J Periodontol 2008;79:979–86. [DOI] [PubMed] [Google Scholar]
  • 115.Mikuls TR, Payne JB, Yu F, et al. Periodontitis and Porphyromonas gingivalis in patients with rheumatoid arthritis. Arthritis Rheumatol 2014;66:1090–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Okada M, Kobayashi T, Ito S, et al. Periodontal treatment decreases levels of antibodies to Porphyromonas gingivalis and citrulline in patients with rheumatoid arthritis and periodontitis. J Periodontol 2013;84:e74–84. [DOI] [PubMed] [Google Scholar]
  • 117.Ortiz P, Bissada NF, 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;80:535–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Hitchon CA, Chandad F, Ferucci ED, et al. Antibodies to Porphyromonas gingivalis are associated with anticitrullinated protein antibodies in patients with rheumatoid arthritis and their relatives. J Rheumatol 2010;37:1105–12. [DOI] [PubMed] [Google Scholar]
  • 119.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;36:1004–10. [DOI] [PubMed] [Google Scholar]
  • 120.Moen K, Brun JG, 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;24:656–63. [PubMed] [Google Scholar]
  • 121.Marchesan JT, Gerow EA, Schaff R, et al. Porphyromonas gingivalis oral infection exacerbates the development and severity of collagen-induced arthritis. Arthritis Res Ther 2013;15:R186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Wegner N, Lundberg K, Kinloch A, et al. Autoimmunity to specific citrullinated proteins gives the first clues to the etiology of rheumatoid arthritis. Immunol Rev 2010;233:34–54. [DOI] [PubMed] [Google Scholar]
  • 123.Brusca SB, Abramson SB, Scher JU. Microbiome and mucosal inflammation as extra-articular triggers for rheumatoid arthritis and autoimmunity. Curr Opin Rheumatol 2014;26:101–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Hilty M, Burke C, Pedro H, et al. Disordered microbial communities in asthmatic airways. PLoS One 2010;5:e8578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Segal LN, Alekseyenko AV, Clemente JC, et al. Enrichment of lung microbiome with supraglottic taxa is associated with increased pulmonary inflammation. Microbiome 2013;1:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Makrygiannakis D, Hermansson M, Ulfgren AK, et al. Smoking increases peptidylarginine deiminase 2 enzyme expression in human lungs and increases citrullination in BAL cells. Ann Rheum Dis 2008;67:1488–92. [DOI] [PubMed] [Google Scholar]
  • 127.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;64:1756–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Fung I, Garrett JP, Shahane A, et al. Do bugs control our fate? The influence of the microbiome on autoimmunity. Curr Allergy Asthma Rep 2012;12:511–19. [DOI] [PubMed] [Google Scholar]
  • 129.Yeoh N, Burton JP, Suppiah P, et al. The role of the microbiome in rheumatic diseases. Curr Rheumatol Rep 2013;15:314. [DOI] [PubMed] [Google Scholar]
  • 130.Taneja V Arthritis susceptibility and the gut microbiome. FEBS Lett 2014. pii: S0014–5793(14)00421–9. Published Online First. doi:10.1016/j.febslet.2014.05.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Abdollahi-Roodsaz S, Joosten LA, Koenders MI, et al. Stimulation of TLR2 and TLR4 differentially skews the balance of T cells in a mouse model of arthritis. J Clin Invest 2008;118:205–16. http://www.ncbi.nlm.nih.gov/pubmed/?term=Stimulation+of+TLR2+and+TLR4+differentially+skews+the+balance+of+T+cells+in+a+mouse+model+of+arthritis [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Wu HJ, Ivanov II, Darce J, et al. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 2010;32:815–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Gomez A, Luckey D, Yeoman CJ, et al. Loss of sex and age driven differences in the gut microbiome characterize arthritis-susceptible 0401 mice but not arthritis-resistant 0402 mice. PLoS ONE 2012;7:e36095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Dorożyńska I, Majewska-Szczepanik M, Marcińska K, et al. Partial depletion of natural gut flora by antibiotic aggravates collagen induced arthritis (CIA) in mice. Pharmacol Rep 2014;66:250–5. [DOI] [PubMed] [Google Scholar]
  • 135.Vaahtovuo J, Munukka E, Korkeamäki M, et al. Fecal microbiota in early rheumatoid arthritis. J Rheumatol 2008;35:1500–5. [PubMed] [Google Scholar]
  • 136.Scher JU, Sczesnak A, Longman RS, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife 2013;2:e01202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Zhou L, Li X, Ahmed A, et al. Gut microbe analysis between hyperthyroid and healthy individuals. Curr Microbiol 2014;69:675–80. [DOI] [PubMed] [Google Scholar]
  • 138.Penhale WJ, Young PR. The influence of the normal microbial flora on the susceptibility of rats to experimental autoimmune thyroiditis. Clin Exp Immunol 1988;72:288–92. [PMC free article] [PubMed] [Google Scholar]
  • 139.Szymula A, Rosenthal J, Szczerba BM, et al. T cell epitope mimicry between Sjögren’s syndrome Antigen A (SSA)/Ro60 and oral, gut, skin and vaginal bacteria. Clin Immunol 2014;152:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Vieira SM, Pagovich OE, Kriegel MA. Diet, microbiota and autoimmune diseases. Lupus 2014;23:518–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Costello ME, Elewaut D, Kenna TJ, et al. Microbes, the gut and ankylosing spondylitis. Arthritis Res Ther 2013;15:214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Schaeverbeke T, Truchetet ME, Richez C. Gut metagenome and spondyloarthritis. Joint Bone Spine 2013;80:349–52. [DOI] [PubMed] [Google Scholar]
  • 143.Wallis D, Asaduzzaman A, Weisman M, et al. Elevated serum anti-flagellin antibodies implicate subclinical bowel inflammation in ankylosing spondylitis: an observational study. Arthritis Res Ther 2013;15:R166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Hacquard-Bouder C, Ittah M, Breban M. Animal models of HLA-B27-associated diseases: new outcomes. Joint Bone Spine 2006;73:132–8. [DOI] [PubMed] [Google Scholar]
  • 145.Stebbings S, Munro K, Simon MA, et al. Comparison of the faecal microflora of patients with ankylosing spondylitis and controls using molecular methods of analysis. Rheumatology (Oxford) 2002;41:1395–401. [DOI] [PubMed] [Google Scholar]
  • 146.Stebbings SM, Taylor C, Tannock GW, et al. The immune response to autologous bacteroides in ankylosing spondylitis is characterized by reduced interleukin 10 production. J Rheumatol 2009;36:797–800. [DOI] [PubMed] [Google Scholar]
  • 147.Lin P, Bach M, Asquith M, et al. HLA-B27 and Human β2-Microglobulin Affect the Gut Microbiota of Transgenic Rats. PLoS ONE 2014;9:e105684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Wang Y, Kasper LH. The role of microbiome in central nervous system disorders. Brain Behav Immun 2014;38:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Bhargava P, Mowry EM. Gut microbiome and multiple sclerosis. Curr Neurol Neurosci Rep 2014;14:492. [DOI] [PubMed] [Google Scholar]
  • 150.Berer K, Krishnamoorthy G. Microbial view of central nervous system autoimmunity. FEBS Lett 2014;588:4207–13. [DOI] [PubMed] [Google Scholar]
  • 151.Lee YK, Menezes JS, Umesaki Y, et al. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA 2011;108(Suppl):4615–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Yokote H, Miyake S, Croxford JL, et al. NKT cell-dependent amelioration of a mouse model of multiple sclerosis by altering gut flora. Am J Pathol 2008;173:1714–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Ochoa-Repáraz J, Mielcarz DW, Ditrio LE, et al. Role of gut commensal microflora in the development of experimental autoimmune encephalomyelitis. J Immunol 2009;183:6041–50. [DOI] [PubMed] [Google Scholar]
  • 154.Ochoa-Repáraz J, Mielcarz DW, Haque-Begum S, et al. Induction of a regulatory B cell population in experimental allergic encephalomyelitis by alteration of the gut commensal microflora. Gut Microbes 2010;1:103–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Berer K, Mues M, Koutrolos M, et al. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 2011;479:538–41. [DOI] [PubMed] [Google Scholar]
  • 156.Ochoa-Reparaz J, Mielcarz DW, Ditrio LE. Central nervous system demyelinating disease protection by the human commensal Bacteroides fragilis depends on polysaccharide A expression. J Immunol 2010;185:4101–8. [DOI] [PubMed] [Google Scholar]
  • 157.Ochoa-Reparaz J, Mielcarz DW, Wang Y. A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal Immunol 2010;3:487–95. [DOI] [PubMed] [Google Scholar]
  • 158.Mao Y-K, Kasper DL, Wang B, et al. Bacteroides fragilis polysaccharide A is necessary and sufficient for acute activation of intestinal sensory neurons. Nat Commun 2013;4:1465. [DOI] [PubMed] [Google Scholar]
  • 159.Wang Y, Telesford KM, Ochoa-Repáraz J, et al. An intestinal commensal symbiosis factor controls neuroinflammation via TLR2-mediated CD39 signalling. Nat Commun 2014;5:4432. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES