You are on page 1of 14

REVIEWS

How the microbiota shapes


rheumatic diseases
Tom Van de Wiele1, Jens T. Van Praet24, Massimo Marzorati1, Michael B. Drennan2,3
and Dirk Elewaut2,3
Abstract | The human gut harbours a tremendously diverse and abundant microbial community
that correlates with, and even modulates, many health-related processes. The mucosal interfaces
are particularly active sites of microorganismhost interplay. Growing insight into the
characteristic composition and functionality of the mucosal microbiota has revealed that the
microbiota is involved in mucosal barrier integrity and immune function. This involvement affects
proinflammatory and anti-inflammatory processes not only at the epithelial level, but also at
remote sites such as the joints. Here, we review the role of the gut microbiota in shaping local
and systemic immune responses and how disturbances in the hostmicroorganism interplay
can potentially affect the development and progression of rheumatic diseases. Increasing our
understanding of how to promote hostmicroorganism homeostasis could therefore reveal
novel strategies for the prevention or alleviation of rheumatic disease.

The human body is colonized by an abundant and genet Peyers patches and cryptopatches (the existence of the
ically diverse microbial community, the microbiota. latter being controversial in humans), have therefore
Although the number of our microbial colonizers is one evolved to enable the immune system to differentiate
1
Laboratory of Microbial order of magnitude higher than the number of our cells, between protective and tolerogenic responses57. Not
Ecology and Technology the number of genes encoded by the human microbiome surprisingly, breaching the integrity of the mucosal
(LabMET), Faculty of
(the genome of all microbiota) exceeds that encoded by surfaces can result in inflammation in the intestine but
Bioscience Engineering,
Ghent University, Coupure the human genome by 100fold. The human microbiota also in other body regions. The microbiota contributes
Links 653, Ghent, B-9000, is considered a major player in health-related processes, to the systemic immune effects of local inflamma
Belgium. either by direct influence or through interaction with tion in the mucosa and the resulting pathologies. Gut
2
Laboratory for Molecular other health determinants, including genetics, diet, life inflammation and the microbiota have been associated
Immunology and
Inflammation, Department
style, medical practices, hygiene and the exposome (the with musculoskeletal diseases such as spondyloarthri
of Rheumatology, totality of environmental factors humans are exposed to tis (SpA, which includes ankylosing spondylitis [AS])
Ghent University Hospital, during their lifetime). The rapid rise of several omics and rheumatoid arthritis (RA), and might also have a
De Pintelaan 185, techniques and their application in the context of large role in other rheumatic diseases such as systemic lupus
Ghent, B-9000, Belgium.
research initiatives, such as the Human Microbiome erythematosus (SLE).
3
Unit for Molecular
Immunology and Project1 in the USA and MetaHIT2 in Europe, have In this Review, we highlight progress made in the
Inflammation, revolutionized the field by shedding light on correlations past few years on the role of the human microbiota in
VIB Inflammation Research between health status and microbiome composition, maintaining immune homeostasis, and how shifts in
Center, Ghent University, specific expression of genes, translation into proteins or microbiome composition can contribute to rheumatic
Fiers-Schell-Van Montagu
building, Technologiepark
production of specific metabolites. The influence of the diseases. We also discuss current strategies to modulate
927, B-9052 Ghent human microbiota on health is being increasingly rec altered microbiotas and how this modulation could have
(Zwijnaarde), Belgium. ognized not to be solely confined to the gut region. For therapeutic applications.
4
Division of Nephrology example, whole-genome shotgun data from the Human
and Infectious Diseases,
Microbiome Project has revealed the microbiome to be The human gut microbiota
AZ SintJan Brugge
Oostende AV, Ruddershove 10, composed of up to 30% of Clostridium clusters IV and The human intestine is colonized by a complex micro
8000 Bruges, Belgium. XIVa3, many of which have immune-modulating effects4. bial ecosystem, which can be considered as a separate
Correspondence to D.E. One of the main tasks of the mucosal immune sys organ within the human host. This human microbiota
dirk.elewaut@ugent.be tem is to maintain a balance between protection against has a coding capacity that exceeds that of the liver by a
doi:10.1038/nrrheum.2016.85 potentially harmful microorganisms and tolerance to factor of 150 (see BOX1 for more facts and figures on
Published online 16 Jun 2016 dietary antigens. Organized lymphoid nodules, such as the human gut and its microbiota). High-throughput

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 1



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points tissues20. Thousands of small molecules derived from


microorganisms present in the gut or from food and
Gut microbiota shape immune responses drug intake are expected to be present in humans21,22.
Both innate and adaptive immunity are influenced by gut microbiota, locally in the As the functional characterization of individual
gut as well as systemically microbiotas progresses, growing evidence is showing
Intestinal dysbiosis is a feature of several inflammatory rheumatic disorders that the gut microbiota is involved in key activities
Development of anticitrullinated protein antibodies is modulated by HLA shared linked to health and disease (BOX2). The human micro
epitope alleles, smoking and specialized microbiota at mucosal sites biota comprises up to 100 trillion cells, which are
Gut inflammation in spondyloarthritis is linked to a more severe disease course and exposed to the host through a mucus-covered surface
risk of developing Crohn disease and is associated with intestinal dysbiosis area of 32m2 (REF.23). Therefore, any factor that dis
Restoring intestinal homeostasis by altered microbiota is an attractive therapeutic rupts the hostmicroorganism equilibrium (such as
strategy to combat rheumatic diseases depletion and/or thinning of the mucus layer; drastic
changes in diet behaviour and/or malnourishment; use
of antibiotics; inflammation; infection with pathogens;
culture-independent sequencing analyses have shown gastrointestinal surgery; etc.) might affect homeostasis
that microbiota from the gastrointestinal tract (GIT) and have a dramatic effect in the modulation of immune
are more complex than microbial populations from functions of the host by the microbiota2427. The result
other body sites, with more than 1,000 microbial spe ing microbial imbalance, also termed dysbiosis, can lead
cies identified to date8,9. Most of the phylotypes (approx to elevated host inflammation and, subsequently, to the
imately 90%) identified in the GIT belong to two phyla, development of intestinal disorders28. Dysbiosis has been
Bacteroidetes and Firmicutes, whereas Actinobacteria, associated with colon discomfort, and with obesity and
Proteobacteria and Verrucomicrobia are represented related metabolic disorders, such as diabetes, nonalcoholic
with a lower abundance10. The complexity of the gut steatohepatitis and cardiovascular diseases29.
microbiota is increased by the presence of methanogenic A critical window in establishing this hostmicrobial
archaea (such as Methanobrevibacter smithii), yeasts and interplay occurs early in life. Early infancy seems to be
viruses (mainly phages)11. Attempts have been made to a critical period for intestinal colonization, which has
identify a core microbiota with a specific composition. marked effects on the maturation of the immune system.
However, although only two phyla are dominant in the Modification of this process of colonization by mode of
GIT microbiota, significant interpersonal variability delivery at birth, breastfeeding or administration of anti
exists, as evidenced by interindividual differences in biotics has been linked to long-term health outcomes.
the colonization and composition of hundreds of spe For example, the lack of contact with the mothers vag
cies within these phyla; differences in life events, age, inal and perineal microbiota for children born through
geographic origin, diet, disease, lifestyle and/or drug caesarian section results in a different faecal microbiota,
uptake lie at the basis of this interindividual variability. the composition of which is determined moreseo by the
The set of genes of each individual can also influence maternal skin microbiota and the microorganisms in the
the composition of the gut microbiota12. hospital environment than the microbiota of children
Some species that have a specific function within the born through vaginal delivery29. These differences in
gut ecosystem have been identified in the gut micro early colonization are still distinguishable after several
biota, including Faecalibacterium prausnitzii, a socalled years and have been suggested as a possible underlying
peacekeeper with recognized anti-inflammatory prop factor for the prevalence of type1 diabetes mellitus,
erties13, and Roseburia intestinalis and Ruminococcus asthma and coeliac disease among individuals born
bromii, which are specialist primary degraders of indi through caesarian section3032. Breastfeeding lowers the
gestible fibres14,15 and providers of butyrate16. These and risk of asthma and coeliac disease later in life33,34, whereas
similar findings have led to a gradual shift in the focus antibiotic use in the first year of life enhances the risk of
of research in this area, from the study of the compo inflammatory bowel disease (IBD) in childhood35.
sition of the microbiota to the study of its functional
activity. Much research on the microbiota has aimed to The gut microbiota and immunity
identify the core ecological functions of this organ and Immune recognition of gut microorganisms
its influence on human health13. Turnbaugh etal.17, for The mucosal microbiota has great potential to interact
example, showed that a core gut microbiome can exist with the host, both in a positive and in a negative man
at the level of shared functional genes, even though the ner, owing to hostmicroorganism proximity over a
composition of the microbial community is as unique large surface area in the gut. Therefore, the immune sys
as an individuals fingerprint. Consequently, metatran tem needs to be vigilant, which is illustrated by the fact
scriptomics and metaproteomics have been introduced that gut-associated lymphoid tissue (GALT) represents
as tools to investigate the gene expression levels of intes 70% of the bodys immune system, and 80% of all IgA-
tinal bacteria using faecal samples18,19. More recently, producing plasma cells reside in the intestinal lamina
metabolomics approaches have also been successfully propria36. Both the innate and adaptive immune systems
used in the analysis of the microbiota. These approaches can respond to microbiota. Constant signalling between
involve the unbiased quantitative and qualitative analysis microorganisms and the host exists in the gut epithe
of the complete set of small molecules (molecular weight lium37. Intestinal epithelial cells and dendritic cells pro
<1,000 Da) present in cells, organisms, body fluids or truding beyond the epithelium are crucial for the initial

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 1 | The human gut and its microbiome: facts and figures
The whole digestive tract is ~9m long. The small intestine is the longest part, at ~7m
The mean total mucosal surface of the digestive tract is ~32m2 on average23
The entire microbiota in a person weighs only 0.52kg (wet weight)
The intestinal tract is a nutrient-rich environment containing up to 100 trillion (1014) microorganisms, the vast majority
of which reside in the colon where densities approach 10111012 cells per ml, the highest recorded densities for any
microbial habitat
At birth, the gastrointestinal tract is sterile. Breast milk has a crucial role in shaping the composition of the microbial
community via transmission of the milk microbiota, including species (for example, Bifidobacterium spp.) selected by
milk oligosaccharides, and maternal antibodies to the infant gut183
The activity and composition of the gut microbiota change from infancy to old age in response to the genetic
background, diet, immune system and health status of the host184
The dominant bacterial groups in different parts of the gastrointestinal tract reflect physiological differences along
the length of the gut (such as oxygen availability, presence of antimicrobial peptides, availability of carbon sources
and pH)183. The oral microbiota is frequently dominated by Firmicutes (for example, Streptococcus salivarius) and
Actinobacteria. The healthy human stomach is dominated by Prevotella, Streptococcus, Veillonella, Rothia and
Haemophilus genera. In the small intestine, the Lactobacillaceae and Enterobacteriaceae families dominate, whereas
the colon is mainly characterized by the presence of species from the families Bacteroidaceae, Prevotellaceae,
Rikenellaceae, Lachnospiraceae and Ruminococcaceae. The mucosa is enriched with mucin degraders (for example,
Bacteroides fragilis and Akkermansia muciniphila).
Today, 7.3 billion humans live on Earth. Together, we represent a reservoir of 10231024 gut microbial cells
Each individual has a unique signature gut microbiota as personal as a fingerprint
The total number of bacteria in our gut is ~10 times greater than the total number of our somatic and germ cells
The number of functional genes in gut microorganisms is ~150 times higher than the number of genes in the human
genome
The human microbiome is defined as the collective genomes of the microorganisms (bacteria, bacteriophages, fungi,
protozoa and viruses) that live inside and on the human body

recognition of mucosal and luminal microorganisms (TFH) cells39. Maturation of Bcells into high-affinity IgA-
(FIG.1). The important distinction between trustworthy secreting plasma cells occurs within Peyers patches in
microorganisms and (opportunistic) pathogens, which neonates and assists in maintaining an immune barrier
determines whether the immune system is regulated or within the intestine into adulthood. Some low-affinity
activated, is fulfilled by pattern-recognition receptors IgAs have also been shown to be generated in isolated
(PRRs). PRRs comprise membrane-associated Toll-like lymphoid follicles and in the lamina propria in a Tcell-
receptors (TLRs), cytosolic nucleotide oligomerization independent manner, and require innate lymphoid cells
domain (NOD)-like receptors and secretory collectins. as well as TLR and/or inflammatory cytokine signalling
The role of PRRs involves recognition of microorganism- for maturation40,41. The role of innate lymphoid cell pop
associated molecular patterns, which leads to the rapid ulations in the regulation of inflammation has received
engagement of other essential components of the innate much attention in the past few years42.
immune system, including those that produce antimi
crobial peptides. Such a response is mostly aspecific, so Microenvironments in the gut mucosa
damage to the endogenous microbiota often occurs as a The gut microbiota cannot be considered as one unique
result38. A more specific response is needed for long-term population, but more as a collection of ecosystems that
protection. colonize different microenvironments in the gut 10.
In the intestine, microbial antigens can be recognized A longitudinal gradient exists in the composition and
by a variety of antigen-presenting cells, including den functionality of the microbiota over the length of the
dritic cells, macrophages and Bcells. Before recognition human gut (from the terminal ileum, to the proximal
can take place, the antigens need to cross the epithelial colon, to the distal colon). Microbiota specifically asso
barrier, which can occur either via intestinal epithelial ciated with nutrient platforms (food-particle-associated
cells or via other specialized cell types, such as M cells. surfaces) also exist in the gut lumen. In addition, a
Dendritic cells can sample antigens directly in the intesti cross-sectional gradient from the gut lumen to the
nal lumen, after which they typically migrate to the mes mucosal environment is present. The mucus layer has
enteric lymph nodes. Here, dendritic cells can modulate a particular double structure, with an inner layer, close
Tcell activation and maturation into specialized func to the epithelium, and an outer layer. The inner layer is
tional subsets (Thelper (TH) cell subsets such as TH1, TH2 rigid because of elevated levels of mucin2; owing to this
or TH17 cells, or regulatory T (TREG) cells) upon antigen rigidity and to the presence of elevated levels of secretory
recognition. The production of IgA by Bcells, which IgA and antimicrobial peptides, the inner mucus layer
has a pivotal role in antimicrobial protection within is typically devoid of microorganisms43. Colonization by
the intestine, is mainly regulated by follicular helper T mucosal microbiota is more likely to occur in the outer

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 3



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

In addition to butyrate-producing Clostridia, other


Box 2 | Functions of the human gut microbiota
typical mucosal microorganisms also have effects on the
Digestion and energy harvesting from otherwise indigestible food components, by intestinal immune function of the host. For example,
modulation of the absorptive capacity of the intestinal epithelium and production of Bacteroides fragilis can produce polysaccharide A, which
enzymes that are absent in humans185 affects the immune system in the early stages of coloni
Supporting and priming the immune system zation. The immune system of germ-free mice is typically
Providing nutrients to the gut epithelium (such as short-chain fatty acids), thereby skewed towards a TH2cell-controlled humoral immune
exerting a trophic function and influencing the differentiation of gut epithelial cells186 response. Mono-association of germ-free mice with PSA-
Resistance against colonization by potential pathogens by filling in all the available producing B.fragilis or treatment with polysaccharide A
functional niches (that is, competition with the pathogen for physical space and stimulates the development of TH1cell-mediated immu
nutrients) and maximizing the functional capacity of the entire ecosystem187 nity53, which, in turn, protects against and suppresses
Influencing satiety through the production of short-chain fatty acids via fermentation inflammation54. Another example of a pronounced
of fibres, which can affect the release of gastrointestinal satiety hormones, such as immune-modulatory function of the microbiota is the
peptide YY and glucagon-like peptide188 ability of segmented filamentous bacteria (SFB) to direct
Gut bacteria are also thought to influence our state of mind by producing neuroactive the immune system of mice towards increased production
compounds that act on the brain189 of TH17 cells, thereby protecting the animals from infec
Brain:
tious agents55. Our research group also showed that SFB
Gut bacteria produce present during the neonatal colonization process in mice
neuroactive compounds are important microbial components in the development
of systemic autoimmunity in adult life56.

Regulation of systemic immunity


Immune system: The role of the microbiota in regulating host immune
Gastrointestinal tract:
Gut bacteria regulate responses is best emphasized in mice housed under
immune system germ-free conditions. Analysis of the innate and adap
Gut bacteria inuence development and
satiety, digestion and priming tive immune systems of germ-free mice revealed that a
resistance against colonization,
and provide nutrients to
microbiota-free environment is associated with reduced
the gut epithelium production of antimicrobial peptides, which results in
enhanced susceptibility to microbial infections. For
example, the microbiota contributes to the production
of the antimicrobial molecules REG3 and REG3 in
mucus layer, which is lessNature Reviews
rigid and | Rheumatology
contains lower levels mice57, and impaired production of these molecules is
of IgA and antimicrobial peptides43. Not unexpectedly, associated with increased susceptibility to infection with
studies on tissue samples obtained by gut biopsies or Listeria monocytogenes and Yersinia pseudotuberculosis,
analysis of the entire mucus layer have revealed the respectively58,59. The development of innate immune
mucosal microbiota to be fundamentally different from phagocytes such as neutrophils is also impaired in germ-
its luminal or faecal counterparts44,45. In addition to the free mice, which results in impaired host defence against
presence of higher levels of host immune effectors in the Escherichia coli K1 and Klebsiella pneumoniae60. In addi
mucosa than in the lumen or faeces46, this difference can tion, neonatal exposure to microorganisms limits the
be attributed to the ability of microorganisms to adhere expansion of invariant natural killer Tcells in the lung
to mucus47 and/or their ability to degrade mucins48. In and colon of specific-pathogen-free (SPF) mice, which
general, the mucosal microbiome is proportionally more makes the animals less susceptible to ovalbumin-induced
abundant in Firmicutes (especially Clostridium cluster allergic asthma than germ-free mice61. Observations in
XIVa) than in Bacteroidetes10,49. human monozygotic and dizygotic twins have shown
that non-heritable factors such as the microbiota have a
Regulation of intestinal immunity dramatic effect upon the immune system, which suggests
Certain microbiota, particularly butyrate-producing that the gut microbiota might contribute to much of the
Clostridia, can modulate TREGcells. Administration immune variation that is seen in humans62.
of butyrate-producing Clostridia such as F.prausnitzii
and Butyricicoccus pullicaecorum to animal models of Beyond the gut: the skin microbiota
inflammation seems to lower the occurrence of macro The presence of bacteria in the human body is not lim
scopic lesions to the intestinal mucosa50,51. Observations ited to the gut. In fact, the human skin the bodys
in model systems invitro, animal models and healthy largest organ hosts a high number of commensal bac
human individuals indicate that F.prausnitzii, in particu teria63. More than 1010 microbial cells cover the human
lar, resides preferentially close to the gut epithelium52. body and, as occurs for the GIT, different skin microen
This proximity might enable the delivery of butyrate vironments with distinct pH, temperature, moisture and
close to the epithelium, which could strengthen the gut sebum content are also characterized by different micro
barrier. Atarashi etal.4 demonstrated that mono-as bial groups. Metagenomic analyses have identified four
sociation of germ-free mice with butyrate-producing phyla of bacteria in the human skin64, which are mainly
Clostridia results in upregulation of FOXP3, which is lipophilic bacteria (including Propionibacterium spp.and
crucial for the function of TREG cells. Corynebacterium spp.) and commensal staphylococci.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

The long-established link between IBD and SpA


supports the role of the microbiota in human rheu
matic disease. Furthermore, human observational
studies have assessed whether the composition of the
faecal microbiome differs between patients and controls,
using sequencing of 16S ribosomal RNA (rRNA) genes,
whole-microbial genome sequencing and metabolomic
analysis. However, the results of these analyses do not
Pattern recognition reflect the composition of the luminal or mucosal micro
receptors present on System biomes. Also, no studies have addressed the risk of rheu
immune and epithelial cells ic im matic disease with regards to differences in the neonatal
in the gut recognize mu
Intest ni colonization of the gut by mode of delivery, breastfeeding
microbeassociated ina
l im
or antibiotic use in early childhood with the exception of

ty
molecular patterns present Muco
on the gut microbiota sal

m
a recent study which analysed the frequency of breast

un
Imm

ity
m
Gradients of microbiota u feeding in patients with AS versus controls70. Of inter

icr
ne
present in the gut lumen est, breastfeeding was found to be protective against AS

oen
reco ition
and mucosa create distinct Gut
development, further suggesting a potentially important

vironment
micro-environmental microbiota

gn
ecosystems in the gut role of gut microbiota in the onset of the disease.
Various mucosal microbiota
are known to regulate Evidence from animal studies
intestinal immune function Modulation of experimentally induced arthritis. Studies
of the host in mice with Tcell-activating genetic modifications (such
Activation of both the as SKG7173 and K/BN mice74) and in mice deficient for
innate and adaptive the IL1 receptor antagonist (IL1Ra)75 have shown that
immune systems by the gut arthritis disease activity is diminished under germ-free
microbiota in turn regulates
systemic immune responses or SPF conditions. Similarly, in rat models of adjuvant-
induced and streptococcal-cell-wall-induced arthritis
and in HLAB*27 transgenic rats, the disease pheno
type is modulated by housing7678. Monocolonization of
germ-free IL1Radeficient mice with the gut-residing
commensal bacterium Lactobacillus bifidus resulted in
arthritis with incidence and severity scores compara
Figure 1 | Relationship between gut microbiota and immune function of the host. ble to those observed in conventionally housed mice75.
Nature
Microbial ecosystems present within microenvironments Reviews
in the | Rheumatology
gut elicit intestinal Likewise, introduction of SFB in germ-free K/BN
immune responses following recognition of microbial antigens. The development of mice resulted in the onset of arthritis74. Furthermore,
intestinal immunity against gut microbiota subsequently shapes systemic immune the periodontal pathogens Porphyromonas gingivalis
responses of the host. and P.nigrescans markedly aggravate the severity of
arthritis in mice with collagen-induced arthritis (CIA)79.
Treatment with enrofloxacin, an antibiotic that targets
Gram-negative bacteria, exacerbated arthritis in the CIA
Under normal healthy conditions, the skin microbiota model80. Modulation of the gut microbiota with antibiotic
lives in homeostasis with the highly sophisticated treatment also influenced the development of arthritis in
immune system of the epidermis and the dermis, which the K/BN model74. In these mice, antibiotic treatment
comprises a network of epithelial cells, lymphocytes and disrupted the gut flora, which was associated with dimin
antigen-presenting cells. Changes in this balance and ished numbers of TH17 cells and attenuated arthritic
in the composition of the skin microbiota have been pathology74. The mechanisms underlying the differences
associated with the pathogenesis of psoriasis6569. in the responses to antibiotic treatment in the CIA and
K/BN models are not known, but could be related to
The microbiota in rheumatic disease differences in the microorganisms that are involved in
Different types of evidence link the microbiota to disease pathogenesis in each model.
rheumatic disease. In some studies of animal models In the humanized transgenic HLADRB1 mouse
of rheumatic disease the composition of the micro model, the composition of the faecal gut microbiota dif
biota was compared between diseased animals. These fered between mice carrying the arthritis susceptibility
comparisons have also been made in studies involving genetic variant (DRB1*0401) and genetically resistant
germ-free and gnotobiotic mice, in which animals were counterparts (HLADRB1*0402)81. This difference was
colonised with a defined microflora. Other studies have associated with increased gut permeability and differ
assessed the consequences of modulation of the micro ential transcription of TH17 regulatory network genes
flora through the use of antibiotics. Some of the limi in the jejunum81. Moreover, the faecal microbiome of
tations of these animal studies are their limited sample HLADRB1*0401 mice did not vary with sex and age,
sizes and the difficulty to mimic the complex multifactorial whereas HLADRB1*0402 mice had a sex-dependent
pathogenesis of human disease. and age-dependent microbiome. In HLAB*27 transgenic

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 5



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

rats, presence of the HLAB27 antigen was associated diversity of the gut microbiota differ between control
with an altered microbial composition of the gut82. An and diseased mice, and modulation of the gut micro
increase in the relative abundance of Prevotella spp. biome by treatment with vitaminA had a variable effect
and Bacteroides vulgatus and a decrease in the relative on lupus symptoms in different organs91. By lowering
abundance of Rikenellaceae was observed in HLAB*27 the pH of drinking water, Johnson etal.92 found that
transgenic animals when compared with controls82. (SWRNZB)-F1 (SNF1) mice developed nephritis at
Hoentjen etal.83 showed that the administration of pre a slower pace and had lower circulating levels of anti
biotics reduced the severity of colitis and prevented the nuclear antibodies (ANAs) than mice that were given
development of arthritis in these rats. This effect was water with a neutral pH. This intervention changed the
associated with alterations in the composition of the composition of the gut microbiota and modulated the
intestinal microbiota, decreased levels of proinflamma immune response in the gut mucosa92. Overall, these
tory cytokines and increased levels of immunomodu data suggest that the presence of the microbiota is not
latory molecules in the cecum and colon, respectively. required for disease onset and development in these
These studies suggest that HLA genes can have a role in models, but modulation of the intestinal flora influences
the microbial colonization of the gut and that dysbiosis disease progression.
might be present in genetically susceptible animals. Our research team identified an unexpected link
Some regulatory B (BREG) cell subsets that have a between the gut microbiota and development of ANAs56.
role in restraining excessive inflammation are depend Surprisingly, the appearance of ANAs in adult life was
ent on the microbiota. Following induction of arthritis, linked to neonatal colonization of the gut with commen
IL1 and IL6, which directly promote the differentia sal flora. The formation of ANAs was found in approx
tion of BREG cells and the production of IL10 by these imately 25% of lymphotoxin-deficient mice lacking
cells, were only produced in conventionally housed Peyers patches, cryptopatches and isolated lymphoid
mice, whereas antibiotic-treated mice did not pro follicles, all of which are components of normal GALT.
duce these cytokines84. Bcell-specific deletion of the Neonatal blockade of lymphotoxin, which affects the
genes encoding the IL6 receptor (IL6R) or the IL1 formation of isolated lymphoid follicles, resulted in the
receptor 1 (IL1R1) resulted in reduced numbers of occurrence of ANAs with variable specificities, includ
IL10producing Bcells and exacerbated arthritis84. The ing antibodies against the U1 ribonucleoprotein complex
gut microbiota was also shown to modulate arthritis in (U1RNP), SSA/Ro and DNA topoisomerase I. By con
experimental models of gout in which disease pathogen trast, blockade during fetal or adult life did not result
esis depends on IL1. Monosodium urate monohydrate in autoimmunity. Elimination of gut flora by treatment
crystals cause gout by activating the NLRP3 inflamma with broad-spectrum antibiotics or by blocking lympho
some, which leads to IL1 production and neutrophil toxin signalling under germ-free conditions reduced
recruitment. In germ-free mice, the production of short- ANA production. Furthermore, the gut microbiota dif
chain fatty acids that are necessary for adequate inflam fered between ANA-positive and ANA-negative animals,
masome assembly and IL1 production is decreased85. with particularly marked differences being observed
This effect depends on the metabolite-sensing receptor for Methylobacterium spp. belonging to the SFB group.
free fatty acid receptor 2 (also known as Gprotein cou Germ-free lymphotoxin-deficient animals monocolo
pled receptor 43)85. Overall, observations in some exper nized with SFB produced more ANAs than lymphotoxin-
imental models of arthritis show that microbial triggers deficient controls monocolonized with E.coli, which
by commensal bacteria are required to promote disease indicates that SFB can modulate the development of
in genetically predisposed animals. generalized autoimmunity in adult life.

Modulation of antinuclear antibody formation and Spondyloarthritis


systemic autoimmunity. The course of lupus-like dis Approximately 20% of patients with inflammatory bowel
ease and autoantibody production in mouse models of disease (IBD) develop arthritis in the joints of the lower
generalized autoimmunity are not profoundly different and upper limbs or inflammation of the spine, which
in animals raised under germ-free or under conven are features that characterize SpA93,94. The incidence of
tional housing conditions8689. However, no data on the these manifestations of musculoskeletal disease might
effects of elimination of gut microbiota with antibiotics be even higher in patients with Crohn disease and indi
are available in these models. Of note, these models, viduals with longstanding IBD95,96. AS, the prototypic
including NZB, MRL/lpr and pristane-induced lupus, form of SpA, occurs in up to 10% of patients with IBD97.
lack essential features of human disease, such as a bias The existence of a pathogenic link between the gut and
towards the female sex and stochastic distribution of joints is also apparent in enterogenic reactive arthritis, a
the probability of developing the disease. Nevertheless, disease in which an infectious enterocolitis triggers joint
indirect data suggest that gut flora might modulate dis inflammation98.
ease in these models. Studies have shown that caloric Importantly, ~50% of patients with SpA have micro
restriction or other dietary interventions, such as sup scopic signs of inflammation in the gut that are not
plementation with vitamins A, D or E, or with polyun associated with clinical gastrointestinal symptoms. This
saturated fatty acids, prevented progression of lupus-like inflammation can have the same histological character
disease in NZB/W mice90. In the MRL/lpr model, the istics as acute inflammation of the gut, in which neutro
most widely used of these models, the composition and phils predominate, in a way that resembles the response

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

to acute bacterial enterocolitis. However, one-third were found between patients and controls, but a higher
of patients with SpA have chronic microscopic inflam prevalence of sulfate-reducing bacteria was found in
mation that is characterized by a mononuclear inflamma patients with AS109. More recently, microbiome analy
tory infiltrate and structural remodelling of the intestinal ses in patients with SpA have been conducted using
mucosa, which resembles the histological characteristics high-throughput 16S rRNA gene sequencing. Costello
of Crohn disease98. Mucosal inflammation has a pro etal.110 found a higher abundance of Lachnospiraceae,
found effect on long-term outcomes in patients with Veillonellaceae, Prevotellaceae, Porphyromonadaceae
SpA. Most patients with normal histology or acute intes and Bacteroidaceae in the terminal ileum of patients
tinal lesions have transient arthritis. Clinical remission with AS than in healthy controls. Stoll etal.111 showed
of joint inflammation is associated with resolution of gut that F.prausnitzii abundance in the stools of patients
inflammation, whereas the presence of gut inflamma with enthesitis-related arthritis, a subtype of juve
tion is associated with persistent joint inflammation99,100. nile SpA, is reduced compared with that in controls,
Chronic inflammatory lesions in the gut are a risk fac whereas Bacteroides spp. and Akkermansia muciniphila
tor for development of Crohn disease; in one study, 20% were identified as disease-associated agents in subsets of
of patients with chronic intestinal lesions developed patients. Differences in the humoral responses to these
clinically overt IBD over a 5year period100. Even in the bacteria have been proposed to contribute to some
early stages of SpA, chronic microscopic gut inflam degree to the development of disease112.
mation is strongly associated with the extent of spinal SpA has also been linked with skin inflammation
inflammation101,102. through its association with psoriasis. Evidence showed
The intestinal microbiota has been extensively stud that 30% of patients with psoriasis develop PsA113,
ied in patients with IBD. A consistent finding of these although the origin of this pathogenic process is not
studies has been decreased diversity, that is, lower clear. Microbial and environmental factors could have
richness and evenness of the intestinal microbiota, in a role in this process, but a link between the micro
patients with IBD103. Richness refers to the variety of biota or the environment with PsA has not yet been
unique organisms present in a sample, whereas evenness established114. A study in patients with PsA or psoriasis
relates to the likelihood that two sequences chosen at reported lower frequencies of certain beneficial taxa,
random represent the same species. One study reported particularly Akkermansia spp. and Ruminococcus spp115.
on changes in the faeces of children with ulcerative colitis In patients with PsA, the frequency of these taxa was
when compared with faeces from healthy control children. lower than their frequency in patients with psoriasis.
Patients with ulcerative colitis had lower diversity of These observations are similar to earlier findings in
microbial species than controls; among the individuals patients with IBD, who also have different microbiome
with ulcerative colitis, those refractory to corticosteroid profiles to those of healthy individuals116. Further studies,
therapy had lower species counts than steroid-responsive however, are needed to fully understand the biological
patients104. Likewise, a study of children who were implications of these observations.
newly diagnosed with Crohn disease or ulcerative coli
tis showed a decreased microbiota diversity in patients Rheumatoid arthritis
with Crohn disease (but not in patients with ulcerative Susceptibility genes have been shown to have an impor
colitis) when compared with that in healthy controls105. tant role in the development of RA, but environmental
Substantial alterations in the taxonomic composition of factors are also likely to be critical in this process, as
the microbiota have also been reported in patients with inferred by the fairly low concordance rate of RA in
IBD. Although the findings of individual studies vary, monozygotic twins117. Development of anti-citrullinated
common observations include increases in the abun protein antibodies (ACPAs), which are highly specific
dance of Proteobacteria and decreased abundance of for RA, occurs well before disease onset and is linked to
Firmicutes, among which F.prausnitzii106. These differ environmental factors such as smoking117. This obser
ences are associated with functional consequences, such vation raised the idea that mucosal sites other than the
as a decreased production of short-chain fatty acids, gut might contribute to the pathogenesis of RA. Over the
which are essential for the health of enterocytes and also past few years, several studies highlighted that changes
have a direct anti-inflammatory effect107. in microbiota composition in the oral cavity, gut and
A strong association between bowel inflammation possibly the lung might facilitate the onset of RA.
and all forms of SpA (including AS, juvenile SpA and Several observational studies have found an associ
psoriatic arthritis (PsA)9195,108) has been recognized for ation between RA and periodontal disease. Periodontal
several decades, as well as the link between gut inflam disease occurs more frequently and tends to be more
mation and disease progression to AS and Crohn dis severe in patients with RA versus healthy controls118
ease. However, surprisingly little is currently known and new-onset RA has been linked with the presence
about the complex relationship between the micro of periodontal disease119. Moreover, treatment of perio
biota, genetics and inflammation in SpA. Few studies dontal disease decreases the severity of RA120. Much of
have directly addressed the role of the gut microbiota in the research on the relationship between the oral micro
this relationship. A 2002 study evaluated the composi biome and RA has been focused on P.gingivalis, because
tion of the faecal microflora of 15 patients with AS and these bacteria have the unique property of harbouring a
15 matched controls by denaturing gradient gel electro gene that encodes peptidyl arginine deiminase (PAD), an
phoresis. No specific differences in colonization profiles enzyme that catalyses the conversion of arginine residues

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 7



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

into citrulline121,122. This role is of particular interest Other rheumatic diseases


because citrullination of proteins (such as vimentin, Data on the putative link between the microbiota and
keratin and enolase) is associated with the production systemic autoimmune diseases other than SpA and RA
of ACPAs under certain conditions123126. Therefore, are limited in humans141,142. Environmental factors are
P.gingivalis could contribute to the induction of ACPAs, critical in the pathogenesis of SLE, the prototypic dis
a strong predictor of the onset of RA. This finding was ease for generalized autoimmunity, with development
supported by additional experimental work invitro and of autoantibodies preceding onset of clinical disease143.
invivo123126. Conversely, treatment of periodontal dis A 2015 study has pinpointed a potential role for the
ease might lower the levels of circulating ACPAs and microbiota in SLE via an effect of the microbiota in low
antibodies against P.gingivalis127. Finally, DNA of P.gin ering the threshold for activation of autoreactive Bcells56.
givalis and P.nigrescens was found in serum and synovial
fluid of patients with RA128. Unidirectional or multidirectional interplay?
In a small study involving low-throughput 16S rRNA How the microbiota, lifestyle and/or environmental
hybridization, Vaahtovuo etal.129 identified dysbiosis factors, and host-derived factors such as genetics and
in the faecal microbiota of patients with newly diag immune responses are interrelated is a key problem to
nosed RA, whereas dysbiosis was not present in the solve. Which factors are causes and which are conse
faecal microbiota of control patients with fibromyal quences? This chicken-oregg paradigm has not been
gia. Another study used PCR primers for Lactobacillus solved at present but some general principles have
DNA to show that the faecal microbiota of patients with emerged, especially from the IBD field.
RA contained increased diversity of Lactobacilli com The changes in the microbiota of patients with IBD
pared with the microbiota of healthy controls130. More do not directly prove these alterations are responsible for
recently, 16S rRNA sequencing showed a strong corre the inflammatory phenotype of these patients, as they
lation between disease and the presence of Prevotella could be secondary to the inflammatory process itself
copri and loss of Bacteroides in the faecal microbiome in or caused by therapy. However, even though some med
untreated patients with new-onset RA131. Finally, bacte ications used to treat IBD, particularly antibiotics, are
rial rRNA from naturally occurring commensals such as likely to alter the contents of the microbiota, alterations
Clostridium spp. have been isolated from synovial tissue in the microbiota were nevertheless identified in newly
of patients with RA but not patients with osteoarthritis diagnosed patients naive to immunosuppressive ther
or those with closed traumatic knee injuries132. apy105. A role for an altered microbiota in IBD is further
The relationship between the respiratory mucosa, substantiated by therapeutic responses to interventions
which harbours its own characteristic microbiota133135, that alter the microbiota. A meta-analysis of 35 stud
and RA is also receiving increasing attention. This inter ies involving patients with Crohn disease or ulcerative
relationship is complicated by the potential influence of colitis concluded that addition of probiotics to stand
cigarette smoking, which is associated with increased ard therapy in patients with ulcerative colitis, although
airway inflammation and, in some studies, has also not in those with Crohn disease, resulted in improved
been linked to changes in the microbiota in airways, remission rates144. Conversely, antibiotics can improve
particularly in the relative abundance of Prevotella and remission rates in patients with Crohn disease, but their
Porphyromonas136. The literature, however, is not entirely role in ulcerative colitis is less clear145.
consistent in this area133,134,136. One key observation that Given the reciprocity between bowel and joint inflam
makes the lung a particularly interesting site to examine mation in SpA, the intestinal microbiota could have a role
is the strong link between smoking, citrullination and in SpA as well. This role could take place either through
RA. Cigarette smoking leads to increased expression of a direct mechanism, whereby entire microorganisms or
PAD and protein citrullination in the bronchoalveolar their fragments traffic to the joints causing inflammation,
lavage cells of smokers without arthritis137. Additionally, or indirectly, whereby bacteria trigger an autoimmune
PAD polymorphisms, such as PADI4, have been shown reaction against self antigens by molecular mimicry or by
to predispose male smokers to developing RA138. ACPAs lowering the threshold of activation of autoreactive cells.
and rheumatoid factor have also been found in induced Alternatively, intestinal bacteria could skew the immune
sputa of patients with early-stage RA139. However, the system toward a proinflammatory state, which would ulti
relationship between smoking and autoimmunity in mately lead to joint inflammation. A proposed mechanism
RA might be more complex than previously thought, as to explain the link between gut and joint inflammation is
citrullination of vimentin, a major autoantigen in RA, the aberrant trafficking of lymphocytes, macrophages and
was found in lungs of both non-smokers and smokers innate lymphoid cells primed in the intestinal mucosa to
with chronic obstructive coronary disease (COPD), the joint146. The association of citrullination in the lung
which suggests that citrullination in the lungs of smokers and RA highlights that other mucosal surfaces could also
is mainly due to inflammation140. Altogether, available be important. In addition, the development of autoim
evidence indicates that the lung might be an early site of munity in adult life could be a consequence of neonatal
autoimmune-related injury and a site of generation of changes in the hostmicroorganism interplay.
RArelated autoimmunity. Thus, generation of RArelated We propose two possible models that could explain
autoimmunity could be triggered or at least modulated by the relationship between the microbiota and the host.
the presence of proinflammatory microbiota derived from In the linear or unidirectional model a primary cause ini
periodontal tissue. tiates a one-way directional process. For example, disease

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Prenatal immunity Postnatal immunity Autoimmunity

Lumen

Epithelial cell M cell Mucus Goblet cell

Immature
Peyers patch Peyers patch

LT-12 LT-12
LT receptor LT receptor

Lamina Cryptopatch Immature isolated


propria lymphoid follicle
Mature isolated lymphoid follicle

Mesenteric IL-17
lymph node
IL-17
Dendritic cell IgA+ plasma cell
ROR+ LTi cell Commensal bacterium
Antinuclear
B cell SFB antibodies
T cell Dimeric IgA

Figure 2 | Unidirectional hostmicroorganism interplay. An example in which antinuclear antibodies are induced
by lymphotoxin deficiency is shown. In the postnatal phase, the intestinal lumen is colonized by microbiota.
Nature Formation
Reviews | Rheumatology
of secondary lymphoid organs depends on the TNF cytokine lymphotoxin 12 (LT-12) and its interaction with the
lymphotoxin (LT) receptor. In the neonatal phase, LT-12 is indispensable for formation of isolated lymphoid follicles.
Ablation of LT-12 in the neonatal period, but not during fetal or adult life, is associated with absence of formation of
isolated lymphoid follicles and with microbiota shifts characterized by expansion of segmented filamentous bacteria
(SFB). SFB are strong inducers of IL17driven responses that promote spontaneous production of antinuclear antibodies.
Thus, neonatal colonization of the intestine by SFB in the absence of gut-associated lymphoid tissues is a predisposing
factor for production of antinuclear antibodies in adult life. Adapted from Van Praet, J. T. etal. Commensal microbiota
influence systemic autoimmune responses. EMBO J. 34, 466474 (2015) and Knoop, K. A. & Newberry, R. D. Isolated
lymphoid follicles are dynamic reservoirs for the induction of intestinal IgA. Front. Immunol 3, 84 (2012)56,190

could be induced in an individual with a particular formation of isolated lymphoid follicles, along with an
genetic background following an environmental trig expansion of SFB56. Similarly, the secretion of lympho
ger (FIG.2). This occurrence would in turn result in toxin by intestinal innate lymphoid cells has been shown
changes in the microbiota, which could influence local to regulate the production of IgA and microbiota com
immune responses in the intestinal tract, for example position in the gut of adult animals147, as does TLR5 or
by modulation of the formation and maturation of NLRP6 deficiency in mice148,149. These findings suggest
GALT. These events would lead to changes in systemic that the gut microbiota can be shaped by the genetic
immune responses, loss of tolerance and occurrence of background in a feed-forward manner. Genetics also
systemic autoimmunity. An example to which such a seems to have a critical role in shaping the gut micro
model applies is the induction of ANAs, including anti biota in humans150. In a large study of twins, many
bodies against U1RNP, SSA/Ro and DNA topoisomerase microbial taxa were influenced by the genetics of the
I, following neonatal depletion of the TNF superfamily host, particularly the family Christensenellaceae, which
cytokine lymphotoxin56 (FIG.2). This depletion ablates formed a cooccurrence network with other heritable

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 9



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

bacteria and with methanogenic archaea. Interestingly, Lifestyle changes (such as smoking cessation in RA)
this network was associated with metabolic phenotypes. might be useful yet difficult to implement in a large
If the linear model is accurate, its unidirectionality could proportion of patients. Furthermore, although gene
have important consequences in the ability of therapy to environment interactions are involved in the onset of
restore balance in the composition of the microbiota. disease, evidence suggests that environmental factors
An alternative model is the multidirectional model, important in rheumatic diseases can have an effect years
in which the interrelationship between genetics, the before clinical disease becomes apparent160.
microbiota, environment and immune responses is not Specific emphasis can be placed on the immune-
unidirectional and is more plastic (FIG.3). Here, the host modulating potential of several gut microorganisms, such
microorganism interplay reaches a steady state that is as Bacteroides fragilis strains or members of Clostridium
dependent on an equilibrium achieved between the dif clusters IV and XIVa, which may modulate PsA. Although
ferent variables. In some diseases (such as AS) the genetic most of the latter organisms (that is, members of
component is the major component of disease, yet the Clostridium clusters IV and XIVa) have butyrate produc
occurrence of clinical onset relies on environmental, tion as a core function, their immune-modulating effect
lifestyle (for example, diet, smoking status) or microbial can also be derived from compounds in their secretome or
changes. In other instances (such as reactive arthritis), other antigens. This area of research needs further inves
the genetic component can be much less prominent, with tigation, but administration of these microorganisms to
the microbial component being the dominating trigger. animal models has already yielded promising results161.
In RA, established risk factors, including smoking status The outcomes of the Human Microbiome Project1
and sex, influence the gut microbiota, with the effect of and MetaHIT2 have confirmed that the resident micro
smoking on ACPA production being particularly obvi biota has a crucial role in maintaining the health of the
ous in patients carrying HLADRB1 shared-epitope host. Conversely, dysbiosis is associated with potential
alleles137,151155. The principal difference between the health problems. Therefore, biotherapeutic strategies
multidirectional and the linear models is the reversibil designed to preserve or restore the human microbiota
ity of the multidirectional model. Lack of equilibrium might be an interesting approach for the treatment of
between the four main components of the model (genet rheumatic diseases.
ics, microbiota, environment and immune response) In the past 20years, much research effort has been
results in onset of disease. Reestablishing homeosta focused on the modulation of the colonic microbiota and
sis could, from a conceptual viewpoint, be achieved by related metabolic processes by prebiotics and probiotics
targeting either of these components. to improve the health of the host162166. The prebiotic
Substantial evidence indicates that some of the concept has been defined as the selective stimulation of
observed relationships between the host genetics, the growth and/or activity(ies) of one or a limited number of
environment and the microbiota are species-dependent. microbial genus(era)/species in the gut microbiota that
Christensenellaceae are highly heritable, whereas the confer(s) health benefits to the host (REF.167). The effects
Bacterioidetes community is mostly shaped by envi of nutrients with prebiotic properties on gut health have
ronmental factors and most of the species in this group received particular attention. Interestingly, improvements
are not heritable. Members of the Bacterioidetes have in gut function have been associated with systemic effects
been shown to respond to dietary interventions156,157. on the host physiology. Dietary fructans and arabinoxy
Modulation of the composition of the gut microbiota lans have been shown to decrease fat mass, steatosis, lipae
by food components might also modulate inflamma mia, hyperglycaemia, gut permeability, and intestinal and
tory responses in the gut. Emulsifiers (such as carboxy systemic inflammation24,167169. The concept of prebiotics
methylcellulose and polysorbate80) are detergent-like was mostly based on the fact that inulin-type fructans
molecules that are ubiquitous components of processed were able to increase the abundance of bifidobacteria in
foods. These substances can increase bacterial transport the gut. However, the relevance of the bifidogenic effect
across epithelia by affecting mucusbacterial inter perse is debatable, as more than 100 bacterial species
actions. Intake of emulsifiers promoted ulcerative colitis are modulated when a prebiotic approach is used and
in mice predisposed to this disorder and was associated several species (Roseburia spp., A.muciniphila, F.praus
with low-grade inflammation and obesity and/or the nitzii, etc.) are associated with improvements in host
metabolic syndrome in wild-type hosts158. health168,169. Although the prebiotic approach is attrac
tive, as it concerns the dietary modulation of indigenous
How can homeostasis be restored? intestinal microorganisms that are already adapted to the
Genetics is the most complex component of the host host170, it has been primarily used so far in a preventive
microbiota relationship to modulate. Proofofprinciple manner. However, some therapeutic applications for this
application of gene therapy for rheumatic diseases has approach have been suggested. A severely disrupted gut
been done in animal models, but clinical practice appli microbiota would benefit from either the introduction
cation is still a long way ahead159. The modulation of of key microbial species, such as Lactobacillus casei 01
environmental factors, the modulation of the immune in patients with RA171,172, or the introduction of a general
response (an approach our group is currently investi microbiota population, as has been reported for severe
gating), and targeting of the microbiota are more obvi dysbiosis in cases of recurrent C.difficile infection173.
ous strategies for influencing the hostmicroorganism The efficacy and safety of such approaches, however,
interplay. need to be confirmed in controlled trials

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Systemic sclerosis

Genetics

In
tis
yli

am
nd Microbiome Immunity

ma
Environmental
spo

tor
factors
ing

y bo
ylos

wel
Ank

Microbiome

disea
Genetics

se
Immunity

Immunity

Microbiome

Immunity

Genetics

Microbiome
Rh
eu
ma
toid
arth
ritis

Figure 3 | Multidirectional model of hostmicroorganism interplay. In this model, the interaction between host
genetics, environmental factors, the microbiota and immunity is plastic. Lack of balance between these factors
contributes to onset of disease, but the relative contribution of each of these four components varies from disease
Nature Reviews | Rheumatology
to disease. For example, the genetic contribution to disease in ankylosing spondylitis is more prominent than that
in rheumatoid arthritis or inflammatory bowel disease.

A systematic review of the data available on the use with risks of transmission of other diseases, which raises
of probiotics in a clinical context showed that the pro questions over the widespread applicability of FMT in
biotics with the greatest ability to restore the microbiota less acute and/or life-threatening pathologies177. An inter
have the strongest clinical efficacy174. As the gut micro mediate solution between use of single bacterial strains
biota is a complex network of species interacting with and FMT is the use of synthetic (probiotic) consortia
the host, such a restoration could be accomplished by that include the major taxa of gut bacteria or have the
acting on single or multiple nodes of the network, at a most represented functionalities178. An example of this
compositional and/or at a functional level175. The sim approach is the use of a synthetic mixture of micro
plest application of this concept is the faecal material organisms, isolated from an individual on the basis of
transplant (FMT), in which faecal microbial slurry is their culturability, as a therapeutic agent to cure CDAD179.
transferred from a healthy donor to a diseased recipient, Another example is the potential use of a 17strain mix
thereby replacing the whole dysbiotic microbiota of the of Clostridia for treatment of inflammatory diseases161.
recipient176. This form of bacteriotherapy has been mostly At the nutritional level, enteral nutrition has been shown
used to treat antibiotic-resistant infections (such as to have similar efficacy in treating juvenile IBD to that
C.difficile-associated diarrhoea [CDAD]). Yet, the poorly of conventional corticosteroids180. Yet, proof of efficacy
characterized nature of faecal transplants is associated and production of pharmaceutical-grade products will

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 11



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

require a big effort in the future, starting from the precise of particular interest in RA and in PsA, respectively.
determination of a causative link between changes in The influence of the microbiota on rheumatic disease is
the composition and activity of the microbiota and the also time-dependent under certain conditions, for example
development of a given pathology175. during the neonatal period.
An even more futuristic approach to favour human At present, management of the microbiota seems
health by managing the metabolic interactions between unlikely to become a self-standing solution for therapy
the microbiota and the host is the engineering of spe of rheumatic diseases. However, when dealing with a
cific bacterial strains. This approach could be used to multifactorial disease, a multifactorial approach to ther
enhance drug therapies, to produce metabolites with apy could be a useful strategy at certain stages of dis
drug-like activities, to rewire smart bacteria to switch ease. Among other factors (genetics, immunity and the
on and off the production of anti-inflammatory mole environment), the modulation of the microbiota and its
cules or to correct abnormalities in signalling pathways interaction with the host might be a strategy to control
involved in disease pathogenesis181,182. or prevent rheumatic diseases.
Whether changes in lifestyle practices, such as hygiene,
Conclusions antibiotic use, diet or smoking status, which all influence
Increasing evidence shows that the microbiota influ various microorganisms in multiple manners, contribute
ences a wide spectrum of rheumatic diseases, including to the onset or to the severity and evolution of rheumatic
SpA, PsA and RA. Of interest, the critical sites where diseases is yet an unexplored question. Research into the
hostmicroorganism interactions modulate inflam mechanisms whereby therapeutically altered micro
matory responses might not be identical in each of organisms could modulate rheumatic diseases, for example
these diseases. The gut (ileum and colon) is the most by activating immune regulatory cells, is still in its infancy,
studied site in SpA, whereas the lung and the skin are yet it is an interesting avenue to pursue.

1. Turnbaugh,P.J. etal. The human microbiome project. 16. Louis,P. & Flint,H.J. Diversity, metabolism and 31. Thavagnanam,S., Fleming,J., Bromley,A.,
Nature 449, 804810 (2007). microbial ecology of butyrate-producing bacteria from Shields,M.D. & Cardwell,C.R. A meta-analysis of
2. Qin,J. etal. A human gut microbial gene catalogue the human large intestine. FEMS Microbiol. Lett. 294, the association between Caesarean section and
established by metagenomic sequencing. Nature 464, 18 (2009). childhood asthma. Clin. Exp. Allergy 38, 629633
5965 (2010). 17. Turnbaugh,P.J. etal. A core gut microbiome in obese (2008).
3. Karlsson,F.H., Nookaew,I., Petranovic,D. & and lean twins. Nature 457, 480484 (2009). 32. Decker,E. etal. Cesarean delivery is associated with
Nielsen,J. Prospects for systems biology and 18. Verberkmoes,N.C. etal. Shotgun metaproteomics of celiac disease but not inflammatory bowel disease in
modeling of the gut microbiome. Trends Biotechnol. the human distal gut microbiota. ISME J. 3, 179189 children. Pediatrics 125, e1433e1440 (2010).
29, 251258 (2011). (2009). 33. Kull,I., Almqvist,C., Lilja,G., Pershagen,G. &
4. Atarashi,K. etal. Induction of colonic regulatory 19. Kolmeder,C.A. etal. Comparative metaproteomics Wickman,M. Breast-feeding reduces the risk of
Tcells by indigenous Clostridium species. Science and diversity analysis of human intestinal microbiota asthma during the first 4years of life. J.Allergy Clin.
331, 337341 (2011). testifies for its temporal stability and expression of Immunol. 114, 755760 (2004).
5. Moghaddami,M., Cummins,A. & Mayrhofer,G. core functions. PLoS ONE 7, e29913 (2012). 34. Akobeng,A.K., Ramanan,A.V., Buchan,I. &
Lymphocyte-filled villi: comparison with other 20. Koek,M.M., Jellema,R.H., van der Greef,J., Heller,R.F. Effect of breast feeding on risk of coeliac
lymphoid aggregations in the mucosa of the human Tas,A.C. & Hankemeier,T. Quantitative metabolomics disease: a systematic review and meta-analysis of
small intestine. Gastroenterology 115, 14141425 based on gas chromatography mass spectrometry: observational studies. Arch. Dis. Child. 91, 3943
(1998). status and perspectives. Metabolomics 7, 307328 (2006).
6. Pabst,O. etal. Cryptopatches and isolated lymphoid (2011). 35. Shaw,S.Y., Blanchard,J.F. & Bernstein,C.N.
follicles: dynamic lymphoid tissues dispensable 21. Tuohy,K.M. etal. Studying the human gut microbiota Association between the use of antibiotics in the first
for the generation of intraepithelial lymphocytes. in the trans-omics era focus on metagenomics and year of life and pediatric inflammatory bowel disease.
Eur. J.Immunol. 35, 98107 (2005). metabonomics. Curr. Pharm. Des. 15, 14151427 Am. J.Gastroenterol. 105, 26872692 (2010).
7. Lugering,A. etal. CCR6 identifies lymphoid tissue (2009). 36. Faria,A.M. & Weiner,H.L. Oral tolerance. Immunol.
inducer cells within cryptopatches. Clin. Exp. Immunol. 22. Lin,H.M., Helsby,N.A., Rowan,D.D. & Rev. 206, 232259 (2005).
160, 440449 (2010). Ferguson,L.R. Using metabolomic analysis to 37. Mowat,A.M. & Agace,W.W. Regional specialization
8. Claesson,M.J. etal. Comparative analysis of understand inflammatory bowel diseases. Inflamm. within the intestinal immune system. Nat. Rev.
pyrosequencing and a phylogenetic microarray for Bowel Dis. 17, 10211029 (2011). Immunol. 14, 667685 (2014).
exploring microbial community structures in the 23. Helander,H.F. & Fandriks,L. Surface area of the 38. Akira,S., Uematsu,S. & Takeuchi,O. Pathogen
human distal intestine. PLoS ONE 4, e6669 digestive tract revisited. Scand. J.Gastroenterol. recognition and innate immunity. Cell 124, 783801
(2009). 49, 681689 (2014). (2006).
9. Sankar,S.A., Lagier,J.C., Pontarotti,P., Raoult,D. 24. Delzenne,N.M., Neyrinck,A.M., Backhed,F. & 39. Fagarasan,S., Kawamoto,S., Kanagawa,O. &
& Fournier,P.E. The human gut microbiome, a Cani,P.D. Targeting gut microbiota in obesity: effects Suzuki,K. Adaptive immune regulation in the gut:
taxonomic conundrum. Syst. Appl. Microbiol. 38, of prebiotics and probiotics. Nat. Rev. Endocrinol. 7, Tcell-dependent and Tcell-independent IgA
276286 (2015). 639646 (2011). synthesis. Annu. Rev. Immunol. 28, 243273
10. Eckburg,P.B. etal. Diversity of the human 25. Cho,J.H. The genetics and immunopathogenesis of (2010).
intestinal microbial flora. Science 308, 16351638 inflammatory bowel disease. Nat. Rev. Immunol. 8, 40. Tsuji,M. etal. Requirement for lymphoid tissue-
(2005). 458466 (2008). inducer cells in isolated follicle formation and
11. Lozupone,C.A., Stombaugh,J.I., Gordon,J.I., 26. Ogura,Y. etal. Expression of NOD2 in Paneth cells: Tcell-independent immunoglobulin A generation
Jansson,J.K. & Knight,R. Diversity, stability and a possible link to Crohns ileitis. Gut 52, 15911597 in the gut. Immunity 29, 261271 (2008).
resilience of the human gut microbiota. Nature 489, (2003). 41. Fagarasan,S. & Honjo,T. Intestinal IgA synthesis:
220230 (2012). 27. De Filippo,C. etal. Impact of diet in shaping gut regulation of front-line body defences. Nat. Rev.
12. Ley,R.E. The genemicrobe link. Nature 518, S7 microbiota revealed by a comparative study in children Immunol. 3, 6372 (2003).
(2015). from Europe and rural Africa. Proc. Natl Acad. Sci. USA 42. Sonnenberg,G.F. & Artis,D. Innate lymphoid
13. Velasquez-Manoff,M. Gut microbiome: the 107, 1469114696 (2010). cells in the initiation, regulation and resolution of
peacekeepers. Nature 518, S3S11 (2015). 28. Round,J.L. & Mazmanian,S.K. The gut microbiota inflammation. Nat. Med. 21, 698708 (2015).
14. Mirande,C. etal. Dietary fibre degradation and shapes intestinal immune responses during health and 43. Johansson,M.E. etal. The inner of the two Muc2
fermentation by two xylanolytic bacteria Bacteroides disease. Nat. Rev. Immunol. 9, 313323 (2009). mucin-dependent mucus layers in colon is
xylanisolvens XB1AT and Roseburia intestinalis 29. Delzenne,N.M. & Cani,P.D. Interaction between devoid of bacteria. Proc. Natl Acad. Sci. USA 105,
XB6B4 from the human intestine. J.Appl. Microbiol. obesity and the gut microbiota: relevance in nutrition. 1506415069 (2008).
109, 451460 (2010). Annu. Rev. Nutr. 31, 1531 (2011). 44. Zoetendal,E.G. etal. Mucosa-associated bacteria
15. Ze,X., Duncan,S.H., Louis,P. & Flint,H.J. 30. Algert,C.S., McElduff,A., Morris,J.M. & in the human gastrointestinal tract are uniformly
Ruminococcus bromii is a keystone species for the Roberts,C.L. Perinatal risk factors for early onset distributed along the colon and differ from the
degradation of resistant starch in the human colon. of type1 diabetes in a 20002005 birth cohort. community recovered from feces. Appl. Environ.
ISME J. 6, 15351543 (2012). Diabet. Med. 26, 11931197 (2009). Microbiol. 68, 34013407 (2002).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

45. Lepage,P. etal. Biodiversity of the mucosa-associated 70. Montoya,J. etal. Patients with ankylosing spondylitis 94. Salvarani,C. etal. Musculoskeletal manifestations
microbiota is stable along the distal digestive tract in have been breast fed less often than healthy controls: in a population-based cohort of inflammatory bowel
healthy individuals and patients with IBD. Inflamm. a casecontrol retrospective study. Ann. Rheum. Dis. disease patients. Scand. J.Gastroenterol. 36,
Bowel Dis. 11, 473480 (2005). 75, 879882 (2016). 13071313 (2001).
46. LievinLe Moal,V. & Servin,A.L. The front line of 71. Hida,S., Miura,N.N., Adachi,Y. & Ohno,N. Cell wall 95. Veloso,F.T., Carvalho,J. & Magro,F. Immune-related
enteric host defense against unwelcome intrusion of -glucan derived from Candida albicans acts as a systemic manifestations of inflammatory bowel
harmful microorganisms: mucins, antimicrobial trigger for autoimmune arthritis in SKG mice. Biol. disease. A prospective study of 792 patients. J.Clin.
peptides, and microbiota. Clin. Microbiol. Rev. 19, Pharm. Bull. 30, 15891592 (2007). Gastroenterol. 23, 2934 (1996).
315337 (2006). 72. Baillet,A.C. etal. High Chlamydia burden promotes 96. Ditisheim,S. etal. Inflammatory articular disease in
47. Roos,S. & Jonsson,H. A high-molecular-mass cell- tumor necrosis factor-dependent reactive arthritis in patients with inflammatory bowel disease: result of
surface protein from Lactobacillus reuteri 1063 SKG mice. Arthritis Rheumatol. 67, 15351547 the Swiss IBD Cohort Study. Inflamm. Bowel Dis. 21,
adheres to mucus components. Microbiology 148, (2015). 25982604 (2015).
433442 (2002). 73. Benham,H. etal. Interleukin23 mediates the 97. Orchard,T.R. etal. The prevalence, clinical features
48. Derrien,M., Vaughan,E.E., Plugge,C.M. & de intestinal response to microbial 1,3glucan and the and association of HLAB27 in sacroiliitis associated
Vos,W.M. Akkermansia muciniphila gen. nov., sp. development of spondyloarthritis pathology in SKG with established Crohns disease. Aliment. Pharmacol.
nov., a human intestinal mucin-degrading bacterium. mice. Arthritis Rheumatol. 66, 17551767 (2014). Ther. 29, 193197 (2009).
Int. J.Syst. Evol. Microbiol. 54, 14691476 (2004). 74. Wu,H.J. etal. Gut-residing segmented filamentous 98. Jacques,P. & Elewaut,D. Joint expedition: linking
49. Frank,D.N. etal. Molecular-phylogenetic bacteria drive autoimmune arthritis via Thelper 17 gut inflammation to arthritis. Mucosal Immunol. 1,
characterization of microbial community imbalances in cells. Immunity 32, 815827 (2010). 364371 (2008).
human inflammatory bowel diseases. Proc. Natl Acad. 75. Abdollahi-Roodsaz,S. etal. Stimulation of TLR2 99. Cuvelier,C. etal. Histopathology of intestinal
Sci. USA 104, 1378013785 (2007). and TLR4 differentially skews the balance of Tcells inflammation related to reactive arthritis. Gut 28,
50. Sokol,H. etal. Faecalibacterium prausnitzii is an in a mouse model of arthritis. J.Clin. Invest. 118, 394401 (1987).
anti-inflammatory commensal bacterium identified by 205216 (2008). 100. Mielants,H. etal. The evolution of
gut microbiota analysis of Crohn disease patients. 76. Ginsburg,I. Can chronic and self-perpetuating arthritis spondyloarthropathies in relation to gut histology.
Proc. Natl Acad. Sci. USA 105, 1673116736 (2008). in the human be caused by arthrotropic undegraded III. Relation between gut and joint. J.Rheumatol.
51. Eeckhaut,V. etal. Butyricicoccus pullicaecorum in microbial cell wall constituants? A working hypothesis. 22, 22792284 (1995).
inflammatory bowel disease. Gut 62, 17451752 Rheumatol. Rehabil. 16, 141149 (1977). 101. Van Praet,L. etal. Degree of bone marrow
(2013). 77. Taurog,J.D. etal. The germfree state prevents oedema in sacroiliac joints of patients with axial
52. Khan,M.T. etal. The gut anaerobe development of gut and joint inflammatory disease spondyloarthritis is linked to gut inflammation
Faecalibacterium prausnitzii uses an extracellular in HLAB27 transgenic rats. J.Exp. Med. 180, and male sex: results from the GIANT cohort.
electron shuttle to grow at oxic-anoxic interphases. 23592364 (1994). Ann. Rheum. Dis. 73, 11861189 (2014).
ISME J. 6, 15781585 (2012). 78. Khare,S.D., Luthra,H.S. & David,C.S. 102. Van Praet,L. etal. Microscopic gut inflammation in
53. Mazmanian,S.K., Liu,C.H., Tzianabos,A.O. & Spontaneous inflammatory arthritis in HLAB27 axial spondyloarthritis: a multiparametric predictive
Kasper,D.L. An immunomodulatory molecule of transgenic mice lacking 2microglobulin: a model of model. Ann. Rheum. Dis. 72, 414417 (2013).
symbiotic bacteria directs maturation of the host human spondyloarthropathies. J.Exp. Med. 182, 103. Nagalingam,N.A. & Lynch,S.V. Role of the
immune system. Cell 122, 107118 (2005). 11531158 (1995). microbiota in inflammatory bowel diseases. Inflamm.
54. Mazmanian,S.K., Round,J.L. & Kasper,D.L. 79. Marchesan,J.T. etal. Porphyromonas gingivalis oral Bowel Dis. 18, 968984 (2012).
A microbial symbiosis factor prevents intestinal infection exacerbates the development and severity of 104. Michail,S. etal. Alterations in the gut microbiome
inflammatory disease. Nature 453, 620625 (2008). collagen-induced arthritis. Arthritis Res. Ther. 15, of children with severe ulcerative colitis. Inflamm.
55. Ivanov,I.I. etal. Induction of intestinal Th17 cells by R186 (2013). Bowel Dis. 18, 17991808 (2012).
segmented filamentous bacteria. Cell 139, 485498 80. Dorozynska,I., Majewska-Szczepanik,M., 105. Hansen,R. etal. Microbiota of denovo pediatric IBD:
(2009). Marcinska,K. & Szczepanik,M. Partial depletion of increased Faecalibacterium prausnitzii and reduced
56. Van Praet,J.T. etal. Commensal microbiota influence natural gut flora by antibiotic aggravates collagen bacterial diversity in Crohns but not in ulcerative
systemic autoimmune responses. EMBO J. 34, induced arthritis (CIA) in mice. Pharmacol. Rep. 66, colitis. Am. J.Gastroenterol. 107, 19131922
466474 (2015). 250255 (2014). (2012).
57. Cash,H.L., Whitham,C.V., Behrendt,C.L. & 81. Gomez,A. etal. Loss of sex and age driven differences 106. Huttenhower,C., Kostic,A.D. & Xavier,R.J.
Hooper,L.V. Symbiotic bacteria direct expression in the gut microbiome characterize arthritis- Inflammatory bowel disease as a model for translating
of an intestinal bactericidal lectin. Science 313, susceptible *0401 mice but not arthritis-resistant the microbiome. Immunity 40, 843854 (2014).
11261130 (2006). *0402 mice. PLoS ONE 7, e36095 (2012). 107. Segain,J.P. etal. Butyrate inhibits inflammatory
58. Brandl,K., Plitas,G., Schnabl,B., DeMatteo,R.P. 82. Lin,P. etal. HLAB27 and human 2microglobulin responses through NFB inhibition: implications for
& Pamer,E.G. MyD88mediated signals induce the affect the gut microbiota of transgenic rats. PLoS ONE Crohns disease. Gut 47, 397403 (2000).
bactericidal lectin RegIII and protect mice against 9, e105684 (2014). 108. Schatteman,L. etal. Gut inflammation in psoriatic
intestinal Listeria monocytogenes infection. 83. Hoentjen,F. etal. Reduction of colitis by prebiotics in arthritis: a prospective ileocolonoscopic study.
J.Exp. Med. 204, 18911900 (2007). HLAB27 transgenic rats is associated with microflora J.Rheumatol. 22, 680683 (1995).
59. Dessein,R. etal. Toll-like receptor 2 is critical for changes and immunomodulation. Inflamm. Bowel Dis. 109. Stebbings,S. etal. Comparison of the faecal
induction of Reg3 expression and intestinal clearance of 11, 977985 (2005). microflora of patients with ankylosing spondylitis and
Yersinia pseudotuberculosis. Gut 58, 771776 (2009). 84. Rosser,E.C. etal. Regulatory Bcells are induced by controls using molecular methods of analysis.
60. Deshmukh,H.S. etal. The microbiota regulates gut microbiota-driven interleukin1 and interleukin6 Rheumatology (Oxford) 41, 13951401 (2002).
neutrophil homeostasis and host resistance to production. Nat. Med. 20, 13341339 (2014). 110. Costello,M.E. etal. Intestinal dysbiosis in ankylosing
Escherichia coli K1 sepsis in neonatal mice. Nat. Med. 85. Vieira,A.T. etal. A role for gut microbiota and the spondylitis. Arthritis Rheumatol. http://dx.doi.
20, 524530 (2014). metabolite-sensing receptor GPR43 in a murine org/10.1002/art.38967 (2014).
61. Olszak,T. etal. Microbial exposure during early life model of gout. Arthritis Rheumatol. 67, 16461656 111. Stoll,M.L. etal. Altered microbiota associated with
has persistent effects on natural killer Tcell function. (2015). abnormal humoral immune responses to commensal
Science 336, 489493 (2012). 86. Mizutani,A. etal. Pristane-induced autoimmunity in organisms in enthesitis-related arthritis. Arthritis Res.
62. Brodin,P. etal. Variation in the human immune germ-free mice. Clin. Immunol. 114, 110118 (2005). Ther. 16, 486 (2014).
system is largely driven by non-heritable influences. 87. Maldonado,M.A. etal. The role of environmental 112. Stebbings,S.M., Taylor,C., Tannock,G.W.,
Cell 160, 3747 (2015). antigens in the spontaneous development of Baird,M.A. & Highton,J. The immune response to
63. Belkaid,Y. & Segre,J.A. Dialogue between skin autoimmunity in MRL-lpr mice. J.Immunol. 162, autologous bacteroides in ankylosing spondylitis is
microbiota and immunity. Science 346, 954959 63226330 (1999). characterized by reduced interleukin 10 production.
(2014). 88. East,J., Prosser,P.R., Holborow,E.J. & Jaquet,H. J.Rheumatol. 36, 797800 (2009).
64. Grice,E.A. & Segre,J.A. The skin microbiome. Autoimmune reactions and virus-like particles in 113. Gao,Z., Tseng,C.H., Strober,B.E., Pei,Z. &
Nat. Rev. Microbiol. 9, 244253 (2011). germ-free NZB mice. Lancet 1, 755757 (1967). Blaser,M.J. Substantial alterations of the cutaneous
65. Takemoto,A., Cho,O., Morohoshi,Y., Sugita,T. & 89. Fernandes,G., Friend,P., Yunis,E.J. & Good,R.A. bacterial biota in psoriatic lesions. PLoS ONE 3,
Muto,M. Molecular characterization of the skin fungal Influence of dietary restriction on immunologic e2719 (2008).
microbiome in patients with psoriasis. J.Dermatol. function and renal disease in (NZB NZW) F1 mice. 114. Eppinga,H., Konstantinov,S.R., Peppelenbosch,M.P.
42, 166170 (2015). Proc. Natl Acad. Sci. USA 75, 15001504 (1978). & Thio,H.B. The microbiome and psoriatic arthritis.
66. Jagielski,T. etal. Distribution of Malassezia species 90. Hsieh,C.C. & Lin,B.F. Dietary factors regulate Curr. Rheumatol. Rep. 16, 407 (2014).
on the skin of patients with atopic dermatitis, cytokines in murine models of systemic lupus 115. Scher,J.U. etal. Decreased bacterial diversity
psoriasis, and healthy volunteers assessed by erythematosus. Autoimmun. Rev. 11, 2227 (2011). characterizes the altered gut microbiota in patients
conventional and molecular identification methods. 91. Liao,X. etal. Paradoxical effects of all-trans-retinoic with psoriatic arthritis, resembling dysbiosis in
BMC Dermatol. 14, 3 (2014). acid on lupus-like disease in the MRL/lpr mouse inflammatory bowel disease. Arthritis Rheumatol.
67. Alekseyenko,A.V. etal. Community differentiation of model. PLoS ONE 10, e0118176 (2015). 67, 128139 (2015).
the cutaneous microbiota in psoriasis. Microbiome 1, 92. Johnson,B.M., Gaudreau,M.C., AlGadban,M.M., 116. Manichanh,C. etal. Reduced diversity of faecal
31 (2013). Gudi,R. & Vasu,C. Impact of dietary deviation on microbiota in Crohns disease revealed by a
68. Statnikov,A. etal. Microbiomic signatures of disease progression and gut microbiome composition metagenomic approach. Gut 55, 205211 (2006).
psoriasis: feasibility and methodology comparison. in lupus-prone SNF1 mice. Clin. Exp. Immunol. 181, 117. McInnes,I.B. & Schett,G. The pathogenesis of
Sci. Rep. 3, 2620 (2013). 323337 (2015). rheumatoid arthritis. N.Engl. J.Med. 365, 22052219
69. Fahlen,A., Engstrand,L., Baker,B.S., Powles,A. 93. Palm,O., Moum,B., Jahnsen,J. & Gran,J.T. (2011).
& Fry,L. Comparison of bacterial microbiota in Fibromyalgia and chronic widespread pain in patients 118. Wolff,B. etal. Oral status in patients with early
skin biopsies from normal and psoriatic skin. with inflammatory bowel disease: a cross sectional rheumatoid arthritis: a prospective, casecontrol
Arch. Dermatol. Res. 304, 1522 (2012). population survey. J.Rheumatol. 28, 590594 (2001). study. Rheumatology (Oxford) 53, 526531 (2014).

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 13



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

119. Scher,J.U. etal. Periodontal disease and the oral 143. Arbuckle,M.R. etal. Development of autoantibodies 167. Roberfroid,M. etal. Prebiotic effects: metabolic and
microbiota in new-onset rheumatoid arthritis. before the clinical onset of systemic lupus health benefits. Br. J.Nutr. 104, S1S63 (2010).
Arthritis Rheum. 64, 30833094 (2012). erythematosus. N.Engl. J.Med. 349, 15261533 168. Neyrinck,A.M. etal. Prebiotic effects of wheat
120. AlKatma,M.K., Bissada,N.F., Bordeaux,J.M., (2003). arabinoxylan related to the increase in bifidobacteria,
Sue,J. & Askari,A.D. Control of periodontal infection 144. Ghouri,Y.A. etal. Systematic review of randomized Roseburia and Bacteroides/Prevotella in diet-induced
reduces the severity of active rheumatoid arthritis. controlled trials of probiotics, prebiotics, and obese mice. PLoS ONE 6, e20944 (2011).
J.Clin. Rheumatol. 13, 134137 (2007). synbiotics in inflammatory bowel disease. Clin. Exp. 169. Everard,A. etal. Responses of gut microbiota and
121. McGraw,W.T., Potempa,J., Farley,D. & Travis,J. Gastroenterol. 7, 473487 (2014). glucose and lipid metabolism to prebiotics in genetic
Purification, characterization, and sequence analysis 145. Kerman,D.H. & Deshpande,A.R. Gut microbiota and obese and diet-induced leptin-resistant mice. Diabetes
of a potential virulence factor from Porphyromonas inflammatory bowel disease: the role of antibiotics in 60, 27752786 (2011).
gingivalis, peptidylarginine deiminase. Infect. Immun. disease management. Postgrad. Med. 126, 719 170. Van Loo,J. etal. Functional food properties of
67, 32483256 (1999). (2014). non-digestible oligosaccharides: a consensus report
122. Rosenstein,E.D., Greenwald,R.A., Kushner,L.J. & 146. Van Praet,L., Jacques,P., Van den Bosch,F. & from the ENDO project (DGXII AIRIICT941095).
Weissmann,G. Hypothesis: the humoral immune Elewaut,D. The transition of acute to chronic bowel Br. J.Nutr. 81, 121132 (1999).
response to oral bacteria provides a stimulus for the inflammation in spondyloarthritis. Nat. Rev. 171. Alipour,B. etal. Effects of Lactobacillus casei
development of rheumatoid arthritis. Inflammation Rheumatol. 8, 288295 (2012). supplementation on disease activity and inflammatory
28, 311318 (2004). 147. Kruglov,A.A. etal. Nonredundant function of soluble cytokines in rheumatoid arthritis patients:
123. Liao,F. etal. Porphyromonas gingivalis may play an LT3 produced by innate lymphoid cells in intestinal a randomized double-blind clinical trial.
important role in the pathogenesis of periodontitis- homeostasis. Science 342, 12431246 (2013). Int. J.Rheum. Dis. 17, 519527 (2014).
associated rheumatoid arthritis. Med. Hypotheses 148. Vijay-Kumar,M. etal. Metabolic syndrome and 172. Vaghef-Mehrabany,E. etal. Probiotic
72, 732735 (2009). altered gut microbiota in mice lacking Toll-like supplementation improves inflammatory status in
124. Wegner,N. etal. Peptidylarginine deiminase from receptor 5. Science 328, 228231 (2010). patients with rheumatoid arthritis. Nutrition 30,
Porphyromonas gingivalis citrullinates human 149. Elinav,E. etal. NLRP6 inflammasome regulates 430435 (2014).
fibrinogen and -enolase: implications for colonic microbial ecology and risk for colitis. Cell 145, 173. van Nood,E. etal. Duodenal infusion of donor feces
autoimmunity in rheumatoid arthritis. Arthritis 745757 (2011). for recurrent Clostridium difficile. N.Engl. J.Med.
Rheum. 62, 26622672 (2010). 150. Goodrich,J.K. etal. Human genetics shape the gut 368, 407415 (2013).
125. Kinloch,A.J. etal. Immunization with Porphyromonas microbiome. Cell 159, 789799 (2014). 174. McFarland,L.V. Use of probiotics to correct dysbiosis
gingivalis enolase induces autoimmunity to mammalian 151. Kokkonen,H. etal. Associations of antibodies against of normal microbiota following disease or disruptive
-enolase and arthritis in DR4IEtransgenic mice. citrullinated peptides with human leukocyte antigen- events: a systematic review. BMJ Open 4, e005047
Arthritis Rheum. 63, 38183823 (2011). shared epitope and smoking prior to the development (2014).
126. Hitchon,C.A. etal. Antibodies to Porphyromonas of rheumatoid arthritis. Arthritis Res. Ther. 17, 125 175. Lemon,K.P., Armitage,G.C., Relman,D.A. &
gingivalis are associated with anticitrullinated protein (2015). Fischbach,M.A. Microbiota-targeted therapies:
antibodies in patients with rheumatoid arthritis and 152. Too,C.L. etal. Smoking interacts with HLADRB1 an ecological perspective. Sci. Transl. Med. 4,
their relatives. J.Rheumatol. 37, 11051112 (2010). shared epitope in the development of anti-citrullinated 137rv5 (2012).
127. Okada,M. etal. Periodontal treatment decreases protein antibody-positive rheumatoid arthritis: results 176. Khoruts,A., Dicksved,J., Jansson,J.K. &
levels of antibodies to Porphyromonas gingivalis and from the Malaysian Epidemiological Investigation of Sadowsky,M.J. Changes in the composition of the
citrulline in patients with rheumatoid arthritis and Rheumatoid Arthritis (MyEIRA). Arthritis Res. Ther. human fecal microbiome after bacteriotherapy for
periodontitis. J.Periodontol. 84, e74e84 (2013). 14, R89 (2012). recurrent Clostridium difficile-associated diarrhea.
128. Martinez-Martinez,R.E. etal. Detection of 153. Mahdi,H. etal. Specific interaction between J.Clin. Gastroenterol. 44, 354360 (2010).
periodontal bacterial DNA in serum and synovial fluid genotype, smoking and autoimmunity to citrullinated 177. de Vrieze,J. Medical research. The promise of poop.
in refractory rheumatoid arthritis patients. J.Clin. -enolase in the etiology of rheumatoid arthritis. Science 341, 954957 (2013).
Periodontol. 36, 10041010 (2009). Nat. Genet. 41, 13191324 (2009). 178. Allen-Vercoe,E. etal. A Canadian Working Group
129. Vaahtovuo,J., Munukka,E., Korkeamaki,M., 154. Lundstrom,E., Kallberg,H., Alfredsson,L., report on fecal microbial therapy: microbial
Luukkainen,R. & Toivanen,P. Fecal microbiota in early Klareskog,L. & Padyukov,L. Geneenvironment ecosystems therapeutics. Can. J.Gastroenterol. 26,
rheumatoid arthritis. J.Rheumatol. 35, 15001505 interaction between the DRB1 shared epitope and 457462 (2012).
(2008). smoking in the risk of anti-citrullinated protein 179. Petrof,E.O. etal. Stool substitute transplant therapy
130. Zhang,X. etal. The oral and gut microbiomes are antibody-positive rheumatoid arthritis: all alleles for the eradication of Clostridium difficile infection:
perturbed in rheumatoid arthritis and partly normalized are important. Arthritis Rheum. 60, 15971603 RePOOPulating the gut. Microbiome 1, 3 (2013).
after treatment. Nat. Med. 21, 895905 (2015). (2009). 180. Dziechciarz,P., Horvath,A., Shamir,R. &
131. Scher,J.U. etal. Expansion of intestinal Prevotella 155. Kallberg,H. etal. Genegene and geneenvironment Szajewska,H. Meta-analysis: enteral nutrition in active
copri correlates with enhanced susceptibility to interactions involving HLADRB1, PTPN22, and Crohns disease in children. Aliment. Pharmacol. Ther.
arthritis. eLife 2, e01202 (2013). smoking in two subsets of rheumatoid arthritis. 26, 795806 (2007).
132. Cuchacovich,R. etal. Detection of bacterial DNA in Am. J.Hum. Genet. 80, 867875 (2007). 181. Holmes,E. etal. Therapeutic modulation
Latin American patients with reactive arthritis by 156. Wu,G.D. etal. Linking long-term dietary patterns with of microbiotahost metabolic interactions.
polymerase chain reaction and sequencing analysis. gut microbial enterotypes. Science 334, 105108 Sci. Transl. Med. 4, 137rv6 (2012).
J.Rheumatol. 29, 14261429 (2002). (2011). 182. Sonnenburg,J.L. Microbiome engineering. Nature
133. Morris,A. etal. Comparison of the respiratory 157. David,L.A. etal. Diet rapidly and reproducibly alters 518, S10 (2015).
microbiome in healthy nonsmokers and smokers. the human gut microbiome. Nature 505, 559563 183. Donaldson,G.P., Lee,S.M. & Mazmanian,S.K.
Am. J.Respir. Crit. Care Med. 187, 10671075 (2013). (2014). Gut biogeography of the bacterial microbiota.
134. Erb-Downward,J.R. etal. Analysis of the lung 158. Chassaing,B. etal. Dietary emulsifiers impact the Nat. Rev. Microbiol. 14, 2032 (2016).
microbiome in the healthy smoker and in COPD. mouse gut microbiota promoting colitis and metabolic 184. Ottman,N., Smidt,H., de Vos,W.M. & Belzer,C. The
PLoS ONE 6, e16384 (2011). syndrome. Nature 519, 9296 (2015). function of our microbiota: who is out there and what do
135. Hilty,M. etal. Disordered microbial communities in 159. Evans,C.H., Ghivizzani,S.C. & Robbins,P.D. they do? Front. Cell. Infect. Microbiol. 2, 104 (2012).
asthmatic airways. PLoS ONE 5, e8578 (2010). Gene therapy of the rheumatic diseases: 1998 to 185. Sekirov,I., Russell,S.L., Antunes,L.C. & Finlay,B.B.
136. Segal,L.N. etal. Enrichment of lung microbiome with 2008. Arthritis Res. Ther. 11, 209 (2009). Gut microbiota in health and disease. Physiol. Rev.
supraglottic taxa is associated with increased 160. Edwards,C.J. & Cooper,C. Early environmental 90, 859904 (2010).
pulmonary inflammation. Microbiome 1, 19 (2013). factors and rheumatoid arthritis. Clin. Exp. Immunol. 186. Aziz,Q., Dore,J., Emmanuel,A., Guarner,F. &
137. Makrygiannakis,D. etal. Smoking increases 143, 15 (2006). Quigley,E.M. Gut microbiota and gastrointestinal
peptidylarginine deiminase 2 enzyme expression in 161. Atarashi,K. etal. Treg induction by a rationally health: current concepts and future directions.
human lungs and increases citrullination in BAL cells. selected mixture of Clostridia strains from the human Neurogastroenterol. Motil. 25, 415 (2013).
Ann. Rheum. Dis. 67, 14881492 (2008). microbiota. Nature 500, 232236 (2013). 187. Lawley,T.D. & Walker,A.W. Intestinal colonization
138. Gan,R.W. etal. Relationship between air pollution 162. Gibson,S.A., McFarlan,C., Hay,S. & resistance. Immunology 138, 111 (2013).
and positivity of RArelated autoantibodies in MacFarlane,G.T. Significance of microflora in 188. Delzenne,N.M. & Cani,P.D. A place for dietary
individuals without established RA: a report on SERA. proteolysis in the colon. Appl. Environ. Microbiol. fibre in the management of the metabolic syndrome.
Ann. Rheum. Dis. 72, 20022005 (2013). 55, 679683 (1989). Curr. Opin. Clin. Nutr. Metab. Care 8, 636640
139. Willis,V.C. etal. Sputum autoantibodies in patients 163. Marzorati,M. etal. In vitro modulation of the human (2005).
with established rheumatoid arthritis and subjects at gastrointestinal microbial community by plant-derived 189. Schmidt,C. Mental health: thinking from the gut.
risk of future clinically apparent disease. Arthritis polysaccharide-rich dietary supplements. Int. J.Food Nature 518, S1215 (2015).
Rheum. 65, 25452554 (2013). Microbiol. 139, 168176 (2010). 190. Knoop,K.A. & Newberry,R.D. Isolated lymphoid
140. Lugli,E.B. etal. Expression of citrulline and 164. Voreades,N., Kozil,A. & Weir,T.L. Diet and the follicles are dynamic reservoirs for the induction of
homocitrulline residues in the lungs of non-smokers development of the human intestinal microbiome. intestinal IgA. Front. Immunol. 3, 84 (2012).
and smokers: implications for autoimmunity in Front. Microbiol. 5, 494 (2014).
rheumatoid arthritis. Arthritis Res. Ther. 17, 9 (2015). 165. Saad,N., Delattre,C., Urdaci,M., Schmitter,J.M. & Author contributions
141. Hevia,A. etal. Intestinal dysbiosis associated with Bressollier,P. An overview of the last advances in T.V.d.W., J.T.V.P., M.M., M.B.D. and D.E. researched data for
systemic lupus erythematosus. mBio 5, e0154801514 probiotic and prebiotic field. Lebenson. Wiss. Technol. the article. T.V.d.W., J.v.P and D.E. made substantial contribu-
(2014). 50, 116 (2013). tions to the discussion of content. All authors contributed to
142. Castelino,M., Eyre,S., Upton,M., Ho,P. & Barton,A. 166. Preidis,G.A. & Versalovic,J. Targeting the human the writing of the manuscript. T.V.d.W., J.v.P., M.B.D. and D.E.,
The bacterial skin microbiome in psoriatic arthritis, microbiome with antibiotics, probiotics, and reviewed/edited the manuscript before submission.
an unexplored link in pathogenesis: challenges and prebiotics: gastroenterology enters the
opportunities offered by recent technological advances. metagenomics era. Gastroenterology 136, Competing interests statement
Rheumatology (Oxford) 53, 777784 (2014). 20152031 (2009). The authors declare no competing interests.

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like