You are on page 1of 11

ARTHRITIS & RHEUMATOLOGY

Vol. 68, No. 1, January 2016, pp 35–45


DOI 10.1002/art.39259
C 2016, American College of Rheumatology
V

REVIEW

Microbiome in Inflammatory Arthritis


and Human Rheumatic Diseases

Jose U. Scher,1 Dan R. Littman,2 and Steven B. Abramson1

Introduction asymptomatic carriers are a common feature of many


infectious diseases and that several microorganisms are
Ever since its modern conception as a medical dis-
fastidious in nature, with complex nutritional require-
cipline, the study of microorganisms has paralleled the
ments for growth. The latter fundamentally prevented
many technological advances in microbiology. In the 17th
the study of bacteria within the context of a dynamic bio-
century, the inventor of the microscope, Antony van
logic community, the role of commensal taxa, the down-
Leeuwenhoek, was also the first to describe—in the
stream molecular events, and the resulting immune
plaque of his own gums—the millions of microorganisms
interactions between microorganisms and their host.
(or “animalcules”) that reside within us. It is that multi- Consequently, only the prevalent microbiologic
tude of microbes populating most human body cavities techniques in the past were used to characterize unique
and surfaces, including its genetic and enzymatic composi- agents capable of triggering clinical rheumatic syn-
tion, that defines our microbiome (1). dromes. Over time, the search led to correlative studies
For centuries, the role of the microbiome as a of specific bacteria and viruses in the etiopathogenesis
potential determinant to health and disease has been of these disorders, most notably rheumatoid arthritis
rather ignored. This has been true in most fields of (RA), psoriasis, inflammatory bowel disease (IBD), and
human research, but particularly so in autoimmune and the related spondyloarthritides (SpA), including anky-
rheumatic conditions. The reasons are multifactorial. losing spondylitis (AS) and reactive arthritis (ReA).
Chief among those was the advent of Koch’s postulates The revolution of culture-independent, high-
in the late 1800s, which exerted a profound influence on throughput microbial DNA sequencing, in parallel with
how investigators thought about causality in medicine the resurgence of interest in and further understanding
(2). Unknown at the time, however, were the facts that of mucosal immunity, has exponentially advanced our
knowledge of the interplay between our microbes and
Supported by the NIH (National Institute of Arthritis and ourselves. That profound, bidirectional interaction and
Musculoskeletal and Skin Diseases grants K23-AR-064318 to Dr. its consequences in physiology and disease have led to a
Scher and RC2-AR-058986 through the American Recovery and
Reinvestment Act of 2009 to Drs. Littman and Abramson), the Judith whole new field of research. Despite the relative novelty
and Stewart Colton Center for Autoimmunity, and the Riley Family of the human microbiome as a discipline, a substantial
Foundation.
1
Jose U. Scher, MD, Steven B. Abramson, MD: New York
body of evidence has accumulated addressing its potential
University School of Medicine and New York University Hospital for involvement in the pathogenesis of rheumatic disease (3).
Joint Diseases, New York, New York; 2Dan R. Littman, MD, PhD: In this article, we will critically review the available data
Kimmel Center for Biology and Medicine of the Skirball Institute
and New York University School of Medicine, New York, New York.
from animal and human studies, focusing on the role of
Drs. Scher, Littman, and Abramson have a patent application the intestinal microbiome in RA, psoriatic arthritis
pending on microbiota and byproducts for diagnostics and therapeu- (PsA), and SpA. The role of the microbiome in autoim-
tics in rheumatic disease.
Address correspondence to Jose U. Scher, MD, Division of munity and in other rheumatic diseases has been reviewed
Rheumatology, New York University Hospital for Joint Diseases, elsewhere (4,5) and is beyond the scope of this article. We
301 East 17th Street, Room 1608, New York, NY 10003. E-mail: include a description of the prospects of microbiome
Jose.Scher@nyumc.org.
Submitted for publication March 10, 2015; accepted in manipulation for therapeutic purposes and conclude by
revised form June 23, 2015. delineating challenges and opportunities in the field.
35
36 SCHER ET AL

Human microbiome and mucosal immunity in physiology produce interferon-g (IFNg), both Th2 and Th17 cells
and disease (producing interleukin-4 [IL-4] and IL-17, respectively)
are stimulated by extracellular microorganisms. In con-
With the advent of massively parallel sequencing
trast, Treg cells actively prevent these proinflammatory
technologies and ever more sophisticated multi-omics
properties via generation of antiinflammatory cytokines
methodologies, the human microbiome is now better
such as IL-10. This fine homeostatic equilibrium is
understood in both its composition and its functionality. It
needed to maintain a state of basal “physiologic in-
is now recognized that more than 100 trillion cells inhabit-
flammation” in the lamina propria (11) (Figure 1).
ing our human bodies are rather prokaryotic in nature. At
With this in mind, the National Institutes of
any given time, we carry 3–6 pounds of bacteria that contain
Health Human Microbiome Project was launched (12)
roughly 3 million protein-coding genes (6). We have also
to address 2 central questions. First, is there a core
come to realize that different biologic niches are populated
human microbiome? Second, do perturbations in com-
by unique microbial consortia that can survive only under
position of the microbiome and/or its metabolites corre-
certain nutritional conditions. For example, the intestinal
late with human disease states? Concomitant with the
microbiome is entirely differentiated from that of the skin
launch of this project, the technological revolution in
or the genitourinary tract (7), and this is in part related to
DNA sequencing and multi-omics has provided the nec-
the various metabolic functions that derive from our own
essary scientific tools (13,14). Most analytic approaches
physiologic needs. In fact, our intestinal microbes (repre-
in human studies have used novel bioinformatics tools
sented by more than 1,000 different species) have coevolved
coupled with parallel DNA sequencing platforms 1) to
in a mutually beneficial manner and provide the enzymatic
describe the taxonomic relative abundance of bacterial
machinery to help us degrade complex polysaccharides
species in a given sample (taking advantage of 16S ribo-
from the diet and extract essential vitamins and amino acids
somal RNA [rRNA] gene amplification and sequencing)
required for our evolutionary success. As a result, this new
and/or 2) to better understand the overall functional
knowledge all but challenged our understanding of the self,
enzymatic potential of a microbiome of interest (using
leading to the notion that we should consider human beings
whole-genome shotgun sequencing for metagenomics
to be supraorganisms (8).
analysis). This has been complemented at times by the
This evolutionary process of coadaptation between
use of metabolomics (measuring the actual content of
intestinal microbiome and host immune responses is now
bacterial metabolites, such as short- and medium-chain
increasingly appreciated. In fact, it is now well established
fatty acids), proteomics, and metatranscriptomics (15).
that the intestinal microbiota shape the immune system and
These methods evaluate the possibility that associations
modulate homeostasis in healthy states or promote inflam-
with disease states may depend on the presence of cer-
mation when dysbiosis occurs (9).
tain bacterial components or byproducts and not neces-
In order to keep this massive antigenic load com-
sarily on the presence of the microorganism per se.
partmentalized in the intestinal lumen, mammals have
Multiple examples of microbiome correlations
developed multiple protective mechanisms (Figure 1). with metabolic, neoplastic, and autoimmune diseases have
These include a physicochemical barrier in the form of a now been reported, including those in obesity and diabe-
mucus layer, antimicrobial proteins, and secretory IgA tes mellitus (16,17), gastrointestinal cancer (18), ather-
(sIgA) that keep the microbiota away from epithelial osclerosis (19), and psoriasis (20). As expected, many of
cells. Evading bacteria encounter tightly adherent intes- the original contributions to the field in autoimmunity
tinal epithelial cells, a mechanical barrier containing were first reported in the literature on IBD. Notably, IBD
sensors of pathogen-associated molecular patterns (i.e., patients have a dysbiotic process characterized by a
Toll-like receptors [TLRs]), and a variety of antibacte- decrease in intestinal (beneficial) microbiota diversity
rial molecules. The innate immune cells, most notably (21,22) and an increase in enterobacteria (23).
macrophages and dendritic cells (DCs), continuously
sense the lumen and survey for detrimental antigens
Perspectives on the role of the intestinal
(10). Ultimately, antigens are presented by major histo-
microbiome in rheumatology
compatibility complex class II molecules and interact
with B or T cell receptors to induce adaptive immune Long before the study of the microbiome became
responses. Depending on the microbial antigen, a spe- technically possible, the suggestion had been proposed
cific cytokine milieu is then generated to influence a that intestinal microorganisms were involved in autoim-
specific type of CD41 T cell differentiation. While Th1 munity leading to clinical inflammatory arthritides. At
cells develop in response to intracellular pathogens and the dawn of the 20th century, Carl Warden suggested in
MICROBIOME IN RHEUMATIC DISEASES 37

Figure 1. Homeostasis of intestinal microbiome in physiologic states. Commensal microbiota and pathobionts are normal residents of the gut
lumen. A protective barrier in mammalians and other hosts is composed of a mucus layer, epithelial cells (producing antibacterial peptides), and
innate/adaptive immune cells residing in the lamina propria. Their fundamental role is to communicate with the microbiota to promote a state
of physiologic inflammation in order to actively tolerate a constant antigenic load that promotes nutritional, metabolic, and immune benefits.
APCs 5 antigen-presenting cells; TGFb 5 transforming growth factor b; IL-10 5 interleukin-10; TNF 5 tumor necrosis factor; sIgA 5 secretory
IgA; NK 5 natural killer; IELs 5 intraepithelial lymphocytes.

his “toxemic factor hypothesis” that overabundant gram- jejunoileal bypass surgery. This disruption of the normal
negative anaerobes in the intestinal canal produced a gastrointestinal anatomy, and consequently of its populat-
noxious substance that, once absorbed, was responsible ing microbial communities, often led to bacterial over-
for RA development (24). Modern microbiologic reports growth and antibody production that ultimately promoted
cemented the notion that gut-infecting (or colonizing) synovial inflammation (and a pathognomonic form of
bacteria could induce distal arthritis. Several of the SpA, dermatitis) (26).
most notably IBD-related arthritis, ReA, and axial SpA, As a result, multiple attempts to alter the compo-
are clear examples of this. Other arthritides rarely seen sition of intestinal bacteria have led to several therapeu-
currently are perhaps even more relevant as proof-of- tic regimens in inflammatory arthritis. A classic example
principle that intestinal dysbiosis can lead to distal synovi- is sulfasalazine. This drug, which was rationally designed
al inflammation. This is the case with Whipple’s disease, to combine an antibacterial sulfonamide with a salicylate
in which the presence of Tropheryma whipplei is neces- through an azo bond (27,28), was ultimately deemed a
sary—although not always sufficient—for the develop- disease-modifying antirheumatic drug (DMARD) and
ment of articular compromise (25). Even more intriguing approved by the Food and Drug Administration for the
is an arthritis syndrome triggered in many instances by treatment of RA, IBD, and AS. Other medications with
38 SCHER ET AL

Table 1. Murine models of RA-like and spondyloarthritis-like disease known to be modulated by the intestinal microbiome or by bacteria-
derived metabolites*
Animal Manipulation Phenotype Germ-free effect Taxa/molecule involved Ref.
Rat Adjuvant-induced arthritis RA-like synovitis Arthritis Escherichia coli, Bacteroides species 42, 43
Rat and Collagen-induced arthritis RA-like synovitis Arthritis Type II collagen, LPS 44, 45
mouse
Rat SCW-induced arthritis RA-like synovitis Arthritis Streptococcus pyogenes cell wall 46
Mouse IL-1rn–/– RA-like synovitis No arthritis Lactobacillus bifidus 47
Mouse K/BxN transgenic RA-like synovitis No arthritis Segmented filamentous bacteria 48
Rat HLA–B27/human b2-microglobulin Colitis, psoriasis, and arthritis No disease Bacteroides species 62, 63
transgenic
Mouse HLA–B27/ankylosing enthesopathy Enthesitis and ankylosis No disease Bacteroides/Enterococcus species, 68
Veillonella/Staphylococcus
species (LPS suppresses
enthesitis)
Mouse SKG (ZAP-70 single-point Arthritis, psoriasiform lesions, No disease b-glucan, gut commensal microbiota 69, 70
mutation) and colitis

* RA 5 rheumatoid arthritis; LPS 5 lipopolysaccharide; SCW 5 streptococcal cell wall; IL-1rn–/– 5 interleukin-1 receptor antagonist knockout.

antibiotic properties (hydroxychloroquine and tetracy- epidemiologic studies also suggest a correlation between
clines) have also been included in the therapeutic arma- gastrointestinal and urinary infections and decreased risk
mentarium for RA. of RA (41), indicating the possibility of disease develop-
ment in response to intestinal microbiota perturbation.
Rheumatoid arthritis There is an abundant body of literature address-
ing the role of the microbiome in inflammatory arthritis
Evidence that RA follows the epidemiology of an (Table 1). In fact, multiple animal models have estab-
infectious vector is both intriguing and debated. In fact, lished a biologic connection between the presence of
only skeletal remains of American Indians show erosive microbiota and development of synovitis. Kohashi et al
changes reminiscent of RA, a finding not seen in Euro- demonstrated that adjuvant-induced arthritis in rats
pean or Asian archaeological sites (29). Consistent with developed only under germ-free conditions (within isola-
this, an increased prevalence of RA has been reported in tors voided of all microorganisms). However, convention-
several American Indian and Alaska Native populations ally raised animals displayed only a mild phenotype,
(e.g., up to 8% in the case of Chippewa Indians). Prior suggesting a potential immunomodulatory effect of the
investigations have also implicated bacterial triggers microbiome. This same group further reported a pro-
in the pathogenesis of RA (30,31). Several studies— tective role of certain gram-negative enterobacteria
reviewed extensively elsewhere (32)—have pointed (Escherichia coli and Bacteroides species) when intro-
toward periodontitis and the oral microbiota (e.g., duced into otherwise germ-free arthritis-susceptible rats
Porphyromonas gingivalis) as inciting factors (33,34). (42,43). Collagen-induced arthritis (CIA) is also modu-
Although P gingivalis represents a relevant taxon given lated by bacteria or its components, since only germ-free
its intrinsic ability to citrullinate peptides through the animals develop disease after intradermal injection of
enzyme peptidylarginine deiminase, it is possible that native type II collagen (44). Furthermore, bacteria-
periodontal inflammation per se elicited by a variety of derived lipopolysaccharide by itself is also capable of
microorganisms (including Prevotella and Leptotrichia potentiating CIA in mice (45). Similarly, in the strepto-
species) contributes to disease development (35,36). coccal cell wall–induced arthritis model in rats, the pres-
Another possibility is that the distal airways repre- ence of gut microbiota renders animals resistant to joint
sent the actual site of citrullination, perhaps as a conse- inflammation (46).
quence of environmental insults (i.e., smoking) and/or Most recently, investigators have used novel
microbial challenges. This is largely based on work by approaches to define the exposure of an animal to the
Demoruelle et al and Willis et al, who described the pres- gut microbiota and the downstream immune response. In
ence of small airway inflammation and anti–citrullinated so-called gnotobiotic experiments, specifically character-
protein antibodies (ACPAs) in sputum from RA patients ized taxa are incorporated into germ-free (or antibiotic-
(37,38), and on data from Catrina et al and Ytterberg treated) animals and biologic events are described both
et al, who identified shared immunologic citrullinated in vivo and ex vivo. There are several noteworthy exam-
targets in joints and lungs of RA patients (39,40). Recent ples. First, IL-1 receptor antagonist–knockout (IL-1rn2/2)
MICROBIOME IN RHEUMATIC DISEASES 39

Table 2. Human rheumatic diseases associated with altered intestinal microbiome*


Human rheumatic disease Technology used Taxa/molecule implicated† Ref.
Early RA Gas–liquid chromatography Anaerobic bacteria 53
Early RA 16S rRNA hybridization and DNA staining #Bifidobacteria, #Bacteroides– 54
Porphyromonas–Prevotella
New-onset RA 16S rRNA gene amplification and whole-genome "Prevotella copri, #Bacteroides 55
shotgun sequencing
AS Denaturing gradient gel electrophoresis "Sulfate-reducing bacteria 76
AS 16S rRNA gene amplification "Lachnospiraceae and Prevotellaceae, 77
#Ruminococcaceae and Rikenellaceae
JIA (ERA) 16S rRNA gene amplification #Faecalibacterium prausnitzii 78
New-onset PsA 16S rRNA gene amplification #Akkermansia and Ruminococcus, 79
#medium-chain fatty acids,
#intestinal RANKL

* RA 5 rheumatoid arthritis; rRNA 5 ribosomal RNA; AS 5 ankylosing spondylitis; JIA 5 juvenile idiopathic arthritis; ERA 5 enthesitis-related
arthritis; PsA 5 psoriatic arthritis.
†# 5 decreased; " 5 increased.

mice develop a spontaneous autoimmune T cell–medi- Despite these advances in understanding the
ated arthritis when raised in conventional cages. While intestine–joint biologic connection, data on humans have
germ-free animals do not become arthritic, Lactobacillus been scarce (Table 2). Eerola et al initially characterized
bifidus–monocolonized IL-1rn2/2 mice develop a high intestinal flora in RA patients using gas–liquid chroma-
incidence of severe joint disease in a Th17- and TLR-4– tography and found that levels of several anaerobic bacte-
dependent manner (47). ria accounted for most differences between RA patients
Other studies have made use of gnotobiotic tools. and controls (although gas–liquid chromatography tech-
The K/BxN T cell receptor–transgenic model is caused nology cannot discriminate directionality) (53). Using
by T cell–driven production of autoantibodies against low-throughput hybridization, another study revealed
glucose-6-phosphate isomerase. Inflammatory arthritis is lower levels of the Bacteroides–Porphyromonas–Prevotella
also mitigated in germ-free animals, mostly due to a defi- bacterial group in patients with early RA compared to
ciency of Th17 cells in both the lamina propria and the controls without arthritis (54). Although these findings
periphery. Introduction of the commensal segmented fila- were of interest, the lack of polymerase chain reaction
mentous bacteria is sufficient for development of disease primer specificity rendered the results virtually impossi-
phenotype. Conversely, antibiotic use strongly hampers ble to interpret at the lower taxonomic levels.
inflammatory arthritis in K/BxN mice (48). This is highly Most recently, our group characterized the intes-
relevant since, for the first time, specific intestinal com- tinal microbiome of DMARD/steroid-naive patients
mensal microbiota demonstrated the ability to drive adap- with new-onset RA, using 16S rRNA gene amplification
tive T cell differentiation locally, followed by systemic and whole-genome shotgun sequencing. Compared to
disease at distal sites. While this remains a matter of in- patients with chronic RA, PsA patients, and healthy
tense mechanistic research, lamina propria Th17 cell pro- controls, the cohort of patients with new-onset RA had
liferation in physiologic states requires the activation of a significantly higher abundance of Prevotella copri (55)
local DCs to produce polarizing cytokines (49) and the (Figure 2). This observation is intriguing because most
ability of T cell receptors to specifically recognize com- other studies have shown that the prevalence of Prevo-
mensal intestinal bacterial antigens (50). tella is typically low in healthy individuals (with the
Taken together, these findings suggest that pertur- exception of some aboriginal populations). The reasons
bations in the composition of intestinal microbiota may be for this remain unclear, but it is possible that it relates
sufficient to trigger arthritis in a variety of experimental to dietary factors (as fiber-consuming subjects—and
murine models. However, potentially joint-protective (or children in particular—have very high intestinal levels of
joint-deleterious) properties might also depend on host Prevotella [56]), geographic variability, or a combination
genetic background and sex (51). For example, arthritis- of both. The relationship between fiber intake and RA
susceptible (HLA–DRB1*0401–transgenic) mice show incidence, however, is yet to be studied.
alterations in intestinal microbiota composition and in- Several other findings were considered of inter-
creases in gut permeability when compared to arthritis- est. First, increases in Prevotella correlated with a reduc-
resistant controls (52). tion in reportedly beneficial microbes (i.e., Bacteroides)
40 SCHER ET AL

Figure 2. Effects of dysbiosis in rheumatic and autoimmune diseases. When the homeostatic balance in the composition of the microbial com-
munity of the intestinal lumen is perturbed, a state of dysbiosis ensues and a downstream local and systemic proinflammatory response is eli-
cited. This has been largely demonstrated in multiple animal models. An accumulating body of evidence in humans suggests that a similar
perturbation can be associated with multiple rheumatic and autoimmune disorders, which could potentially explain the source of various
cytokine-induced systemic inflammatory responses. Commensal microbiota and pathobionts are normal residents of the gut lumen. A protective
barrier in mammalians and other hosts is composed of a mucus layer, epithelial cells (producing antibacterial peptides), and innate/adaptive
immune cells residing in the lamina propria. Their fundamental role is to communicate with the microbiota to promote a state of physiologic
inflammation in order to actively tolerate a constant antigenic load that promotes nutritional, metabolic, and immune benefits. # 5 decreased;
; 5 not decreased or increased; " 5 increased; MCFAs 5 medium-chain fatty acids; sIgA 5 secretory IgA; IL-17 5 interleukin-17; TNF 5 tumor
necrosis factor; ACPAs 5 anti–citrullinated protein antibodies; NKs 5 natural killer cells; IELs 5 intraepithelial lymphocytes.

in patients with new-onset RA. Interestingly, the relative increase in DHFR-high (i.e., Bacteroides-overabundant)
abundance of P copri correlated inversely with the pres- microbiota in RA patients may help explain, at least par-
ence of “shared-epitope” genes, suggesting that the com- tially, why only some RA patients respond adequately to
position of the human intestinal microbiome could also oral MTX. Finally, colonization of mice revealed the abili-
be partially dependent on the host genome. Second, ty of P copri to dominate the intestinal microbiota, result-
there were significant differences between the meta- ing in an increased sensitivity to Dextran sulfate sodium
genomes of the cohorts. Although this has not yet been (DSS)–induced colitis, a model of IBD. Other investiga-
fully studied, these variations may be functionally relevant tors have addressed the potential for local and systemic
as predictors of treatment response. This relates to the proinflammatory properties of the genus Prevotella,
fact that levels of dihydrofolate reductase (DHFR) are including its role in DSS-induced colitis and osteomyelitis
high in metagenome samples rich in Bacteroides, poten- (57,58).
tially competing with host DHFR for methotrexate Taken together, these studies reveal a potential role
(MTX) binding and metabolism. If this is the case, an of the gut microbiome in the pathogenesis of RA. However,
MICROBIOME IN RHEUMATIC DISEASES 41

these findings remain mostly correlative and do not neces- Work performed in other models has also shed
sarily represent causation. Further validating human studies light on the requirement of gut bacteria for SpA inflam-
and mechanistic approaches in gnotobiotic mice are needed mation. In the ankylosing enthesopathy mouse model the
to rigorously evaluate downstream RA immune events in animals spontaneously develop progressive enthesitis and
response to local microbial perturbation. ankylosis. The disease is also HLA–B27 dependent and
triggered by commensal microbiota (68). In the SKG
Spondyloarthritis and psoriatic arthritis model, mice develop SpA-like disease after systemic
injection of b-glucan, a major component of bacterial
Unlike the case with RA, the existence of SpA has and yeast cell walls (69). Furthermore, the incidence and
been documented for centuries, its prevalence is signifi- severity of arthritis and ileitis in SKG mice were depen-
cantly higher in Caucasian populations, its clinical mani- dent on the presence of host microbiota and downstream
festations are much more heterogeneous in nature, genes production of IL-23–dependent IL-17/IL-22 (70). Mice
(particularly HLA–B27) have a relatively larger effect in overexpressing IL-23 through minicircle DNA technology
disease incidence, and there is a prominent role of proin- develop enthesitis and osteoproliferation by activation of
flammatory Th17 cells and their signature cytokine, IL- entheseal resident T cells (71). Taken together, these
17. The epidemiologic relationship between intestinal experiments strongly support a model in which a geneti-
microorganisms, gut inflammation, and SpA is perhaps cally predisposed host reacts to a dysbiotic process by
even more remarkable. This is supported by the estab- altering the immune balance in the lamina propria
lished role of infectious diarrhea in the pathogenesis of toward a proinflammatory state dominated by IL-23 pro-
ReA, and the association between IBD and AS and PsA duction and activation of g/d T cells capable of amplifying
(mostly in the axial forms) (59–61). the response through cytokines such as IL-17 and tumor
This heterogeneity is also evident in the many, necrosis factor. This process ultimately leads to both local
though yet imperfect, animal models of SpA (Table 1). and systemic clinical repercussions in the form of colitis,
Several examples are elemental as proof-of-principle that psoriasis, enthesitis, and arthritis.
the microbiome is a significant contributor to disease. As discussed, AS and PsA patients are at
Pivotal studies first showed spontaneous inflammatory increased risk of developing clinical IBD. Intriguingly,
SpA-like disease in transgenic rats expressing HLA–B27 even a larger proportion of them have microscopic sub-
and multiple copies of human b2-microglobulin. The clinical gut inflammation, characterized by increased
phenotype is characterized by sacroiliitis, peripheral arth- infiltration of IL-22– and IL-23–producing cells (72–75).
ritis, psoriasiform skin inflammation, and colitis (62). Very few reports have described the intestinal micro-
However, when these rats are raised under germ-free biome in these disorders (Table 2). Using denaturing gra-
conditions, they fail to develop inflammatory intestinal or dient gel electrophoresis, Stebbings et al found a higher
peripheral joint disease (63). In contrast, HLA–B27– prevalence of sulfate-reducing bacteria in AS patients
transgenic rats colonized with various commensal Bacter- (76). Using 16S rRNA gene amplification, another study
oides species ultimately develop inflammation, suggesting demonstrated a distinct gut microbial signature in AS
that the presence of specific taxa is sufficient for disease (77). Compared to healthy controls, AS patients had a
initiation and propagation (64). The mechanism by which higher prevalence of several bacterial families in the ter-
this occurs remains elusive, but it is likely to be mediated minal ileum (e.g., Lachnospiraceae and Prevotellaceae)
by a combination of HLA–B27 misfolding and activation and a concomitant decrease in Ruminococcaceae and
of intestinal and circulating Th17 cells, in an IL-23–de- Rikenellaceae families. In children with enthesitis-related
pendent manner (65,66). The directionality of this gene– arthritis, the abundance of Faecalibacterium prausnitzii
microbiome interaction is a subject of intense research. was significantly lower than that in healthy controls (78).
A recent study compared the intestinal microbial com- Our group also defined the intestinal micro-
position of HLA–B27–transgenic rats (prior to disease biome of PsA patients compared to that of psoriasis
development) with that of wild-type controls, revealing patients and healthy controls (Figure 2). While psoriasis
an increase in Prevotella species and a decrease in Rike- patients and PsA patients both showed decreased levels
nellaceae and Akkermansia in the transgenic animals of Coprococcus, PsA patients were further characterized
(67). It is therefore plausible that, as in RA-like disease, by significantly lower levels of Akkermansia and Rumi-
microbiota alterations are a consequence of host genetic nococcus, suggesting a chronological loss of diversity
predisposition or perhaps a superimposed requirement that may correlate with the natural history of disease
for activation of downstream immunologic events in sus- (79). Curiously, studies in IBD revealed similar reduc-
ceptible animals. tions of the Ruminococcaceae family and Akkermansia
42 SCHER ET AL

genus. This state of intestinal dysbiosis in PsA patients notypic characterization of taxonomy and immune effects
correlated with decreased levels of both medium-chain are still needed to understand whether this approach
fatty acids and RANKL in the intestinal lumen, suggest- could eventually be adapted for the treatment of RA, PsA,
ing that microbiome perturbations are linked to altera- or SpA.
tions in mucosal integrity and perhaps dissemination of More specific approaches are also being used.
systemic inflammation. Many novel findings in the liter- One strategy relies on incorporating agents based on
ature on IBD, including the role of the intestinal virome single-strain live organisms or a defined bacterial consor-
in Crohn’s disease– and ulcerative colitis–specific dys- tium, leading to a modification of the host microbiome
biosis (80) and the colitogenic properties of sIgA-coated composition that does not depend on fecal microbial
human bacteria (81), may also prove highly relevant to transplantation. Several animal studies have demonstrat-
SpA and/or PsA and further advance our knowledge of ed successful treatment of autoimmune manifestations
their pathogenesis. with this methodology, including amelioration of IBD by
either Bacteroides fragilis (88) or a cocktail of Clostridia
Intestinal microbiome targeting strategies (89) through induction of colonic Treg cells. This is in
contrast to the procolitogenic and proarthritogenic effects
Therapies targeting intestinal bacteria have been of Bacteroides in HLA–B27–transgenic rats (64) and the
part of the antirheumatic strategy for many decades. detrimental role of Lactobacillus in adjuvant-induced
However, broad-spectrum antibiotic drugs have not arthritis in rats and joint inflammation in IL-1rn2/2 mice
demonstrated efficacy. Despite indirect evidence that (42,47,90). However, other studies have shown a benefi-
certain classes of antibacterials may hypothetically cial effect of Lactobacillus strains in CIA (91). Still, evi-
modulate the risk of RA and juvenile arthritis (82), this dence that probiotics have therapeutic value in RA is
approach is currently unsubstantiated. First, the notion scarce. Studies conducted so far are rather small and usu-
that certain classes of antibiotics target microorganisms ally show modest or no effect (92–94).
based on Gram stain properties has become outdated. Other sophisticated approaches exploit bacteria-
This is based on multiple culture-independent studies derived bioactive effectors. Several efforts use mole-
showing far-reaching taxonomic changes with drugs cules with immunomodulating properties, including
previously thought to be bacteriostatic against just a polysaccharides, structural proteins, and short-chain
few microorganisms (83). Moreover, indiscriminate use fatty acids, with evidence that some may be potential
of antibiotics at an early age could potentially result in treatments for IBD and other systemic autoimmune
clinical manifestations of various conditions, including disorders (88,95–98). Nevertheless, the effectiveness of
obesity and even autoimmune disorders (41,82,84). these compounds and their tolerability and long-term
Most recently, numerous groups are currently properties are yet to be elucidated.
attempting to modulate the composition of bacteria or
their byproducts to abrogate inflammatory responses. The
Conclusions
various methods now being used range from ecosystem-
level interventions to single-target approaches, including The vastness of the human intestinal microbiome
individual species or their metabolites (85). is increasingly being recognized. Its overwhelming diver-
Fecal microbial transplantation is used in an sity, resilience, and functional potential have transformed
attempt to restore a “healthy” intestinal microbiome in a the way we now view physiology and disease. Rapidly
recipient, using the full bacterial community from a close accumulating evidence indicates the existence of an intri-
donor. For decades, fecal microbial transplantation has cate symbiosis between the microbiome and its harboring
proven highly effective (and even curative) in antibiotic- host. In order to take full advantage of the essential
resistant Clostridium difficile infectious colitis (86). Despite byproducts generated by this bioreactor, humans have
striking results, the use of this technique presents its own evolved to actively tolerate this constant and massive
array of challenges, including how to define a healthy antigenic load within the intestinal lumen through a state
microbiome and who to consider an appropriate donor. of low-grade inflammation. However, perturbations in
Nevertheless, fecal microbial transplantation has already the relative composition of commensal microbiota can
been applied to autoimmune diseases, most notably in alter the immune homeostasis of the lamina propria and
IBD. Although the results are based on relatively small elicit aberrant systemic responses.
samples, they remain promising as in some series patients Animal models of inflammatory arthritis have
have responded favorably to fecal microbial transplanta- considerably advanced our mechanistic understanding
tion (87). Well-designed clinical studies paired with phe- of the dysbiotic process in connection with key cellular
MICROBIOME IN RHEUMATIC DISEASES 43

responses and downstream immune events. Given the 8. Zhao L. Genomics: the tale of our other genome. Nature 2010;
465:879–80.
presence of a preclinical phase in many autoimmune 9. Littman DR, Pamer EG. Role of the commensal microbiota in nor-
diseases (i.e., autoantibodies preceding clinically evid- mal and pathogenic host immune responses. Cell Host Microbe
ent inflammation), the microbiome could hypothetical- 2011;10:311–23.
10. Round JL, Mazmanian SK. The gut microbiota shapes intestinal
ly contribute causally through a variety of mechanisms. immune responses during health and disease [published erratum
A state of dysbiosis could lead to a specific adaptive appears in Nat Rev Immunol 2009;9:600]. Nat Rev Immunol 2009;
immunodifferentiation of effector cells (e.g., segmented 9:313–23.
11. Hooper LV, Macpherson AJ. Immune adaptations that maintain
filamentous bacteria–dependent Th17 cell activation), homeostasis with the intestinal microbiota. Nat Rev Immunol
to the provision of neoantigens (i.e., P gingivalis–driven 2010;10:159–69.
citrullination of peptides and generation of ACPAs), 12. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight
R, Gordon JI. The Human Microbiome Project. Nature 2007;
and/or could result in generation of costimulatory sig- 449:804–10.
nals (e.g., in the presence of potentially arthritogenic 13. Bornigen D, Morgan XC, Franzosa EA, Ren B, Xavier RJ, Garrett
ACPAs, disease could be triggered only through a WS, et al. Functional profiling of the gut microbiome in disease-
associated inflammation. Genome Med 2013;5:65.
“second event,” driven by bacterial components and 14. Ursell LK, Haiser HJ, van Treuren W, Garg N, Reddivari L,
consequent cellular immune response). Vanamala J, et al. The intestinal metabolome: an intersection
As more human microbiome studies become between microbiota and host. Gastroenterology 2014;146:1470–6.
15. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R.
available, many questions will require rigorous explora- Diversity, stability and resilience of the human gut microbiota.
tion. These involve the directionality of the dysbiotic Nature 2012;489:220–30.
process (i.e., are microbiome alterations actual inciting 16. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan
A, Ley RE, et al. A core gut microbiome in obese and lean
events in the autoimmune process, or are they instead twins. Nature 2009;457:480–4.
a consequence of ecologic adaptation to the inflamma- 17. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-
tory milieu?), the specificity of commensal antigen rec- wide association study of gut microbiota in type 2 diabetes.
Nature 2012;490:55–60.
ognition by T cells, the timing of microbial pertur- 18. Arthur JC, Perez-Chanona E, Muhlbauer M, Tomkovich S, Uronis
bation in the different phases of disease (i.e., preclinical, JM, Fan TJ, et al. Intestinal inflammation targets cancer-inducing
early, and chronic) and the contribution of communities activity of the microbiota. Science 2012;338:120–3.
19. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT,
in other body niches such as the periodontal and lung et al. Intestinal microbiota metabolism of L-carnitine, a nutrient
(in RA) and skin (in PsA), and the relationship of the in red meat, promotes atherosclerosis. Nat Med 2013;19:576–85.
microbiome and metabolome to immunosuppressive 20. Gao Z, Tseng CH, Strober BE, Pei Z, Blaser MJ. Substantial
alterations of the cutaneous bacterial biota in psoriatic lesions.
drugs (i.e., could the microbiome be involved in pharma- PloS One 2008;3:e2719.
cokinetics of oral DMARDs and/or serve as a predictor 21. Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward
of drug response?). Ultimately, the answer to many of DV, et al. Dysfunction of the intestinal microbiome in inflamma-
tory bowel disease and treatment. Genome Biol 2012;13:R79.
these questions will largely depend on whether we can 22. Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, van
successfully target the microbiome for therapeutic pur- Treuren W, Ren B, et al. The treatment-naive microbiome in
poses in autoimmune and rheumatic diseases. new-onset Crohn’s disease. Cell Host Microbe 2014;15:382–92.
23. Willing BP, Dicksved J, Halfvarson J, Andersson AF, Lucio M,
Zheng Z, et al. A pyrosequencing study in twins shows that gas-
AUTHOR CONTRIBUTIONS trointestinal microbial profiles vary with inflammatory bowel dis-
ease phenotypes. Gastroenterology 2010;139:1844–54.e1.
All authors drafted the article, revised it critically for important
24. Warden CC. The toxemic factor in rheumatoid arthritis. Cal
intellectual content, and approved the final version to be published.
State J Med 1909;7:299–301.
25. Moos V, Schneider T. Changing paradigms in Whipple’s disease
REFERENCES and infection with Tropheryma whipplei. Eur J Clin Microbiol
Infect Dis 2011;30:1151–8.
1. Lederberg J. Infectious history. Science 2000;288:287–93. 26. Ross CB, Scott HW, Pincus T. Jejunoileal bypass arthritis. Bail-
2. Koch R. An address on bacteriological research. Br Med J 1890; lieres Clin Rheumatol 1989;3:339–55.
2:380–3. 27. Svartz N. The treatment of rheumatic polyarthritis with acid azo
3. Scher JU, Abramson SB. The microbiome and rheumatoid arth- compounds. Rheumatism 1948;4:180–5.
ritis. Nat Rev Rheumatol 2011;7:569–78. 28. Svartz N. The primary cause of rheumatoid arthritis is an infec-
4. Chervonsky AV. Influence of microbial environment on autoim- tion—the infectious agent exists in milk. Acta Med Scand 1972;
munity. Nat Immunol 2010;11:28–35. 192:231–9.
5. Yurkovetskiy LA, Pickard JM, Chervonsky AV. Microbiota and 29. Rothschild BM, Turner KR, DeLuca MA. Symmetrical erosive
autoimmunity: exploring new avenues. Cell Host Microbe 2015; peripheral polyarthritis in the Late Archaic Period of Alabama.
17:548–52. Science 1988;241:1498–501.
6. Peterson J, Garges S, Giovanni M, McInnes P, Wang L, Schloss 30. Deighton CM, Gray J, Bint AJ, Walker DJ. Specificity of the
JA, et al. The NIH Human Microbiome Project. Genome Res proteus antibody response in rheumatoid arthritis. Ann Rheum
2009;19:2317–23. Dis 1992;51:1206–7.
7. Human Microbiome Project Consortium. Structure, function and 31. Hoffman RW, O’Sullivan FX, Schafermeyer KR, Moore TL,
diversity of the healthy human microbiome. Nature 2012;486:207–14. Roussell D, Watson-McKown R, et al. Mycoplasma infection
44 SCHER ET AL

and rheumatoid arthritis: analysis of their relationship using 50. Yang Y, Torchinsky MB, Gobert M, Xiong H, Xu M, Linehan
immunoblotting and an ultrasensitive polymerase chain reaction JL, et al. Focused specificity of intestinal TH17 cells towards
detection method. Arthritis Rheum 1997;40:1219–28. commensal bacterial antigens. Nature 2014;510:152–6.
32. Payne JB, Golub LM, Thiele GM, Mikuls TR. The link between 51. Toivanen P, Vaahtovuo J, Eerola E. Influence of major histo-
periodontitis and rheumatoid arthritis: a periodontist’s perspec- compatibility complex on bacterial composition of fecal flora.
tive. Curr Oral Health Rep 2015;2:20–9. Infect Immun 2001;69:2372–7.
33. Mikuls TR, Payne JB, Yu F, Thiele GM, Reynolds RJ, Cannon 52. Gomez A, Luckey D, Yeoman CJ, Marietta EV, Berg Miller
GW, et al. Periodontitis and Porphyromonas gingivalis in ME, Murray JA, et al. Loss of sex and age driven differences in
patients with rheumatoid arthritis. Arthritis Rheumatol 2014;66: the gut microbiome characterize arthritis-susceptible 0401 mice
1090–100. but not arthritis-resistant 0402 mice. PloS One 2012;7:e36095.
34. Dissick A, Redman RS, Jones M, Rangan BV, Reimold A, Griffiths 53. Eerola E, Mottonen T, Hannonen P, Luukkainen R, Kantola I,
GR, et al. Association of periodontitis with rheumatoid arthritis: a Vuori K, et al. Intestinal flora in early rheumatoid arthritis. Br J
pilot study. J Periodontol 2010;81:223–30. Rheumatol 1994;33:1030–8.
35. Scher JU, Ubeda C, Equinda M, Khanin R, Buischi Y, Viale A, 54. Vaahtovuo J, Munukka E, Korkeamaki M, Luukkainen R, Toivanen
et al. Periodontal disease and the oral microbiota in new-onset P. Fecal microbiota in early rheumatoid arthritis. J Rheumatol 2008;
rheumatoid arthritis. Arthritis Rheum 2012;64:3083–94. 35:1500–5.
36. De Aquino SG, Abdollahi-Roodsaz S, Koenders MI, van de Loo 55. Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski
FA, Pruijn GJ, Marijnissen RJ, et al. Periodontal pathogens C, et al. Expansion of intestinal Prevotella copri correlates with
directly promote autoimmune experimental arthritis by inducing enhanced susceptibility to arthritis. Elife 2013;2:e01202.
a TLR2- and IL-1-driven Th17 response. J Immunol 2014;192: 56. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet
4103–11. JB, Massart S, et al. Impact of diet in shaping gut microbiota
37. Demoruelle MK, Weisman MH, Simonian PL, Lynch DA, Sachs revealed by a comparative study in children from Europe and
PB, Pedraza IF, et al. Airways abnormalities and rheumatoid rural Africa. Proc Natl Acad Sci U S A 2010;107:14691–6.
arthritis–related autoantibodies in subjects without arthritis: ear- 57. Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth
ly injury or initiating site of autoimmunity? Arthritis Rheum CJ, et al. NLRP6 inflammasome regulates colonic microbial
2012;64:1756–61. ecology and risk for colitis. Cell 2011;145:745–57.
38. Willis VC, Demoruelle MK, Derber LA, Chartier-Logan CJ, Parish 58. Lukens JR, Gurung P, Vogel P, Johnson GR, Carter RA,
MC, Pedraza IF, et al. Sputum autoantibodies in patients with McGoldrick DJ, et al. Dietary modulation of the microbiome
established rheumatoid arthritis and subjects at risk of future clini- affects autoinflammatory disease. Nature 2014;516:246–9.
cally apparent disease. Arthritis Rheum 2013;65:2545–54. 59. Arvikar SL, Fisher MC. Inflammatory bowel disease associated
39. Catrina AI, Ytterberg AJ, Reynisdottir G, Malmstrom V, Klareskog arthropathy. Curr Rev Musculoskelet Med 2011;4:123–31.
L. Lungs, joints and immunity against citrullinated proteins in rheu- 60. Mielants H, Veys EM, De Vos M, Cuvelier C, Goemaere S, De
matoid arthritis. Nat Rev Rheumatol 2014;10:645–53.
Clercq L, et al. The evolution of spondyloarthropathies in relation
40. Ytterberg AJ, Joshua V, Reynisdottir G, Tarasova NK, Rutishauser
to gut histology. I. Clinical aspects. J Rheumatol 1995;22:2266–72.
D, Ossipova E, et al. Shared immunological targets in the lungs and
61. Li WQ, Han JL, Chan AT, Qureshi AA. Psoriasis, psoriatic
joints of patients with rheumatoid arthritis: identification and vali-
arthritis and increased risk of incident Crohn’s disease in US
dation. Ann Rheum Dis 2015;74:1772–7.
women. Ann Rheum Dis 2013;72:1200–5.
41. Sandberg ME, Bengtsson C, Klareskog L, Alfredsson L, Saevarsdottir
62. Hammer RE, Maika SD, Richardson JA, Tang JP, Taurog JD.
S. Recent infections are associated with decreased risk of rheumatoid
Spontaneous inflammatory disease in transgenic rats expressing
arthritis: a population-based case-control study. Ann Rheum Dis 2015;
74:904–7. HLA-B27 and human b2m: an animal model of HLA-B27-
42. Kohashi O, Kohashi Y, Takahashi T, Ozawa A, Shigematsu N. associated human disorders. Cell 1990;63:1099–112.
Reverse effect of gram-positive bacteria vs. gram-negative bacte- 63. Taurog JD, Richardson JA, Croft JT, Simmons WA, Zhou M,
ria on adjuvant-induced arthritis in germfree rats. Microbiol Fernandez-Sueiro JL, et al. The germfree state prevents devel-
Immunol 1985;29:487–97. opment of gut and joint inflammatory disease in HLA-B27
43. Kohashi O, Kohashi Y, Takahashi T, Ozawa A, Shigematsu N. transgenic rats. J Exp Med 1994;180:2359–64.
Suppressive effect of Escherichia coli on adjuvant-induced arth- 64. Rath HC, Herfarth HH, Ikeda JS, Grenther WB, Hamm TE Jr,
ritis in germ-free rats. Arthritis Rheum 1986;29:547–53. Balish E, et al. Normal luminal bacteria, especially Bacteroides
44. Breban MA, Moreau MC, Fournier C, Ducluzeau R, Kahn MF. species, mediate chronic colitis, gastritis, and arthritis in HLA-
Influence of the bacterial flora on collagen-induced arthritis in B27/human b2 microglobulin transgenic rats. J Clin Invest 1996;
susceptible and resistant strains of rats. Clin Exp Rheumatol 98:945–53.
1993;11:61–4. 65. Glatigny S, Fert I, Blaton MA, Lories RJ, Araujo LM, Chiocchia
45. Caccese RG, Zimmerman JL, Carlson RP. Bacterial lipopolysac- G, et al. Proinflammatory Th17 cells are expanded and induced
charide potentiates type II collagen-induced arthritis in mice. by dendritic cells in spondylarthritis-prone HLA–B27–transgenic
Mediators Inflamm 1992;1:273–9. rats. Arthritis Rheum 2012;64:110–20.
46. Van den Broek MF, van Bruggen MC, Koopman JP, Hazenberg 66. DeLay ML, Turner MJ, Klenk EI, Smith JA, Sowders DP, Colbert
MP, van den Berg WB. Gut flora induces and maintains resis- RA. HLA–B27 misfolding and the unfolded protein response aug-
tance against streptococcal cell wall-induced arthritis in F344 ment interleukin-23 production and are associated with Th17 acti-
rats. Clin Exp Immunol 1992;88:313–7. vation in transgenic rats. Arthritis Rheum 2009;60:2633–43.
47. Abdollahi-Roodsaz S, Joosten LA, Koenders MI, Devesa I, Roelofs 67. Lin P, Bach M, Asquith M, Lee AY, Akileswaran L, Stauffer P,
MF, Radstake TR, et al. Stimulation of TLR2 and TLR4 differen- et al. HLA-B27 and human b2-microglobulin affect the gut
tially skews the balance of T cells in a mouse model of arthritis. microbiota of transgenic rats. PloS One 2014;9:e105684.
J Clin Invest 2008;118:205–16. 68. Sinkorova Z, Capkova J, Niederlova J, Stepankova R, Sinkora J.
48. Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, Commensal intestinal bacterial strains trigger ankylosing enthes-
et al. Gut-residing segmented filamentous bacteria drive auto- opathy of the ankle in inbred B10.BR (H-2k) male mice. Hum
immune arthritis via T helper 17 cells. Immunity 2010;32:815–27. Immunol 2008;69:845–50.
49. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, 69. Ruutu M, Thomas G, Steck R, Degli-Esposti MA, Zinkernagel MS,
et al. Induction of intestinal Th17 cells by segmented filamen- Alexander K, et al. b-glucan triggers spondylarthritis and Crohn’s
tous bacteria. Cell 2009;139:485–98. disease–like ileitis in SKG mice. Arthritis Rheum 2012;64:2211–22.
MICROBIOME IN RHEUMATIC DISEASES 45

70. Rehaume LM, Mondot S, Aguirre de Carcer D, Velasco J, 84. Cho I, Yamanishi S, Cox L, Methe BA, Zavadil J, Li K, et al.
Benham H, Hasnain SZ, et al. ZAP-70 genotype disrupts the rela- Antibiotics in early life alter the murine colonic microbiome and
tionship between microbiota and host, leading to spondyloarthritis adiposity. Nature 2012;488:621–6.
and ileitis in SKG mice. Arthritis Rheumatol 2014;66:2780–92. 85. Olle B. Medicines from microbiota. Nat Biotechnol 2013;31:
71. Sherlock JP, Joyce-Shaikh B, Turner SP, Chao CC, Sathe M, Grein 309–15.
J, et al. IL-23 induces spondyloarthropathy by acting on ROR-gt1 86. Van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal
CD31CD42CD82 entheseal resident T cells. Nat Med 2012;18: EG, de Vos WM, et al. Duodenal infusion of donor feces for
1069–76. recurrent Clostridium difficile. N Engl J Med 2013;368:407–15.
72. Mielants H, Veys EM, Goemaere S, Goethals K, Cuvelier C, De 87. Colman RJ, Rubin DT. Fecal microbiota transplantation as ther-
Vos M. Gut inflammation in the spondyloarthropathies: clinical, apy for inflammatory bowel disease: a systematic review and
radiologic, biologic and genetic features in relation to the type meta-analysis. J Crohns Colitis 2014;8:1569–81.
of histology: a prospective study. J Rheumatol 1991;18:1542–51. 88. Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis
73. Van Praet L, van den Bosch FE, Jacques P, Carron P, Jans L, factor prevents intestinal inflammatory disease. Nature 2008;453:
Colman R, et al. Microscopic gut inflammation in axial spondyloarthri- 620–5.
tis: a multiparametric predictive model. Ann Rheum Dis 2013;72:414–7. 89. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose
74. Ciccia F, Bombardieri M, Principato A, Giardina A, Tripodo C, Y, et al. Induction of colonic regulatory T cells by indigenous Clos-
Porcasi R, et al. Overexpression of interleukin-23, but not interleukin- tridium species. Science 2011;331:337–41.
17, as an immunologic signature of subclinical intestinal inflammation 90. Lehman TJ, Allen JB, Plotz PH, Wilder RL. Lactobacillus casei
in ankylosing spondylitis. Arthritis Rheum 2009;60:955–65. cell wall–induced arthritis in rats: cell wall fragment distribu-
75. Ciccia F, Accardo-Palumbo A, Alessandro R, Rizzo A, Principe tion and persistence in chronic arthritis–susceptible LEW/N and
S, Peralta S, et al. Interleukin-22 and interleukin-22–producing –resistant F344/N rats. Arthritis Rheum 1984;27:939–42.
NKp441 natural killer cells in subclinical gut inflammation in 91. Amdekar S, Singh V, Singh R, Sharma P, Keshav P, Kumar A.
Lactobacillus casei reduces the inflammatory joint damage asso-
ankylosing spondylitis. Arthritis Rheum 2012;64:1869–78.
ciated with collagen-induced arthritis (CIA) by reducing the pro-
76. Stebbings S, Munro K, Simon MA, Tannock G, Highton J,
inflammatory cytokines: Lactobacillus casei: COX-2 inhibitor.
Harmsen H, et al. Comparison of the faecal microflora of pa-
J Clin Immunol 2011;31:147–54.
tients with ankylosing spondylitis and controls using molecular
92. Jenks K, Stebbings S, Burton J, Schultz M, Herbison P, Highton
methods of analysis. Rheumatology (Oxford) 2002;41:1395–401.
J. Probiotic therapy for the treatment of spondyloarthritis: a ran-
77. Costello ME, Ciccia F, Willner D, Warrington N, Robinson PC,
domized controlled trial. J Rheumatol 2010;37:21182125.
Gardiner B, et al. Intestinal dysbiosis in ankylosing spondylitis.
93. Alipour B, Homayouni-Rad A, Vaghef-Mehrabany E, Sharif SK,
Arthritis Rheumatol 2015;67:686–91. Vaghef-Mehrabany L, Asghari-Jafarabadi M, et al. Effects of Lac-
78. Stoll ML, Kumar R, Morrow CD, Lefkowitz EJ, Cui X, Genin tobacillus casei supplementation on disease activity and inflamma-
A, et al. Altered microbiota associated with abnormal humoral tory cytokines in rheumatoid arthritis patients: a randomized
immune responses to commensal organisms in enthesitis-related double-blind clinical trial. Int J Rheum Dis 2014;17:519–27.
arthritis. Arthritis Res Ther 2014;16:486. 94. Pineda M, Thompson SF, Summers K, de Leon F, Pope J, Reid
79. Scher JU, Ubeda C, Artacho A, Attur M, Isaac S, Reddy SM, et al. G. A randomized, double-blinded, placebo-controlled pilot study
Decreased bacterial diversity characterizes the altered gut micro- of probiotics in active rheumatoid arthritis. Med Sci Monit 2011;
biota in patients with psoriatic arthritis, resembling dysbiosis in 17:CR347–54.
inflammatory bowel disease. Arthritis Rheumatol 2015;67:128–39. 95. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA,
80. Norman JM, Handley SA, Baldridge MT, Droit L, Liu CY, Keller Bohlooly YM, et al. The microbial metabolites, short-chain fatty
BC, et al. Disease-specific alterations in the enteric virome in in- acids, regulate colonic Treg cell homeostasis. Science 2013;341:
flammatory bowel disease. Cell 2015;160:447–60. 569–73.
81. Palm NW, de Zoete MR, Cullen TW, Barry NA, Stefanowski J, 96. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi
Hao L, et al. Immunoglobulin A coating identifies colitogenic D, et al. Commensal microbe-derived butyrate induces the differ-
bacteria in inflammatory bowel disease. Cell 2014;158:1000–10. entiation of colonic regulatory T cells. Nature 2013;504:446–50.
82. Arvonen M, Virta LJ, Pokka T, Kroger L, Vahasalo P. Repeated 97. Wang Y, Telesford KM, Ochoa-Reparaz J, Haque-Begum S,
exposure to antibiotics in infancy: a predisposing factor for juve- Christy M, Kasper EJ, et al. An intestinal commensal symbiosis
nile idiopathic arthritis or a sign of this group’s greater suscepti- factor controls neuroinflammation via TLR2-mediated CD39 sig-
bility to infections? J Rheumatol 2015;42:521–6. nalling. Nat Commun 2014;5:4432.
83. Dethlefsen L, Relman DA. Incomplete recovery and individual- 98. Yan F, Cao H, Cover TL, Whitehead R, Washington MK, Polk
ized responses of the human distal gut microbiota to repeated DB. Soluble proteins produced by probiotic bacteria regulate
antibiotic perturbation. Proc Natl Acad Sci U S A 2011;108 intestinal epithelial cell survival and growth. Gastroenterology
Suppl 1:4554–61. 2007;132:562–75.

You might also like