You are on page 1of 13

Clinical Immunology 215 (2020) 108415

Contents lists available at ScienceDirect

Clinical Immunology
journal homepage: www.elsevier.com/locate/yclim

Review Article

Gut microbiota in chronic inflammatory disorders: A focus on pediatric T


inflammatory bowel diseases and juvenile idiopathic arthritis

Amanda Ricciutoa,b, , Philip M. Shermana,b, Ronald M. Laxerb,c
a
Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
b
Department of Paediatrics, Faculty of Medicine, University of Toronto, Canada
c
Division of Rheumatology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada

A R T I C LE I N FO A B S T R A C T

Keywords: The gut microbiota is integral to human health, including maintaining the delicate balance between tolerance
Dysbiosis and protection against potentially harmful pathogens. A growing body of evidence implicates the intestinal
Fecal microbial transplantation microbiome in immune-mediated inflammatory disorders; these data span the spectrum from genetic and en-
Inflammatory bowel disease vironmental disease risk factors, to animal studies (particularly germ-free and gnotobiotic models) and human
Juvenile idiopathic arthritis
studies, including evidence of dysbiosis in diseased individuals compared to healthy populations. In this review,
Microbiota
we summarize both animal and human data supporting a link between the gut microbiota and inflammatory
bowel diseases (IBD) and systemic inflammatory arthritis, as models for chronic inflammatory disorders, while
offering a pediatric focus (pediatric IBD and juvenile idiopathic arthritis). We discuss relevant mechanisms
related to the crosstalk between the gut microbiota and the innate and adaptive immune system. We close with a
brief discussion of emerging microbe-altering interventions, including fecal microbial transplantation and its
immunologic effects.

1. Introduction tremendous interest of late. Numerous studies document differences in


the microbial community structure (i.e., dysbiosis) of individuals af-
Immune-mediated inflammatory disorders (IMIDs), including fected by various diseases (including IMIDs) compared to healthy
chronic inflammatory bowel diseases (IBD) and juvenile inflammatory controls (HCs). Moreover, animal studies demonstrate the transfer-
arthritides (JIA), are highly prevalent in industrialized countries. It is ability of disease phenotype upon transfer of fecal material from dis-
projected, for example, that by the year 2030, the prevalence of IBD eased humans or animals to germ-free (GF) mice, offering particularly
will approach 1% in Canada [1]. More alarming still is that young compelling support for a causal link between gut microbes and disease.
children represent the fastest growing group [2]. While overall in- The reciprocal, multi-faceted interactions between the gut microbiota
cidence rates are stabilizing in the developed world, they are now in- and human immune system add biological plausibility to the hypothesis
creasing in developing countries, posing a growing global burden [3]. that gut microbes are causally implicated in immune-mediated dis-
IMIDs are currently thought to result from a complex interplay be- orders. This has exciting implications for the development of potential
tween host genetics and various environmental factors. A fascinating therapies with microbe-altering properties, such as nutritional inter-
epidemiologic phenomenon is that the risk of certain IMIDs in im- ventions, pro- and prebiotics, antibiotics and fecal microbial trans-
migrants to high prevalence regions approximates that in host re- plantation (FMT).
sidents, such as the risk of ulcerative colitis (UC), an IBD subtype, in In what follows, we review the role of the gut microbiota in both
South Asians [4]. Observations like these, coupled with rapid fluxes in IBD and inflammatory arthropathies, as models for chronic in-
disease rates over relatively short periods of time, indicate that genetic flammatory disorders, while offering a pediatric focus. We begin by
factors cannot be solely to blame. Rather, these findings point strongly summarizing methods used to characterize the microbiota, the influ-
to the role of environmental risk factors. ence of the gut microbiota on the immune system and evidence sup-
Among environmental factors, the gut microbiota has garnered porting an association between the microbiome and both IBD and

Abbreviations: IBD, inflammatory bowel diseases; JIA, juvenile idiopathic arthritis



Corresponding author at: Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8,
Canada.
E-mail addresses: amanda.ricciuto@sickkids.ca (A. Ricciuto), philip.sherman@sickkids.ca (P.M. Sherman), ronald.laxer@sickkids.ca (R.M. Laxer).

https://doi.org/10.1016/j.clim.2020.108415
Received 13 February 2020; Received in revised form 6 April 2020; Accepted 6 April 2020
Available online 09 April 2020
1521-6616/ © 2020 Elsevier Inc. All rights reserved.
A. Ricciuto, et al. Clinical Immunology 215 (2020) 108415

Fig. 1. Functions of the gut microbiota in human


health.
Short chain fatty acids are derived from indigestible
complex carbohydrates; they serve as an energy
source for colonocytes and have important im-
munoregulatory functions. “Colonization resistance”
refers to mechanisms by which gut microbes protect
themselves and the host against incursion by poten-
tially harmful microorganisms.

arthritis. We then review microbial changes reported in both condi- relatively high cost compared with 16S rRNA profiling and the issue of
tions, while highlighting pediatric findings. We close with a discussion reproducibility of findings both within samples and between labora-
of selected future novel therapies, including FMT. tories.

2. Microbiota characterization methods 3. The microbiome in human health

The human body includes a complex mix of mammalian and pro- Most intestinal bacteria belong to 4 phyla: Firmicutes,
karyotic cells, present in roughly equal proportions [5]. The microbial Bacteroidetes, Proteobacteria and Actinobacteria, with the first two
flora harbored by humans is termed the “microbiota.” It includes predominating in health. Microbial diversity increases along the gas-
symbiotic and commensal microorganisms, as well as pathobionts trointestinal tract, from stomach to colon and, at the terminal ileum,
(bacteria with the potential to be pathogenic). The term “microbiome” prevalent species transition from aerobes to facultative and strict
denotes the collective genomes of the microbiota. anaerobes. Microbial populations differ substantially between the gut
Standard culturing techniques are unable to identify at least 80% of lumen and mucosal surfaces; the latter are physically closer to the in-
the human microbiota [6]. The flurry of microbiome-focused research testinal epithelium and, consequently, may exert a greater effect on the
in recent years was spurred by the advent of technologies that enable host, including the innate and adaptive arms of the immune system. For
more comprehensive microbial characterization, particularly high example, in trinitrobenzenesulfonic acid (TNBS)-induced colitis in
throughput DNA sequencing. This method uses the 16S small ribosomal mice, the microbiomes of feces, cecum and mucus are distinct, and the
subunit (16S rRNA), a gene unique to bacteria, to identify bacteria and mucus microbiome correlates most closely with disease severity [9].
Archaea [7]. The 16S rRNA gene includes elements that are conserved Moreover, in a human study of functional gastrointestinal disorders,
between most bacteria and hypervariable elements that enable the mucosa-associated Faecalibacterium and Ruminococcus were inversely
differentiation of species. An analogous approach can be applied to correlated with Toll-like receptor (TLR)4 expression while, in luminal
fungi, which uses the internal transcribed spacer (ITS), 18S rRNA or 26S samples, there was no correlation with TLR4 at any taxonomic level
rRNA regions. While these methods have proven extremely useful, they [10]. It is pertinent, therefore, to consider the source of microbes (fecal
have certain limitations. These include a limited ability to differentiate or mucosal) in interpreting findings reported in various studies of the
commensal from pathogenic strains, especially in the setting of hor- gut microbiota.
izontal gene transfer (the exchange of virulence factors between species Over a person's lifetime, the diversity and composition of the mi-
in the same environment) [8], and to identify non-bacterial strains. crobiota fluctuate with age and extrinsic factors, such as diet, lifestyle,
Metagenomics, the study of the collective genomes (bacterial, antibiotics and other environmental exposures. Somewhat surprisingly,
fungal and viral) present in an environmental sample, allows even more the functional capacity of the healthy microbiome is relatively stable
detailed compositional profiling of a microbial community. Shotgun irrespective of its specific composition [11]. The gut microbiota per-
metagenomics refers to a method by which DNA extracted from an forms numerous functions that are integral to human health. These are
environment is sheared into small fragments, which are then se- outlined in Fig. 1 and can be grouped into 3 main categories: nutrition
quenced. Importantly, whole genome sequencing can be used to deduce and metabolism; protection against pathogens (or “colonization re-
microbial function, including enzymatic capacity. Metagenomics is sistance”); and immune system development. The latter is further de-
complemented by the methods of metatranscriptomics, metaproteomics tailed below.
and metabolomics, which respectively examine gene expression, and
protein and metabolite production by a microbial community.
Limitations of whole genome sequencing currently relate to the

2
A. Ricciuto, et al. Clinical Immunology 215 (2020) 108415

4. The effects of the gut microbiota on the human immune system repeated TLR activation by commensal microbes suppresses proin-
flammatory NF-ΚΒ signaling, downstream chemokine (IL-8) production
4.1. The innate and adaptive immune system: a refresher and recruitment of neutrophils. Moreover, PRR triggering activates and
induces antigen-presenting cells (APCs), which in turn promote Treg
The gut faces the remarkable challenge of maintaining an intricate differentiation [16]. In keeping with this, feeding LPS to GF mice
balance between tolerance to the 500 to 1000 commensal bacteria challenged with dextran sulfate sodium (DSS) to induce colitis is pro-
present in the gut and protection against potentially harmful pathogens. tective, and associated with an increase in CD103+ DCs and induction
The predominantly tolerogenic immunologic response to intestinal lu- of Tregs in the distal intestine [17]. Several microbial metabolites also
minal antigens (microbial and dietary) is enabled by the recognition of affect innate immune responses. SCFAs (acetate, propionate and buty-
antigen patterns by innate immune cells, which in turn direct T cell rate) are produced from microbial fermentation of host-indigestible
reactivity. Microbial sensing by mononuclear phagocytes (MNPs) of the complex carbohydrates, largely by Firmicutes and Bacteroidetes. SCFAs
innate system (macrophages, dendritic cells (DCs)) and intestinal epi- influence innate cells via G protein-coupled receptors; they suppress the
thelial cells (ECs) is mediated by pattern recognition receptors (PRRs), production of proinflammatory cytokines, such as TNF-α, IL-6 and ni-
such as TLRs and nucleotide-binding oligomerization domain (NOD)- tric oxide (NO) [18]. Bacterial-derived tryptophan metabolites have the
like receptors (NLRs). TLRs and NLRs recognize pathogen-associated capacity to directly modulate ILC3 functionality via the aryl hydro-
molecular patterns (PAMPs), such as lipopolysaccharide (LPS). TLRs carbon receptor (AhR) [19].
also stimulate the production of antimicrobial peptides (AMPs), such as Within the adaptive arm of the host immune system, the microbiota
regenerating family protein III (Reg III β/γ). Innate lymphoid cells is critical for the development of the Th17 compartment in the gut la-
(ILCs) also belong to the innate arm of the immune system. They do not mina propria [20,21]. This is illustrated by the absence of Th17 cells in
bear antigen-specific T or B cell receptors but share certain functional mice reared under GF conditions [21]. Among commensal bacteria in
characteristics with CD4+ T helper (Th) cells. There are several distinct mice, segmented filamentous bacteria (SFB) have a particularly potent
types; ILC3s constitute the majority of intestinal ILCs. They secrete in- ability to induce Th17 cells; the addition of SFB alone to GF mice is
terleukin (IL)-17 and IL-22 and are generally considered the innate sufficient for Th17 cells to accumulate in the small bowel [22,23].
counterpart of Th17 cells. Based on a more recent study, adhesion of microbes to intestinal ECs is
Within the adaptive arm of the immune system, T helper cells are an important cue for Th17 induction; this was observed with SFB, as
classically categorized into Th1 and Th2 subsets; Th1 considered the well as the murine-specific enteropathogen Citrobacter rodentium, the
primary mediator of responses to extracellular pathogens (through human enteric pathogen Escherichia coli serotype O157:H7 and a mix-
production of interferon (IFN)-γ, which recruits macrophages), and Th2 ture of 20 bacterial strains derived from feces of adult UC patients [24].
considered the primary mediator of response to parasites (through se- There are also ample data to support the role of gut bacteria and
cretion of IL-4, IL-5 and IL-13). IBD includes two main subtypes, UC and their metabolites in promoting Tregs. SCFAs can stimulate colonic Tregs
Crohn's disease (CD). Based on this framework, UC was traditionally directly, through physical inhibition of histone deacetylases (HDACs)
considered to be predominantly Th2-mediated and CD to be Th1- involved in the post-translational regulation of the transcription factor
mediated. However, this paradigm was expanded and refined following FoxP3, or indirectly, via G protein-coupled receptors. SCFAs also pro-
the discovery of complementary Th subsets, including proinflammatory mote the differentiation of CD4+ naïve T cells into Tregs. Mouse and in
Th17 cells (CD4+ IL-17-secreting T cells) and anti-inflammatory T vitro studies support the ability of SCFAs and SCFA-producing bacteria
regulatory (Treg) cells, which express the nuclear transcription factor to induce Tregs and demonstrate their association with FoxP3 expres-
Forkhead box P3 (FoxP3). sion and higher IL-10 levels [25,26]. Colonic Treg abundance correlates
Th17 cells play an important role in mucosal defense, while Tregs with butyrate concentrations and butyrate is protective against the
dampen the proliferation and functions of other T cells. Although Th17 development of colitis in an adoptive T cell transfer model of re-
and Treg cells herald from a common CD4+ naïve T cell, their differ- combination activating gene (RAG)−/− mice (lacking both T and B
entiation is contingent on specific inputs, specifically transforming cells) [27]. Retinoic acid and tryptophan products are additional mi-
growth factor (TGF)-β in the presence of retinoic acid for Tregs, and crobial metabolites with Treg-stimulating effects. Retinoic acid is gen-
TGF-β, IL-6 and IL-21 (in humans) for Th17. The proinflammatory/anti- erated by DCs in a process that requires commensal support. This re-
inflammatory balance between Th17 and Treg cells is now recognized tinoic acid then leads to increased gut-homing IL-10-producing Tregs
to be fundamental to the etiopathogenesis of T-cell mediated IMIDs like and enhanced Treg differentiation [28]. Microbial tryptophan meta-
IBD and JIA [12,13]. The complexity of the Th framework continues to bolites (kynurenines), produced by bacteria like Bifidobacterium longum,
grow, with more recent data demonstrating the existence of additional B. thetaiotaomicron and Faecalibacterium prausnitzii, drive intraepithelial
Th subsets, such as regulatory type 1 (Tr1) cells (CD4+ T cells that CD4+ Th cells toward an immunoregulatory phenotype, an effect
possess immunoregulatory properties and produce IL-10, but are mediated by AhR [29–31].
FoxP3-negative) as well as follicular Th (Tfh) cells (which promote In addition to microbial metabolites, several specific bacteria, in-
Th1/Th17 polarization), Th9 cells and Th22 cells. cluding Clostridia, Bacteroides fragilis and Faecalibacterium prausnitzii,
are directly implicated in Treg induction. In seminal work by Atarashi
4.2. The reciprocal interactions between the microbiota and the immune et al. [32,33], the transfer of specific Clostridia strains from human
system stool into GF mice triggered a significant increase in Tregs, which were
low at baseline. This occurred in conjunction with production of anti-
The paramount importance of the gut microbiota in shaping a inflammatory molecules, such as IL-10, TGF-β1, cytotoxic T lymphocyte
healthy immune system is highlighted by the immunologic defects that antigen-4 (CTLA-4) and inducible T cell co-stimulator (ICOS)), as well
arise in mice raised in germ free (GF) environments. These animals as SCFAs. The polysaccharide A (PSA) of the capsule of B. fragilis sti-
display lymphoid hypoplasia in the spleen, lymph nodes, thymus and at mulates differentiation of naïve CD4+ T cells into IL-10-secreting Tregs
mucosal surfaces; they have fewer peripheral CD4+ T cells (with a Th2 or Tr1 cells via plasmacytoid DC stimulation [34,35]. In the absence of
bias); and have lower immunoglobulin (Ig) levels [14,15]. APCs, PSA can directly activate TLR2 on FoxP3+ T cells to trigger the
The influence of the gut microbiota on the immune system is ap- production of immunoregulatory molecules like IL-10 [36]. As dis-
parent within both the innate and adaptive arms. Communication be- cussed in 5.1.3 below, F. prausnitzii has garnered significant attention in
tween gut microbes and innate MNPs is key to maintaining intestinal IBD. This bacterium displays several anti-inflammatory effects, in part
homeostasis. TLR activation by commensal bacteria can induce, but mediated by its product, microbial anti-inflammatory molecule (MAM),
also dampen and regulate, intestinal inflammation. For example, which blocks NF-ΚΒ activation and IL-8 production [37–39]. F.

3
A. Ricciuto, et al. Clinical Immunology 215 (2020) 108415

prausnitzii is associated with a tolerogenic serum cytokine profile, Transfer of these bacteria into IL-10-deficient mice and K/BxN mice (an
characterized by low levels of IL-12 and IFN-γ, and high IL-10 [40]. arthritis model) exacerbates colitis and arthritis, respectively, lending
Thus far, we have described the role of the gut microbiota in support to the pathogenicity of these bacteria. F. prausnitzii, on the
shaping local immune responses. Only recently was it shown that in- other hand, displays a protective effect against colitis in animals; the
testinal microbes influence early lymphocyte development in the oral administration of F. prausnitzii (or its supernatant) mitigates TNBS-
thymus as well, as supported by impaired thymic lymphocyte devel- induced colitis in mice [37].
opment observed in GF mice. Congruent with the above discussion, B.
fragilis monocolonization restored this defect but not when a PSA-de- 5.3. Evidence relating to IBD – human studies
ficient strain was used [41].
A few general observations lend support to the role of the gut mi-
5. Evidence supporting the role of the gut microbiome in IBD and crobiota in IBD pathogenesis. These include the fact that IBD pre-
inflammatory arthropathies dominates in gut regions of high bacterial load (terminal ileum and
colon); fecal diversion is associated with improvement of CD actvity
5.1. Evidence relating to disease risk factors [59,60]; and antibiotic therapy is helpful for selected types of IBD, such
as perianal CD and pouchitis in UC [42].
Both genetic and environmental risk factors for IBD and in- Numerous studies demonstrate a state of dysbiosis in adult- and
flammatory arthritis point to the possible role of the gut microbiome in pediatric-onset IBD. While these reports are consistent in showing dif-
disease pathogenesis. Many of the susceptibility loci for IBD relate to ferences between IBD and control cohorts, they are otherwise hugely
interactions between the immune system and the microbiota. heterogeneous, in term of both the methods employed and the specific
Illustrative genes include NOD2, autophagy-related 16-like 1 findings arising, which limits interpretability. Several such studies are
(ATG16L1), caspase recruitment domain-containing protein 9 (CARD9) discussed below, particularly within the framework of a recent meta-
and C-type lectin domain family 7 member (CLEC7A). NOD2 encodes analysis of the microbiome in IBD [61]. Table 1 provides a summary of
an intracellular PRR. Mice deficient in NOD2 have intestinal dysbiosis microbiota studies in pediatric IBD (discussed below), further high-
and a heightened susceptibility to colitis. Furthermore, in the setting of lighting the heterogeneity of findings between studies. Contributors to
NOD2 deficiency, commensal microbes, like Bacteroides vulgatus, trigger heterogeneity include sample source (fecal vs mucosal), specific
mucosal barrier disruption and inflammatory gene expression [42]. In methods used to characterize the microbiota, medication exposure, as
humans, NOD2 mutations are associated with lower IL-10, increased well as patient age and geographic location of residence.
mucosa-associated bacteria [43], as well as depletion of Faecalibacteria Despite these limitations, a few generalizations can be made. The
and enrichment of Escherichia [44–47]. Many environmental factors first is that IBD is associated with an overall reduction of microbial
that have been identified as risk factors for IBD (and other IMIDs) also diversity (i.e., the total number of bacterial species). Data from the
invoke change in the gut microbiota. Briefly, these include exposure to MetaHIT consortium suggest that, on average, IBD patients have 25%
bouts of acute gastroenteritis, breastfeeding, early antibiotic use, fewer microbial-derived genes than are present in healthy controls
changes in diet (particularly high fat content), cigarette smoking and [62]. Temporal instability is another characteristic of the IBD micro-
the hygiene hypothesis more broadly [42]. biota. This was demonstrated in a longitudinal study, in which 128 IBD
patients were serially sampled. Microbial fluctuations were particularly
5.2. Evidence relating to IBD – animal studies marked in individuals with ileal CD post surgical resection [63].
Secondly, IBD is associated with a loss of commensal bacteria and
Observations of attenuated colitis in GF animals and animals treated enrichment of pathobionts. At a phylum level, this typically translates
with antibiotics offer compelling support for the involvement of the gut into a relative loss of Firmicutes and enrichment of Proteobacteria.
microbiome in the pathogenesis of intestinal inflammation [48–50]. For Among commensal microorganisms, SCFA producers are often de-
example, IL-10−/− mice (a common colitis model) do not develop co- pleted, including multiple Firmicutes, such as Clostridium cluster IV
litis when GF. Intriguingly, the reintroduction of specific bacteria may (which includes Ruminococcaceae family members such as F. praus-
be beneficial (Lactobacillus plantarum 299 V) or harmful (Enterobacter nitzii), Clostridium cluster XIVa (which includes Ruminococcus and
faecalis) in this model [50–52]. Seminal work from the 1990s showed Lachnospiraceae family members such as Roseburia and Blautia),
that the adoptive transfer of naïve CD4+ T cells fails to induce colitis in Leuconostocaceae, Phascolarctobacterium, as well as certain
RAG1−/− mice raised under GF conditions [53,54]. However, upon Bacteroidetes, such as Odoribacter splanchnius [64–66]. Both F. praus-
activation by gut microbial antigens, transfer of CD4+ T cells into se- nitzii and Roseburia are butyrate producers and have been found to
vere combined immunodeficiency (SCID) mice effectively induces co- correlate inversely with disease activity in UC [65]. Decreased abun-
litis [55]. In a more recent study, colonization of GF mice using fecal dance of F. prausnitzii is also a risk factor for post-operative CD recur-
samples from IBD patients led to an increased abundance of Th17 and rence [37]. In addition to these bacteria, certain studies show reduc-
Th2 cells and decreased Tregs, compared to fecal samples from healthy tions of other Firmicutes (such as Lactobacillus), Actinobacteria (such
controls [56]. Moreover, the transfer of IBD microbial samples wor- as Bifidobacterium), Bacteroidetes (such as Bacteroides) and selected
sened disease in a RAG1−/− T cell transfer model. Proteobacteria (such as Sutterella) in IBD populations [66–68], al-
Animal models of colitis reveal enrichment of specific bacteria, such though agreement is imperfect between studies. In light of the anti-
as Bacteroides, Enterobacteriaceae, Porphyromonas, Akkermansia mu- inflammatory effects of SCFAs and organisms like F. prausnitzii and
ciniphila and Clostridium ramosum, which are associated with increased Bacteroides, elaborated above, it is enticing to hypothesize that their
mucosal inflammation [48]. A relatively novel method, termed IgA-seq, loss is causally implicated in disease processes; however, it is exceed-
can be used to identify bacteria coated with IgA. The assumption is that ingly difficult to disentangle whether dysbiotic changes represent cause
these IgA-coated bacteria have a greater capacity to stimulate in- or consequence in the setting of chronic intestinal inflammation.
flammatory responses and, therefore, disease. Using this method, 35 Pathobionts belonging to the Proteobacteria phylum are frequently
bacterial species uniquely coated with IgA were identified in the stool enriched in IBD, including members of the Enterobacteriaceae family,
samples of IBD patients. In mice, these bacteria exacerbated DSS colitis such as E. coli, Shigella, Proteus mirabilis and Klebsiella pneumoniae. AIEC
[57]. Interestingly, compared to CD patients without arthritis, those strains have been isolated from the ileal tissue of CD patients [69].
with peripheral spondyloarthritis (SpA), are enriched in IgA-coated E. Other Proteobacteria reported to be increased in IBD (though with some
coli resembling adherent and invasive E. coli (AIEC) [58]. Colonization inconsistency) include Pasteurellaceae, Veillonellaceae and Desulfovi-
of GF mice with CD-SpA-derived E. coli triggers Th17 expansion. brio. Desulfovibrio can reduce sulfate, generating hydrogen sulfide

4
Table 1
Pediatric studies published since 2010 comparing the gut microbiota in IBD to a control group.
Author Region Patients Sample Method Depleted Enriched Diversity
Year
A. Ricciuto, et al.

CD
Aomatsu 2012 Japan 10 CD Stool 16S rRNA T-RFLP Clostridium IV (including F. prausnitzii), Clostridium Clostridium XIVa (different OTU), Lactobacillales ↓ vs HC/UC
[129] 27 HC XIVa, Bacteroides
Assa 2016 [75] Canada 10 CD TI or ICV bx V6 hypervariable region of F. prausnitzii, Oscillospira, unclassified No difference vs HC
15 HC 16S rRNA Ruminococcaceae
Ijaz 2016 [130] UK 13 + 6 CD Stool 16S rRNA V4 region and Ruminococcaceae, Lachnospiraceae, Enterobacteriaceae, Pasteurellaceae, Veillonella, Dorea, ↓ vs HC
14 HC shotgun metagenomics Phascolarctobacterium, Parabacteroides, Akkermansia, Anaerostipes, Enterococcus, Clostridium_XVIII,
Methanobrevibacter Clostridium XIVa
Kaakoush 2015 Australia 5 CD Stool V1–3 16S rRNA Number of OTUs decreased dramatically upon starting ↓ vs HC
[131] 5 HC pyrosequencing, shotgun exclusive enteral nutrition
metagenomics, Ion Proton
system
Knoll 2017 Germany 5 + 1 CD Stool Whole genome shotgun Nil significant in CD subgroup Nil significant in CD subgroup
[132] 12 HC metagenomics
Makonnen Finland 10 CD Stool 16S rRNA RT-PCR, DGGE of Bifidobacteria, Lachnospiraceae and Coriobacteriaceae; ↑ in patients with
2015 8 HC V6–8 region and culture Bacteroides and Bifidobacteria diversity lower vs HC; medium-high
[133] Ruminococcaceae diversity lower vs HC/UC activity vs HC
Quince 2015 UK 23 CD Stool 16S rRNA sequencing of V4 Coprococcus, Pseudobutyrivibrio, Ruminococcus, Unknown Ruminococcaceae, Parvimonas, Actinomyces, ↓ – decreased further
[115] 8 HC region, shotgun metagenomics Subdoligranulum, Faecalibacterium, Lachnospira, Enterococcus, Peptostreptococcus, Lactococcus, during exclusive
Clostridium, Alistipes, Parabacteroides, Anaerotruncus, Escherichia-Shigella, Eggerthella, Megasphera, enteral nutrition
Roseburia, Turicibacter, Anaerostipes, Catabacter, Prevotella, Gemella, Mogibacterium, Peptoniphilus,
Bifidobacterium, Bilophila, Slackia, unknown Abiotrophia, Streptococcus, Lachnospiraceae
Coriobacteriaceae genus, Dialister Clostridium clostridioformes, S. anginosus, Enterococcus
Lachnospiraceae – specifically E. rectale and Roseburia faecalis, Ruminococcus gnavus

5
inulinivorans
B. adolescentis, Ruminococcus bromii, Eubacterium spp.,
F. prausnitzii, Coprococcus eutactus, Subdoligranulum
variabile
Wang China 11 CD Stool 16S rRNA V3–4 region Anaerostipes, Blautia, Coprococcus, Faecalibacterium, Enterococcus ↓
2018 16 HC Lachnospira, Odoribacter, Roseburia, Ruminococcus,
[134] Sutterella
Sila 2020 Croatia 19 IBD Stool 16S rRNA gene TRFLP Clostridium genus, Firmicutes Streptococcus, Lactococcus, Enterococcus ↓ vs healthy siblings
[135] 20 HC (sib) Proteobacteria
19 HCs
Kowalska 2019 Poland 64 CD Stool 16S rRNA V3–4 region Bifidobacterium adolescentis, Roseburia (R. faecis), F. Enterococcus ↓ vs HC
[136] 18 HC prausnitzii, Gemmiger formicilis, Ruminococcus bromiii,
Veillonellaceae (Dialister), Adlercreutzia, Clostridium
elatum, Coprococcus
Kolho 2016 Finland 36 CD Stool Phylogenetic microarray Clostridium clusters IV and XIVa, F. prausnitzii Bacteroides fragilis, Clostridium ramosum, Eubacterium
[77] 8 HCs (HITChip) cylindroides

UC Depleted Enriched
Aomatsu 2012 Japan 14 UC Stool 16S rRNA TRFLP Clostridium IV, Bacteroides Lactobacillales No difference vs HC
[129] 27 HC
Knoll 2017 Germany 3 + 3 UC Stool Shotgun metagenomics Eubacterium rectale, F. prausnitzii, Bilophila wadsworthia E. coli, Clostridium ramosum, Ruminococcus gnavus, ↓
[132] 12 HC (trend) Fusobacterium nucleatum
Maukonen Finland 12 UC Stool RT-PCR, DGGE of 16S rRNA Bifidobacteria, Lachnospiraceae, Coriobacteriaceae, Bacteroides ↑ in patients with
2015 8 HC gene V6-V8 region, culture Lactobacillus medium-high
Bacteroides and Bifidobacteria diversity lower vs HC, activity vs HC
Ruminococcaceae diversity lower vs HC
(continued on next page)
Clinical Immunology 215 (2020) 108415
Table 1 (continued)

Author Region Patients Sample Method Depleted Enriched Diversity


Year
A. Ricciuto, et al.

Michail 2012 Canada, USA, 27 ASUC Stool 16S rRNA microarray Phylum: Firmicutes (5% decrease), Lentisphaerae, Phylum: Proteobacteria (3.6% increase), Fusobacteria, ↓, especially steroid
[137] Australia, Israel 26 HC Verrucomicrobia Spirochaetes non-responders
Class: Clostridia, Lentisphaerae, Coriobacteridae, Class: Actinobacteridae, Bacilli, Erysipelotrichi,
Verrucomicrobiae Fusobacteria, Alphaproteobacteria, Betaproteobacteria,
Deltaproteobacteria, Epsilonproteobacteria,
Gammaproteobacteria, Spirochetes
Species: E. coli
Shah 2016 [76] USA 10 UC Colonic bx 16S rRNA V4-V6 region Roseburia, Akkermansia Pasteurellaceae, RF32, Phenylobacterium, Similar UC vs HC
13 HC Haemophilus, F. prausnitzii (numeric only)
Sila 2020 Croatia 19 IBD Stool 16S rRNA gene T-RFLP Clostridium genus, Firmicutes Streptococcus, Lactococcus, Enterococcus ↓ vs healthy siblings
[138] 20 HC (sib) Proteobacteria
19 HCs
Turner 2019 Israel, Canada, 28 UC Stool 16S rRNA V4 Illumina MiSeq Gammaproteobacteria ↓ vs remission
[139] USA, Europe (vs remission)
Kolho 2016 Finland 26 UC Stool Phylogenetic microarray Sutterella wadsworthensis, C. difficile
[77] 8 HCs (HITChip)

Bx – biopsy; CD – Crohn's disease; DGGE – denaturing gradient gel electrophoresis; HC – healthy control; ICV – ileocecal valve; OTU – operational taxonomic unit; RT-PCR – reverse transcription polymerase chain
reaction; T- RFLP – terminal-restriction fragment length polymorphism; TI – terminal ileum; UC – ulcerative colitis.

6
Clinical Immunology 215 (2020) 108415
A. Ricciuto, et al. Clinical Immunology 215 (2020) 108415

(H2S), which is of relevance because H2S impairs the utilization of Table 2


butyrate (a source of energy) by colonocytes. Certain Firmicutes, in- Microbiota changes reported in chronic inflammatory bowel diseases.
cluding Ruminococcus gnavus and C. difficile, Bacteroidetes, including Depleted Enriched
Prevotellaceae, and Fusobacteria are also reported to be increased in
IBD [67,68,70]. Bacteria • Firmicutes phylum • Proteobacteria
The above findings are supported by a large pediatric study con- • Clostridium cluster IV phylum

ducted by Gevers et al. which examined fecal and mucosal samples


(Ruminococcaceae like
F. prausnitzii)
• Enterobacteriaceae (E.
coli, including adherent
from 468 treatment-naïve children newly diagnosed with CD and 229
healthy controls [71]. The CD group displayed increased abundances of
• Clostridium
XIVa
cluster and invasive E. coli
(AIEC), Shigella, P.
Pasteurellaceae, Veillonellaceae, Neisseriaceae, Fusobacteriaceae and (Ruminococcaceae and mirabilis, K.
Lachnospiraceae like pneumoniae)
E. coli, and decreased abundances of Bacteroides, Clostridiales, Faeca-
Roseburia and Blautia) • R. gnavus
libacterium, Roseburia, Blautia, Ruminococcus and Lachnospiraceae. It
• Christensenellaceae • Veillonellaceae
is interesting to note that microbial alterations were more marked in • Coriobacteriaceae • Pasteurellaceae
mucosal than fecal samples, and that sample composition was more • Bacteroides • Neisseriaceae
• Bifidobacterium • Actinomyces
• •
similar between ileum and rectum than between feces and mucosa.
Sutterella Fusobacteria
A systematic review of adult and pediatric case-control studies
• Akkermansia • Desulfovibrio
published since 2010 comparing the gut microbiota between IBD and • E. rectale
control cohorts was recently conducted [61]. The most striking finding Fungi [73] • S. cerevisiae • C.C. albicans
is the lack of consistency across studies: among 34 CD and 29 UC stu- • Ascomycota diversity • M. parapsilosis
• Basidiomycota
restricta
dies, no microbial changes were seen in more than three studies.
• Caudovirales diversity
Moreover, only four bacterial groups were consistently altered in three
studies. Small sample sizes and the sources of heterogeneity outlined
Viruses/Bacteriophages •
above were common limitations. Despite this lack of reproducibility,
the authors drew a few conclusions based on more consistent ob- rectale, uncultured Clostridiales and Vibrio species, and a lower abun-
servations. In CD, all three studies examining Christensenellaceae and dance of S. mitis were associated with a greater likelihood of response to
Coriobacteriaceae and six of 11 studies examining F. prausnitzii found a treatment with an anti-tumor necrosis factor-α (TNF-α) monoclonal
decrease, while two studies each of Actinomyces, Veillonella and E. coli antibody [77]. In a more recent study, disruptions of an association
found an increase in these microorganisms. In UC, Eubacterium rectale network including Lachnospiraceae and Ruminococcaceae (SCFA pro-
and Akkermansia were reduced in all three studies, and E. coli pro- ducers) were associated with frequently relapsing disease, a poor clin-
portions were increased in four of nine studies. Diversity was either ical response to anti-TNF-α and a higher risk of CD recurrence after
lower or not different in IBD patients compared to controls. Chris- surgical resection [78].
tensenellaceae and Eubacterium are Firmicutes and butyrate producers. While the focus to date has been on taxonomic classification, a far
Coriobacteriaceae, which belong to the Actinobacteria phylum, include more salient question to pose than “who is there” is “what are they
species such as Collinsella, Eggerthella, Slackia and Atopobium, of doing?” In other words, the functional repertoire of a microbial com-
which some have the ability to transform bile acids. This is pertinent as munity bears more relevance to host health than its composition. Multi-
bile acid metabolites influence both Th17 and Treg cell differentiation omics methods (i.e., metagenomics, metatranscriptomics, metabo-
[72]. lomics, metaproteomics) represent powerful tools to interrogate mi-
It is important to recall that bacteria are not the sole non-mam- crobial function. Using a multi-omics approach, Lloyd-Price and col-
malian inhabitants of the human gut. Fungi (the “mycobiome”) and leagues examined 132 pediatric and adult IBD patients serially over one
viruses (the “virome”) are present as well. Exploration of these elements year [79]. Molecular disruptions of transcription, among Clostridia for
of the human gut microbiota is still in its infancy, but data supporting example, and metabolite pools (SCFAs, bile acids and acylcarnitines)
dysbiosis at these levels are emerging. Studies support overgrowth of C. were apparent. Although the IBD group generally displayed decreased
albicans and C. parapsilosis, and decreased abundance of S. cerevisiae in microbiome stability over time, the relative abundances of Prevotella
IBD [73]. There is also enrichment of Caudovirales bacteriophages and copri remained stable in IBD patients. This is of interest given the link
loss of viral-bacterial correlations in the mucosa of UC patients [74]. between P. copri and inflammatory arthritis, particularly rheumatoid
Bringing together the above findings, Table 2 summarizes some of arthritis (RA) (considered below).
the more consistent dysbiotic changes observed in IBD. Fig. 2 illustrates
the disruption of microbial composition and related aberrant immune
responses in the setting of IBD. 5.4. Evidence relating to inflammatory arthropathies – animal studies
The discussion thus far has collapsed adult and pediatric studies, but
the pediatric population presents several important advantages, in- As is the case for colitis, support for the role of the microbiota in the
cluding temporal proximity to disease origin and less frequent co- pathogenesis of inflammatory arthritis derives from multiple observa-
morbidities, all of which minimize confounding. Table 1 presents a tions of absent arthritis in GF animal models. Examples include the IL-1
summary of the pediatric studies in the systematic review discussed, as receptor antagonist knockout mouse (IL1rn−/−), K/BxN T cell receptor
well as selected subsequent pediatric publications. Although most transgenic mouse, ANKylosing ENThesopathy (ANKENT) mouse, HLA-
findings are aligned with those presented above, it is interesting to note B27 transgenic rat and SKG mouse (point mutation in ZAP-70, a T cell
that the only two studies to demonstrate a numeric increase in F. transduction molecule), all of which develop arthritis under normal
prausnitzii abundance (rather than a decrease) were performed in chil- circumstances, but fail to do so when raised under GF conditions. In all
dren (though one was not statistically significant) [75,76]. Similarly, cases, inflammatory arthritis ensues upon reintroduction of specific
although most studies support a decrease in Bacteroides, two pediatric microbes [80–83]. The IL1rn−/− GF mouse displays dysbiosis char-
studies showed a decrease. Such findings raise the possibility of age- acterized by decreased richness and diversity, and depletion of Rumi-
dependent changes in the gut microbiota. nococcus and Prevotella. K/BxN T cell receptor transgenic mice develop
The potential utility of the gut microbiota as a prognostic and/or inflammatory arthritis due to T cell recognition of a self-antigen. Re-
predictive biomarker of disease course and treatment response is of colonization of the GF K/BxN T cell receptor transgenic mouse with SFB
great interest. In a study including 68 children with IBD, higher base- results in arthritis and reinstatement of Th17 cells in the gut lamina
line abundances of Bifidobacterium, Clostridium colinum, Eubacterium propria. The latter 3 models develop arthritis resembling SpA. In

7
A. Ricciuto, et al. Clinical Immunology 215 (2020) 108415

Fig. 2. Genetic, environmental and immune-mediated microbiome interactions in the pathogenesis of chronic inflammatory bowel diseases (IBD).
In health, the gut microbiota promotes homeostasis and anti-inflammatory processes. In the setting of appropriate genetic and environmental risk factors, dysbiotic
changes occur in IBD and contribute to aberrant immune responses by the innate and adaptive immune system. Reprinted from J Allergy Clin Immunol, 145(1), Glassner
et al., The microbiome and inflammatory bowel disease, 16–27, Copyright (2020), with permission from Elsevier.

humans, SpAs predominantly include ankylosing spondylitis (AS) and dependent manner [90].
psoriatic arthritis (PsA). SpAs are characterized by axial and peripheral Chronic recurrent multifocal osteomyelitis (CRMO) is an autoin-
asymmetric joint involvement, enthesitis and dactylitis. flammatory disorder characterized by bone lesions and fever. CRMO is
Interestingly, gut inflammation occurs frequently with SpA, with associated with pediatric IBD [91]. Mice homozygous for a mutation in
roughly 10–15% of SpA patients developing IBD. Conversely, up to 30% proline-serine-threonine phosphatase interacting protein 2 (Pstpip2),
of IBD patients manifest signs of inflammatory arthritis [84]. Even in termed Pstpip2cmo mice, develop similar manifestations [92]. Experi-
the absence of gastrointestinal symptoms, approximately half of SpA ments in these mice highlight the potential role of the microbiota in
patients have microscopic intestinal inflammation [85]. HLA-B27 CRMO. The critical cytokine for promoting disease in Pstpip2cmo mice is
transgenic rats exhibit a higher abundance of Paraprevotella and lower IL-1β. PSTPIP2 appears to negatively regulate IL-1β secretion in a ca-
Rikenellaceae [86]. The dysbiosis appears to be age-dependent, with a pase-1- and NLRP3 inflammasome-independent fashion and neu-
decrease in Firmicutes and increase in Proteobacteria and Akkermansia trophils, rather than macrophages, are the key innate immune cells
muciniphila, as well as IgA-coated bacteria, with age. These microbial implicated [93]. Interestingly, Pstpip2cmo mice fed a high fat diet (HFD)
changes are associated with increased expression of Th1 and Th17 are completely protected from osteomyelitis; this protection occurs in
proinflammatory cytokines, colonic Th17 expansion, dysregulated AMP conjunction with a microbial shift toward a greater ratio of Lactoba-
production and upregulation of bacterial-specific IgA [87]. Propionate cillus to Prevotella. Moreover, fecal transplantation from protected HFD
administration improves the arthritis in this model [88]. SKG mice re- mice to pre-disease, normal chow Pstpip2cmo mice is associated with
main arthritis-free when treated with antifungals and develop arthritis decreased Prevotella and IL-1β production, and is protective against
upon exposure to fungal elements, findings that highlight the im- clinical disease. Further support for the impact of the microbiota on IL-
portance of the mycobiome [89]. Transferability of phenotype with 1β derives from the finding that CD45+ cells from the colons of GF
microbial transplantation was demonstrated in this model: instillation mice express significantly lower levels of Il1b transcript than specific
of feces from RA patients (characterized by increased P. copri) into pathogen-free mice [94]. Collectively, these observations offer the
BALB/c ZAP-70 mutant mice provokes severe arthritis in a Th17- possibility that microbe-altering interventions could well have

8
A. Ricciuto, et al. Clinical Immunology 215 (2020) 108415

therapeutic implications for autoinflammatory disorders, like CRMO correspond to R. gnavus, as reported in IBD and adult inflammatory
[95].5.1.5 Evidence relating to inflammatory arthritis – Human studies. arthritis; and an increase in Bacteroides. The second theme is that ob-
It is challenging to reconcile microbiota findings in systemic in- servations in adults cannot necessarily be extrapolated to children. In
flammatory arthropathies as this umbrella captures several distinct other words, there may be an age-dependent effect for certain micro-
entities. Nevertheless, the following includes a few notable observations organisms, as already raised in 5.1.5. The surprisingly consistent in-
from adult studies. Firstly, compared to controls, Prevotella copri is crease in Bacteroides in the JIA studies contrasts with the majority of
present at higher abundance in new-onset RA, with a concomitant de- adult arthritis studies, which favour depletion, although, as already
crease in Bacteroides [96]. More recently, Zhou et al. also demonstrated acknowledged, this has not been a universal finding across all studies.
increased P. copri in an AS cohort [97]. In this latter study, P. copri Interestingly, one JIA study reported an increase in F. prausnitzii, rather
abundance fell with treatment and more significantly so in patients than a decrease, invoking the two pediatric IBD studies cited above that
receiving anti-TNF-α or disease-modifying anti-rheumatic drugs, rather similarly documented numerically higher F. prausnitzii abundances in
than non-steroidal anti-inflammatory agents only. In a study of RA and IBD patients than healthy controls. The authors of the JIA review also
SpA, the most striking finding was a 2- to 3-fold increase in the abun- highlight the finding of increased Akkermansia in the arthritis cohort of
dance of R. gnavus (a mucin degrader) in SpA compared to both healthy the 2014 study by Stoll et al., which again departs from the pre-
controls and RA patients [98]. Furthermore, R. gnavus correlated with ponderance of the evidence in adult cohorts. In this case, however,
SpA activity in patients with a history of IBD. A recent study by pediatric IBD studies are typically aligned with adult arthritis (and
Klingberg et al. also demonstrated increased R. gnavus in AS patients adult IBD) studies in suggesting depletion of Akkermansia. It is unclear
[99]. In this study, patients with elevated fecal calprotectin, a bio- whether this reduction in IBD is cause or consequence, the latter po-
marker of intestinal inflammation, had a lower abundance of bacteria tentially due to IBD-related depletion of goblet cell-derived mucins,
with anti-inflammatory effects, like F. prausnitzii and Clostridium, but which are the energy substrate for Akkermansia. Interestingly, there are
higher levels of Streptococcus. animal data to support that Akkermansia muciniphila may be deleterious
Proteobacteria are often enriched in patients with inflammatory in arthritis models, but beneficial in colitis models; this microorganism
arthropathies, including AS (E. coli and Shigella spp. increased) [99] was recently shown to be permissive to arthritis in the K/BxN mouse
and RA populations [98]. Akkermansia muciniphila, another mucin-de- [108],and to augment bacterial invasiveness [109], while ameliorating
grading bacterium, is frequently decreased; it was almost absent in a DSS colitis in mice [110].
PsA cohort [100], and present at lower abundances than controls in the
AS study by Zhou [97]. It should be noted, however, that not all studies 6. Microbe-altering therapies
have consistently supported this finding [99]. Reports have also been
conflicting about Bacteroides, with many showing depletion 6.1. Nutritional interventions
[96,99,101,102], but others enrichment [97,103]. In fact, in one study,
Bacteroides coprophilus was the AS-enriched bacterium with the stron- Numerous potential therapies exist to modulate the gut microbiota.
gest ability to identify AS (from controls) [97]. Zhou et al. used shotgun These include nutritional interventions, pre- and probiotics, oral anti-
metagenomics to investigate microbial function in AS; investigators biotics, as well as fecal microbial transplantation (FMT). Exclusive
identified three major pathways in the AS group, including oxidative enteral nutrition (EEN), the elimination of table food and substitution
phosphorylation, LPS biosynthesis and glycosaminoglycan degradation. with a polymeric or elemental formula for a several-week period, is an
Notably, the metabolism pathway of butanoate, a SCFA, was decreased effective induction treatment for pediatric CD [111]. Notably, there is a
in the AS group and there was a trend toward a decrease in propionate case report and a case series suggesting a similar beneficial effect of
metabolism as well. EEN in JIA [112,113]. Seemingly paradoxically, fecal microbial di-
The potential roles of many of these microorganisms in perpetuating versity appears to decline further with EEN, including a decrease in
or regulating inflammation are considered in the IBD section. In that organisms already depleted at baseline and thought to have anti-in-
vein, it is interesting to note several similarities between the dysbiotic flammatory properties, such as Faecalibacterium [114,115]. Such per-
changes described in IBD and arthritis populations, such as decreased F. plexing findings underscore our incomplete understanding of the
prausnitzii, and increased Proteobacteria and R. gnavus. There are also complex microbial processes at play in these chronic inflammatory
data suggesting that P. copri strains can contribute to human disease by disorders.
inciting chronic inflammation. Prevotella strains primarily act via TLR2
to stimulate APCs to produce Th17-polarizing cytokines. In addition, 6.2. FMT – animal studies
Prevotella strains stimulate ECs to produce IL-8, IL-6 and the chemo-
kine CCL20, which can trigger Th17 immune responses and neutrophil FMT, defined as the restoration of physiologic microbial community
recruitment [104]. Moreover, two RA-specific autoantigens that share by means of transplanting microbiota via feces, is an exciting emerging
sequence homology with P. copri are highly expressed in inflamed RA therapy. FMT has demonstrated efficacy for recurrent C. difficile infec-
synovium [105]. tion [116]. There are 4 randomized controlled trials (RCTs) and nu-
Specific consideration of pediatric studies is worthwhile here as merous case series in IBD, which support transient benefit [117–120].
well. JIA includes 7 subtypes: 1) systemic-onset; 2) persistent or ex- There are no studies of FMT for inflammatory arthritis.
tended oligoarthritis (≤4 joints); 3) Rheumatoid Factor (RF)-negative Animal experiments offer important insight into the immunologic
polyarthritis (≥5 joints); 4) RF-positive polyarthritis; 5) psoriatic; 6) mechanisms mediating the effects of FMT for treating colitis. In one
enthesitis-related arthritis (ERA); and 7) undifferentiated. ERA re- study, the gavage of mucus and feces from healthy mice into mice with
sembles adult SpA and RF-positive polyarthritis resembles RA. A very DSS colitis improved the colitis and was associated with an increase in
recent review indicates that, to date, there have been 7 studies on the IL-10 levels, the number of IL-10-producing CD4+ T cells and invariant
gut microbiota in JIA: 3 focus on children with oligo- or polyarticular NK (iNK) T cells, and a decrease in macrophages, neutrophils, ILC2s,
JIA; 3 focus on ERA; and 1 includes both oligo−/polyarticular JIA and ILC3s and MHC-II-expressing APCs in the lamina propria. There was
ERA [106]. Table 3 summarizes the findings of these studies. also a non-significant increase in FoxP3+ Tregs and upregulation of
As with IBD, despite substantial heterogeneity between JIA studies, AMPs. The increased IL-10 levels normalized when the inflammation
a few general themes have emerged. Firstly, a few microorganisms are resolved and IL-10 receptor blockade abrogated the effects, suggesting
fairly consistently altered in JIA. These include a decrease in F. praus- that the benefit of FMT is IL-10-dependent [121]. Lamina propria
nitzii, in line with reduced microbial genetic potential for butyrate mononuclear cells (LPMCs) exposed to FMT-derived microbiota gen-
production [107]; an increase in Ruminococcaceae, which may erated less TNF-α, IL-1β and IFN-γ in this study, and FMT was

9
A. Ricciuto, et al. Clinical Immunology 215 (2020) 108415

Table 3
Studies of the microbiota in juvenile idiopathic arthritis; adapted from [106].
Author Region Patients Treatment Method Depleted Enriched
Year

Stoll USA 25 ERA DMARDs Amp of V4 rRNA Faecalibacterium prausnitzii Bacteroides


2014 [140] 13 HC Illumina HiSeq, Lachnospiraceae Akkermansia
HiSeq
Large and statistically significant decreased abundance of Faecalibacterium prausnitzii (3.8% vs 10%), smaller decrease in unspecified Lachnospiraceae (7% vs 12%); 2 distinct clusters
on PCoA, one driven by Akkermansia muciniphila and the other by Bacteroides
Tejesvi Finland 13 oJIA, 15 RF- pJIA, 1 RF+, Treatment-naïve Amp of V4-V5 rRNA Firmicutes Bacteroides
2016 [141] pJIA, 1 ERA Ion Torrent
27 HC
At phylum level, relative depletion of Firmicutes and enrichment of Bacteroidetes; at genus level, increased Bacteroides (44% vs 33%)
Aggarwal India 33 ERA DMARD-naïve x Amp of V3 rRNA Prevotellaceae (trend) Bacteroides
2017 [142] 14 HC 6 weeks Illumina HiScan SQ Klebsiella
Strong depletion of Prevotellaceae family (Bacteroidetes phylum) (9% vs 66%); increased abundance of Bacteroides genus (3% vs 0.06%) and Klebsiella genus (82% vs 43%)
Di Paola Italy 10 pJIA, 19 ERA DMARDs V5-V6 Peptostreptococcaceae (ERA) Ruminococcaceae (both)
2016 [143] 29 HC Roche 454 Faecalibacterium (non-ERA) Veillonellaceae (ERA)
Decreased Peptostreptococcaceae (0.3% vs 1.1%); small, non-significant decrease in Faecalibacterium genus (0.2% vs 0.35%); increase in Veillonellaceae (1.4% vs 0.4%); both ERA and
non-ERA JIA showed increased Ruminococcaceae (22% ERA, 27% non-ERA vs 12%)
Muller Netherlands 8 RF- pJIA Treatment -naïve Phyla-specific Alistipes finegoldii Bacteroides fragilis
2017 [144] 22 HC primers; Sanger Prevotella multisacharivorax
PCoA revealed substantial differences in the composition of the Bacteroidetes phyla; at genus level, JIA patients had unspecified increased fecal B. fragilis and decreased Alistipes
finegoldii and Prevotella multisacharivorax
Stoll USA 24 ERA Treatment -naïve Amp of V4 rRNA; F. prausnitzii A1–165 Bacteroides fragilis
2018 [107] 19 HC WGS
Illumina MiSeq
Overall no difference in F. prausnitzii but regulatory strain decreased in ERA at expense of increased non-regulator L2/6 strain; increased B. fragilis (2% vs 0.45%); whole genome
sequencing showed decreased carriage of a pathway resulting in butyrate production
Dijkhuizen Italy, Netherlands Italy: Treatment -naïve V1-V3 Allobaculum Erysipelotrichaceae
2019 [145] 47 oJIA, 24 RF- pJIA, 2 RF+ Roche 454 Gemellaceae F. prausnitzii
pJIA, 3 psJIA, 2 U-JIA Propionibacterium acne Parabacteroides
79 HC Turicibacter Enterococcus
Netherlands: Ruminococcaceae
14 oJIA, 4 RF- pJIA, 2 ERA, 1
psJIA
28 HC

Substantial difference in age between patients and controls in Italian cohort (8 vs 3.9 y); random forest methods in Italian group separated patients and HC with near
100% accuracy (bacteria driving this were Erysipelotrichaceae, F. prausnitzii, Parabacteroides, Enterococcus and Ruminococcaceae (increased in JIA) and
Allobaculum, Gemellaceae, Propionibacterium acnes and Turicibacter (increased in HC); in Dutch cohort, random forest also successfully separated groups but no
single organism differed significantly between groups.
Amp – amplification; ATB – antibiotics; DMARD – disease-modifying anti-rheumatic drug; ERA – enthesitis-related arthritis; HC – healthy control; JIA – juvenile
idiopathic arthritis; o – oligoarticular; p – polyarticular; PCoA – principal coordinate analysis; ps – psoriatic; RF – rheumatoid factor.

associated with an increase in bacterial commensals, including SCFA- microorganisms (such as those belonging to Clostridium clusters IV and
producing taxa, such as Ruminococcaceae and Erysipelotrichaceae. Si- XIVa), which induce immunoregulatory Tregs and IL-10 [119,127,128].
milarly, in another DSS mouse colitis study, FMT led to improvement of In keeping with this finding, in the RCT by Moayyedi et al., the stool of
mucosal inflammation, along with recovery of Bifidobacterium and the donor associated with the highest response rate was significantly
Lactobacillus, as well as an increase in tryptophan [122]. There was enriched in Lachnospiraceae and Ruminococcaceae [120].
also an increase in the expression of IL-10, TGF-β and the ARh receptor,
for which tryptophan metabolites are ligands, as discussed in 4.1.2.
7. Conclusions

6.3. FMT – human studies The gut microbiota is intimately tied to human health and is central
to maintaining immunologic homeostasis. Evidence supporting an as-
Only limited human data exploring the immunologic consequences sociation between perturbations in the gut microbiome and various
of FMT are available. In a randomized controlled trial, peripheral blood human diseases, including IBD and JIA, continues to rapidly accrue.
mononuclear cells (PBMCs) showed a slight increase in gut-homing Despite substantial progress in this field, the specific underlying me-
CD4+ T cells following FMT [117]. Two smaller studies support an chanisms at play (and, at an even more fundamental level, whether
increase in colonic mucosal Tregs after a single FMT infusion, and a perturbations represent cause or consequence) continue to elude in-
decrease in Th1 cells [123,124]. In a pivotal study of 20 UC patients vestigators.
treated with a single FMT by colonoscopic infusion, a positive corre- Numerous unknowns remain to be elucidated. Whether local ma-
lation was seen between CD4+ T cell production of IFN-γ from rectal nipulation of the gut microbiota has the ability to affect and treat sys-
biopsies and the relative abundance of total gut virome [125]. Speci- temic autoimmune disorders (e.g., the use of enteral nutrition or FMT
fically, the bacteriophage Caudovirales, known to be increased in IBD for systemic diseases like JIA) remains to be established. Whether there
compared to healthy controls, was significantly enriched in FMT non- is a “window of opportunity” relative to disease onset beyond which
responders. such interventions are futile is also unclear. The focus of investigations
Generally, successful FMT is associated with increased alpha di- thus far has been on bacterial inhabitants of the gut; future work must
versity or richness and a shift in the recipient microbiota toward that of delineate whether other microbiota constituents (fungi, viruses) are
the donor [126]. The gut microbiota in FMT responders also con- important mediators of gut health and determine their role(s) in various
sistently demonstrates a shift toward butyrate-producing human diseases. Carefully conducted clinical trials with large enough

10
A. Ricciuto, et al. Clinical Immunology 215 (2020) 108415

sample sizes to discern statistical power are needed to more accurately engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-
determine the efficacy of microbe-altering therapeutic interventions. 22, Immunity 39 (2013) 372–385.
[20] K. Atarashi, J. Nishimura, T. Shima, et al., ATP drives lamina propria T(H)17 cell
Such studies must include longer follow-up duration to carefully assess differentiation, Nature 455 (2008) 808–812.
for potential adverse effects of microbial manipulation, particularly at [21] I.I. Ivanov, L. Frutos Rde, N. Manel, et al., Specific microbiota direct the differ-
early life stages. As yet, the long-term safety implications of interven- entiation of IL-17-producing T-helper cells in the mucosa of the small intestine,
Cell Host Microbe 4 (2008) 337–349.
tions like FMT are unknown. Heterogeneity between studies has been a [22] V. Gaboriau-Routhiau, S. Rakotobe, E. Lecuyer, et al., The key role of segmented
significant impediment to progress; accordingly, meticulous attention filamentous bacteria in the coordinated maturation of gut helper T cell responses,
and great effort should now be dedicated to standardizing methods. Immunity 31 (2009) 677–689.
[23] I.I. Ivanov, K. Atarashi, N. Manel, et al., Induction of intestinal Th17 cells by
Finally, the focus must shift from identification to functional character- segmented filamentous bacteria, Cell 139 (2009) 485–498.
ization; this will move the field from a cataloguing of which microbes [24] K. Atarashi, T. Tanoue, M. Ando, et al., Th17 cell induction by adhesion of mi-
are present in the gut, to a careful dissection of what functions various crobes to intestinal epithelial cells, Cell 163 (2015) 367–380.
[25] H. Zeng, H. Chi, Metabolic control of regulatory T cell development and function,
constituents of the intestinal microbiota are playing across the age
Trends Immunol. 36 (2015) 3–12.
continuum, in both health and various chronic diseases, including IBD [26] M. Sun, W. Wu, L. Chen, et al., Microbiota-derived short-chain fatty acids promote
and arthropathies. Only the latter approach will allow us to unravel the Th1 cell IL-10 production to maintain intestinal homeostasis, Nat. Commun. 9
intricate ties between the gut microbiota and human disease. (2018) 3555.
[27] Y. Furusawa, Y. Obata, S. Fukuda, et al., Commensal microbe-derived butyrate
induces the differentiation of colonic regulatory T cells, Nature 504 (2013)
Funding sources 446–450.
[28] G. Bakdash, L.T. Vogelpoel, T.M. van Capel, et al., Retinoic acid primes human
dendritic cells to induce gut-homing, IL-10-producing regulatory T cells, Mucosal
This review did not receive any specific grant from funding agencies Immunol. 8 (2015) 265–278.
in the public, commercial, or not-for-profit sectors. [29] L. Cervantes-Barragan, J.N. Chai, M.D. Tianero, et al., Lactobacillus reuteri in-
duces gut intraepithelial CD4(+)CD8alphaalpha(+) T cells, Science 357 (2017)
806–810.
Acknowledgments [30] H.M. Roager, T.R. Licht, Microbial tryptophan catabolites in health and disease,
Nat. Commun. 9 (2018) 3294.
PMS is the recipient of a Canada Research Chair in Gastrointestinal [31] N. Wilck, M.G. Matus, S.M. Kearney, et al., Salt-responsive gut commensal mod-
ulates TH17 axis and disease, Nature 551 (2017) 585–589.
Disease. [32] K. Atarashi, T. Tanoue, T. Shima, et al., Induction of colonic regulatory T cells by
indigenous Clostridium species, Science 331 (2011) 337–341.
References [33] K. Atarashi, T. Tanoue, K. Oshima, et al., Treg induction by a rationally selected
mixture of clostridia strains from the human microbiota, Nature 500 (2013)
232–236.
[1] S. Coward, F. Clement, E.I. Benchimol, et al., Past and future burden of in- [34] S. Dasgupta, D. Erturk-Hasdemir, J. Ochoa-Reparaz, et al., Plasmacytoid dendritic
flammatory bowel diseases based on modeling of population-based data, cells mediate anti-inflammatory responses to a gut commensal molecule via both
Gastroenterology 156 (5) (2019) 1345–1353, https://doi.org/10.1053/j.gastro. innate and adaptive mechanisms, Cell Host Microbe 15 (2014) 413–423.
2019.01.002 (e4, Epub 2019 Jan 10). [35] S.K. Mazmanian, J.L. Round, D.L. Kasper, A microbial symbiosis factor prevents
[2] E.I. Benchimol, C.N. Bernstein, A. Bitton, et al., Trends in epidemiology of pe- intestinal inflammatory disease, Nature 453 (2008) 620–625.
diatric inflammatory bowel disease in Canada: distributed network analysis of [36] J.L. Round, S.M. Lee, J. Li, et al., The toll-like receptor 2 pathway establishes
multiple population-based provincial health administrative databases, Am. J. colonization by a commensal of the human microbiota, Science 332 (2011)
Gastroenterol. 112 (7) (2017) 1120–1134, https://doi.org/10.1038/ajg.2017.97 974–977.
(Epub 2017 Apr 18). [37] H. Sokol, B. Pigneur, L. Watterlot, et al., Faecalibacterium prausnitzii is an anti-
[3] S.C. Ng, H.Y. Shi, N. Hamidi, et al., Worldwide incidence and prevalence of in- inflammatory commensal bacterium identified by gut microbiota analysis of Crohn
flammatory bowel disease in the 21st century: a systematic review of population- disease patients, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 16731–16736.
based studies, Lancet 390 (2018) 2769–2778. [38] R. Martin, F. Chain, S. Miquel, et al., The commensal bacterium Faecalibacterium
[4] M. Agrawal, S. Shah, A. Patel, et al., Changing epidemiology of immune-mediated prausnitzii is protective in DNBS-induced chronic moderate and severe colitis
inflammatory diseases in immigrants: a systematic review of population-based models, Inflamm. Bowel Dis. 20 (2014) 417–430.
studies, J. Autoimmun. 105 (2019) 102303. [39] M. Zhang, X. Qiu, H. Zhang, et al., Faecalibacterium prausnitzii inhibits inter-
[5] R. Sender, S. Fuchs, R. Milo, Are we really vastly outnumbered? Revisiting the leukin-17 to ameliorate colorectal colitis in rats, PLoS One 9 (2014) e109146.
ratio of bacterial to host cells in humans, Cell 164 (2016) 337–340. [40] M. Lopez-Siles, S.H. Duncan, L.J. Garcia-Gil, et al., Faecalibacterium prausnitzii:
[6] P.B. Eckburg, E.M. Bik, C.N. Bernstein, et al., Diversity of the human intestinal from microbiology to diagnostics and prognostics, ISME J. 11 (2017) 841–852.
microbial flora, Science 308 (2005) 1635–1638. [41] M. Ennamorati, C. Vasudevan, K. Clerkin, et al., Intestinal microbes influence
[7] W.G. Weisburg, S.M. Barns, D.A. Pelletier, et al., 16S ribosomal DNA amplification development of thymic lymphocytes in early life, Proc. Natl. Acad. Sci. U. S. A. 117
for phylogenetic study, J. Bacteriol. 173 (1991) 697–703. (2020) 2570–2578.
[8] J.H. Vineis, D.L. Ringus, H.G. Morrison, et al., Patient-specific bacteroides genome [42] K.L. Glassner, B.P. Abraham, E.M.M. Quigley, The microbiome and inflammatory
variants in pouchitis, mBio 7 (2016). bowel disease, J. Allergy Clin. Immunol. 145 (2020) 16–27.
[9] A.J. Kozik, C.H. Nakatsu, H. Chun, et al., Comparison of the fecal, cecal, and [43] A. Swidsinski, A. Ladhoff, A. Pernthaler, et al., Mucosal flora in inflammatory
mucus microbiome in male and female mice after TNBS-induced colitis, PLoS One bowel disease, Gastroenterology 122 (2002) 44–54.
14 (2019) e0225079. [44] Z. Al Nabhani, G. Dietrich, J.P. Hugot, et al., Nod2: the intestinal gate keeper, PLoS
[10] L.N. Dong, J.P. Wang, P. Liu, et al., Faecal and mucosal microbiota in patients with Pathog. 13 (2017) e1006177.
functional gastrointestinal disorders: correlation with toll-like receptor 2/toll-like [45] D.N. Frank, C.E. Robertson, C.M. Hamm, et al., Disease phenotype and genotype
receptor 4 expression, World J. Gastroenterol. 23 (2017) 6665–6673. are associated with shifts in intestinal-associated microbiota in inflammatory
[11] Structure, function and diversity of the healthy human microbiome, Nature 486 bowel diseases, Inflamm. Bowel Dis. 17 (2010) 179–184.
(2012) 207–214. [46] E. Li, C.M. Hamm, A.S. Gulati, et al., Inflammatory bowel diseases phenotype, C.
[12] C.S. Catana, I. Berindan Neagoe, V. Cozma, et al., Contribution of the IL-17/IL-23 difficile and NOD2 genotype are associated with shifts in human ileum associated
axis to the pathogenesis of inflammatory bowel disease, World J. Gastroenterol. 21 microbial composition, PLoS One 7 (2012) e26284.
(2015) 5823–5830. [47] A. Rehman, C. Sina, O. Gavrilova, et al., Nod2 is essential for temporal develop-
[13] L. Maggi, A. Mazzoni, R. Cimaz, et al., Th17 and Th1 lymphocytes in oligoarticular ment of intestinal microbial communities, Gut 60 (2011) 1354–1362.
juvenile idiopathic arthritis, Front. Immunol. 10 (2019) 450. [48] K.K. Gkouskou, C. Deligianni, C. Tsatsanis, et al., The gut microbiota in mouse
[14] T. Gensollen, S.S. Iyer, D.L. Kasper, et al., How colonization by microbiota in early models of inflammatory bowel disease, Front. Cell. Infect. Microbiol. 4 (2014) 28.
life shapes the immune system, Science 352 (2016) 539–544. [49] T. Hudcovic, R. Stepankova, J. Cebra, et al., The role of microflora in the devel-
[15] D. Mathis, C. Benoist, Microbiota and autoimmune disease: the hosted self, Cell opment of intestinal inflammation: acute and chronic colitis induced by dextran
Host Microbe 10 (2011) 297–301. sulfate in germ-free and conventionally reared immunocompetent and im-
[16] F. Fava, S. Danese, Intestinal microbiota in inflammatory bowel disease: friend of munodeficient mice, Folia Microbiol. (Praha) 46 (2001) 565–572.
foe? World J. Gastroenterol. 17 (2011) 557–566. [50] R.K. Sellon, S. Tonkonogy, M. Schultz, et al., Resident enteric bacteria are ne-
[17] A. Wittmann, P.A. Bron, van S, II, et al., TLR signaling-induced CD103-expressing cessary for development of spontaneous colitis and immune system activation in
cells protect against intestinal inflammation, Inflamm. Bowel Dis. 21 (2015) interleukin-10-deficient mice, Infect. Immun. 66 (1998) 5224–5231.
507–519. [51] E. Balish, T. Warner, Enterococcus faecalis induces inflammatory bowel disease in
[18] D. Chakravortty, N. Koide, Y. Kato, et al., The inhibitory action of butyrate on interleukin-10 knockout mice, Am. J. Pathol. 160 (2002) 2253–2257.
lipopolysaccharide-induced nitric oxide production in RAW 264.7 murine mac- [52] M. Schultz, C. Veltkamp, L.A. Dieleman, et al., Lactobacillus plantarum 299V in
rophage cells, J. Endotoxin Res. 6 (2000) 243–247. the treatment and prevention of spontaneous colitis in interleukin-10-deficient
[19] T. Zelante, R.G. Iannitti, C. Cunha, et al., Tryptophan catabolites from microbiota mice, Inflamm. Bowel Dis. 8 (2002) 71–80.

11
A. Ricciuto, et al. Clinical Immunology 215 (2020) 108415

[53] R. Aranda, B.C. Sydora, P.L. McAllister, et al., Analysis of intestinal lymphocytes in the gut microbiota of transgenic rats, PLoS One 9 (2014) e105684.
mouse colitis mediated by transfer of CD4+, CD45RBhigh T cells to SCID re- [87] M.J. Asquith, P. Stauffer, S. Davin, et al., Perturbed mucosal immunity and dys-
cipients, J. Immunol. 158 (1997) 3464–3473. biosis accompany clinical disease in a rat model of Spondyloarthritis, Arthritis
[54] F. Powrie, S. Mauze, R.L. Coffman, CD4+ T-cells in the regulation of inflammatory Rheum. 68 (2016) 2151–2162.
responses in the intestine, Res. Immunol. 148 (1997) 576–581. [88] M. Asquith, S. Davin, P. Stauffer, et al., Intestinal metabolites are profoundly al-
[55] Y. Cong, S.L. Brandwein, R.P. McCabe, et al., CD4+ T cells reactive to enteric tered in the context of HLA-B27 expression and functionally modulate disease in a
bacterial antigens in spontaneously colitic C3H/HeJBir mice: increased T helper rat model of spondyloarthritis, Arthritis Rheum. 69 (2017) 1984–1995.
cell type 1 response and ability to transfer disease, J. Exp. Med. 187 (1998) [89] H. Yoshitomi, N. Sakaguchi, K. Kobayashi, et al., A role for fungal {beta}-glucans
855–864. and their receptor Dectin-1 in the induction of autoimmune arthritis in genetically
[56] G.J. Britton, E.J. Contijoch, I. Mogno, et al., Microbiotas from humans with in- susceptible mice, J. Exp. Med. 201 (2005) 949–960.
flammatory bowel disease Alter the balance of gut Th17 and RORgammat(+) [90] Y. Maeda, T. Kurakawa, E. Umemoto, et al., Dysbiosis contributes to arthritis
regulatory T cells and exacerbate colitis in mice, Immunity 50 (2019) development via activation of autoreactive T cells in the intestine, Arthritis
212–224 (e4). Rheum. 68 (2016) 2646–2661.
[57] N.W. Palm, M.R. de Zoete, T.W. Cullen, et al., Immunoglobulin a coating identifies [91] G.K. Audu, K. Nikaki, D. Crespi, et al., Chronic recurrent multifocal osteomyelitis
colitogenic bacteria in inflammatory bowel disease, Cell 158 (2014) 1000–1010. and inflammatory bowel disease, J. Pediatr. Gastroenterol. Nutr. 60 (2015)
[58] M. Viladomiu, C. Kivolowitz, A. Abdulhamid, et al., IgA-coated E. coli enriched in 586–591.
Crohn's disease spondyloarthritis promote TH17-dependent inflammation, Sci. [92] J. Grosse, V. Chitu, A. Marquardt, et al., Mutation of mouse Mayp/Pstpip2 causes a
Transl. Med. (2017) 9. macrophage autoinflammatory disease, Blood 107 (2006) 3350–3358.
[59] P.H. Harper, E.C. Lee, M.G. Kettlewell, et al., Role of the faecal stream in the [93] S.L. Cassel, J.R. Janczy, X. Bing, et al., Inflammasome-independent IL-1β mediates
maintenance of Crohn's colitis, Gut 26 (1985) 279–284. autoinflammatory disease in Pstpip2-deficient mice, Proc. Natl. Acad. Sci. 111
[60] P. Rutgeerts, M. Peeters, M. Hiele, et al., Effect of faecal stream diversion on re- (2014) 1072–1077.
currence of Crohn's disease in the neoterminal ileum, Lancet 338 (1991) 771–774. [94] J.R. Lukens, J.M. Gross, C. Calabrese, et al., Critical role for inflammasome-in-
[61] R. Pittayanon, J.T. Lau, G.I. Leontiadis, et al., Differences in gut microbiota in dependent IL-1beta production in osteomyelitis, Proc. Natl. Acad. Sci. U. S. A. 111
patients with vs without inflammatory bowel diseases: a systematic review, (2014) 1066–1071.
Gastroenterology 158 (4) (2020) 930–946, https://doi.org/10.1053/j.gastro. [95] P.J. Ferguson, R.M. Laxer, New discoveries in CRMO: IL-1beta, the neutrophil, and
2019.11.294 (e1, Epub 2019 Dec 5). the microbiome implicated in disease pathogenesis in Pstpip2-deficient mice,
[62] J. Qin, R. Li, J. Raes, et al., A human gut microbial gene catalogue established by Semin. Immunopathol. 37 (2015) 407–412.
metagenomic sequencing, Nature 464 (2010) 59–65. [96] J.U. Scher, A. Sczesnak, R.S. Longman, et al., Expansion of intestinal Prevotella
[63] J. Halfvarson, C.J. Brislawn, R. Lamendella, et al., Dynamics of the human gut copri correlates with enhanced susceptibility to arthritis, Elife 2 (2013) e01202.
microbiome in inflammatory bowel disease, Nat. Microbiol. 2 (2017) 17004. [97] C. Zhou, H. Zhao, X.Y. Xiao, et al., Metagenomic profiling of the pro-inflammatory
[64] X.C. Morgan, T.L. Tickle, H. Sokol, et al., Dysfunction of the intestinal microbiome gut microbiota in ankylosing spondylitis, J. Autoimmun. 102360 (2019).
in inflammatory bowel disease and treatment, Genome Biol. 13 (2012) R79. [98] M. Breban, J. Tap, A. Leboime, et al., Faecal microbiota study reveals specific
[65] K. Machiels, M. Joossens, J. Sabino, et al., A decrease of the butyrate-producing dysbiosis in spondyloarthritis, Ann. Rheum. Dis. 76 (2017) 1614–1622.
species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in [99] E. Klingberg, M.K. Magnusson, H. Strid, et al., A distinct gut microbiota compo-
patients with ulcerative colitis, Gut 63 (2014) 1275–1283. sition in patients with ankylosing spondylitis is associated with increased levels of
[66] A. Basson, A. Trotter, A. Rodriguez-Palacios, et al., Mucosal interactions between fecal calprotectin, Arthritis. Res. Ther. 21 (2019) 248.
genetics, diet, and microbiome in inflammatory bowel disease, Front. Immunol. 7 [100] J.U. Scher, C. Ubeda, A. Artacho, et al., Decreased bacterial diversity characterizes
(2016) 290. the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis
[67] R.B. Sartor, G.D. Wu, Roles for intestinal bacteria, viruses, and fungi in patho- in inflammatory bowel disease, Arthritis Rheum. 67 (2015) 128–139.
genesis of inflammatory bowel diseases and therapeutic approaches, [101] S. Stebbings, K. Munro, M.A. Simon, et al., Comparison of the faecal microflora of
Gastroenterology 152 (2017) 327–339.e4. patients with ankylosing spondylitis and controls using molecular methods of
[68] N.C. Knox, J.D. Forbes, C.L. Peterson, et al., The gut microbiome in inflammatory analysis, Rheumatology (Oxford) 41 (2002) 1395–1401.
bowel disease: lessons learned from other immune-mediated inflammatory dis- [102] R.Y. Tito, H. Cypers, M. Joossens, et al., Brief report: dialister as a microbial
eases, Am. J. Gastroenterol. 114 (2019) 1051–1070. marker of disease activity in spondyloarthritis, Arthritis Rheum. 69 (2017)
[69] A. Darfeuille-Michaud, J. Boudeau, P. Bulois, et al., High prevalence of adherent- 114–121.
invasive Escherichia coli associated with ileal mucosa in Crohn's disease, [103] M.E. Costello, F. Ciccia, D. Willner, et al., Brief report: intestinal dysbiosis in an-
Gastroenterology 127 (2004) 412–421. kylosing spondylitis, Arthritis Rheum. 67 (2015) 686–691.
[70] J.D. Forbes, G. Van Domselaar, C.N. Bernstein, The gut microbiota in immune- [104] J.M. Larsen, The immune response to Prevotella bacteria in chronic inflammatory
mediated inflammatory diseases, Front. Microbiol. 7 (2016) 1081. disease, Immunology 151 (2017) 363–374.
[71] D. Gevers, S. Kugathasan, L.A. Denson, et al., The treatment-naive microbiome in [105] A. Pianta, S.L. Arvikar, K. Strle, et al., Two rheumatoid arthritis-specific auto-
new-onset Crohn's disease, Cell Host Microbe 15 (2014) 382–392. antigens correlate microbial immunity with autoimmune responses in joints, J.
[72] S. Hang, D. Paik, L. Yao, et al., Bile acid metabolites control TH17 and Treg cell Clin. Invest. 127 (2017) 2946–2956.
differentiation, Nature 576 (2019) 143–148. [106] M. Arvonen, P. Vanni, A.N. Sarangi, et al., Microbial orchestra in juvenile idio-
[73] X.V. Li, I. Leonardi, I.D. Iliev, Gut Mycobiota in immunity and inflammatory pathic arthritis: sounds of disarray? Immunol. Rev. 294 (2020) 9–26, https://doi.
disease, Immunity 50 (2019) 1365–1379. org/10.1111/imr.12826.
[74] T. Zuo, X.J. Lu, Y. Zhang, et al., Gut mucosal virome alterations in ulcerative [107] M.L. Stoll, P.F. Weiss, J.E. Weiss, et al., Age and fecal microbial strain-specific
colitis, Gut 68 (2019) 1169–1179. differences in patients with spondyloarthritis, Arthritis. Res. Ther. 20 (2018) 14.
[75] A. Assa, J. Butcher, J. Li, et al., Mucosa-associated Ileal microbiota in new-onset [108] M.L. Stoll, M.K. Pierce, J.A. Watkins, et al., Akkermansia muciniphila is permissive
pediatric Crohn's disease, Inflamm. Bowel Dis. 22 (2016) 1533–1539. to arthritis in the K/BxN mouse model of arthritis, Genes Immun. 20 (2019)
[76] R. Shah, J.L. Cope, D. Nagy-Szakal, et al., Composition and function of the pe- 158–166.
diatric colonic mucosal microbiome in untreated patients with ulcerative colitis, [109] B.P. Ganesh, R. Klopfleisch, G. Loh, et al., Commensal Akkermansia muciniphila
Gut Microbes 7 (2016) 384–396. exacerbates gut inflammation in Salmonella Typhimurium-infected gnotobiotic
[77] K.L. Kolho, K. Korpela, T. Jaakkola, et al., Fecal microbiota in pediatric in- mice, PLoS One 8 (2013) e74963.
flammatory bowel disease and its relation to inflammation, Am. J. Gastroenterol. [110] X. Bian, W. Wu, L. Yang, et al., Administration of Akkermansia muciniphila
110 (2015) 921–930. ameliorates dextran sulfate sodium-induced ulcerative colitis in mice, Front.
[78] B. Yilmaz, P. Juillerat, O. Oyas, et al., Microbial network disturbances in relapsing Microbiol. 10 (2019) 2259.
refractory Crohn's disease, Nat. Med. 25 (2019) 323–336. [111] D.R. Mack, E.I. Benchimol, J. Critch, et al., Canadian Association of
[79] J. Lloyd-Price, C. Arze, A.N. Ananthakrishnan, et al., Multi-omics of the gut mi- Gastroenterology clinical practice guideline for the medical management of pe-
crobial ecosystem in inflammatory bowel diseases, Nature 569 (2019) 655–662. diatric Luminal Crohn's disease, Gastroenterology 157 (2019) 320–348.
[80] S. Abdollahi-Roodsaz, L.A. Joosten, M.I. Koenders, et al., Stimulation of TLR2 and [112] L. Berntson, Anti-inflammatory effect by exclusive enteral nutrition (EEN) in a
TLR4 differentially skews the balance of T cells in a mouse model of arthritis, J. patient with juvenile idiopathic arthritis (JIA): brief report, Clin. Rheumatol. 33
Clin. Invest. 118 (2008) 205–216. (2014) 1173–1175.
[81] R. Rogier, T.H.A. Ederveen, J. Boekhorst, et al., Aberrant intestinal microbiota due [113] L. Berntson, I. Hedlund-Treutiger, K. Alving, Anti-inflammatory effect of exclusive
to IL-1 receptor antagonist deficiency promotes IL-17- and TLR4-dependent ar- enteral nutrition in patients with juvenile idiopathic arthritis, Clin. Exp.
thritis, Microbiome 5 (2017) 63. Rheumatol. 34 (2016) 941–945.
[82] I. Matsumoto, A. Staub, C. Benoist, et al., Arthritis provoked by linked T and B cell [114] K. Gerasimidis, M. Bertz, L. Hanske, et al., Decline in presumptively protective gut
recognition of a glycolytic enzyme, Science 286 (1999) 1732–1735. bacterial species and metabolites are paradoxically associated with disease im-
[83] H.J. Wu, I.I. Ivanov, J. Darce, et al., Gut-residing segmented filamentous bacteria provement in pediatric Crohn's disease during enteral nutrition, Inflamm. Bowel
drive autoimmune arthritis via T helper 17 cells, Immunity 32 (2010) 815–827. Dis. 20 (2014) 861–871.
[84] C. López-Medina, A. Moltó, Update on the epidemiology, risk factors, and disease [115] C. Quince, U.Z. Ijaz, N. Loman, et al., Extensive modulation of the fecal meta-
outcomes of axial spondyloarthritis, Best Pract. Res. Clin. Rheumatol. 32 (2018) genome in children with Crohn's disease during exclusive enteral nutrition, Am. J.
241–253. Gastroenterol. 110 (2015) 1718–1729 (quiz 1730).
[85] E. Gilis, C. Mortier, K. Venken, et al., The role of the microbiome in gut and joint [116] B.H. Mullish, M.N. Quraishi, J.P. Segal, et al., The use of faecal microbiota
inflammation in psoriatic arthritis and Spondyloarthritis, J. Rheumatol. Suppl. 94 transplant as treatment for recurrent or refractory Clostridium difficile infection
(2018) 36–39. and other potential indications: joint British Society of Gastroenterology (BSG)
[86] P. Lin, M. Bach, M. Asquith, et al., HLA-B27 and human beta2-microglobulin affect and Healthcare Infection Society (HIS) guidelines, Gut 67 (2018) 1920–1941.

12
A. Ricciuto, et al. Clinical Immunology 215 (2020) 108415

[117] S.P. Costello, P.A. Hughes, O. Waters, et al., Effect of fecal microbiota transplan- the microbiota of children with newly diagnosed Crohn's disease, Clin. Transl.
tation on 8-week remission in patients with ulcerative colitis: a randomized clin- Gastroenterol. 6 (2015) e71.
ical trial, Jama 321 (2019) 156–164. [132] R.L. Knoll, K. Forslund, J.R. Kultima, et al., Gut microbiota differs between chil-
[118] S. Paramsothy, M.A. Kamm, N.O. Kaakoush, et al., Multidonor intensive faecal dren with inflammatory bowel disease and healthy siblings in taxonomic and
microbiota transplantation for active ulcerative colitis: a randomised placebo- functional composition: a metagenomic analysis, Am. J. Physiol. Gastrointest.
controlled trial, Lancet 389 (2017) 1218–1228. Liver Physiol. 312 (2017) G327–G339.
[119] N.G. Rossen, S. Fuentes, M.J. van der Spek, et al., Findings from a randomized [133] J. Maukonen, K.L. Kolho, M. Paasela, et al., Altered fecal microbiota in paediatric
controlled trial of fecal transplantation for patients with ulcerative colitis, inflammatory bowel disease, J. Crohns. Colitis. 9 (2015) 1088–1095.
Gastroenterology 149 (2015) 110–118.e4. [134] Y. Wang, X. Gao, A. Ghozlane, et al., Characteristics of Faecal microbiota in
[120] P. Moayyedi, M.G. Surette, P.T. Kim, et al., Fecal microbiota transplantation in- paediatric Crohn's disease and their dynamic changes during infliximab therapy, J.
duces remission in patients with active ulcerative colitis in a randomized con- Crohns. Colitis. 12 (2018) 337–346.
trolled trial, Gastroenterology 149 (2015) 102–109.e6. [135] S. Sila, M. Jelic, I. Trivic, et al., Altered gut microbiota is present in newly diag-
[121] C. Burrello, F. Garavaglia, F.M. Cribiu, et al., Therapeutic faecal microbiota nosed pediatric patients with inflammatory bowel disease, J. Pediatr.
transplantation controls intestinal inflammation through IL10 secretion by im- Gastroenterol. Nutr. 70 (2020) 497–502.
mune cells, Nat. Commun. 9 (2018) 5184. [136] K. Kowalska-Duplaga, T. Gosiewski, P. Kapusta, et al., Differences in the intestinal
[122] Y.L. Wei, Y.Q. Chen, H. Gong, et al., Fecal microbiota transplantation ameliorates microbiome of healthy children and patients with newly diagnosed Crohn's dis-
experimentally induced colitis in mice by upregulating AhR, Front. Microbiol. 9 ease, Sci. Rep. 9 (2019) 18880.
(2018) 1921. [137] S. Michail, M. Durbin, D. Turner, et al., Alterations in the gut microbiome of
[123] V. Jacob, C. Crawford, S. Cohen-Mekelburg, et al., Single delivery of high-diversity children with severe ulcerative colitis, Inflamm. Bowel Dis. 18 (2012) 1799–1808.
fecal microbiota preparation by colonoscopy is safe and effective in increasing [138] S. Sila, M. Jelic, I. Trivic, et al., Altered gut microbiota is present in newly diag-
microbial diversity in active ulcerative colitis, Inflamm. Bowel Dis. 23 (2017) nosed pediatric patients with inflammatory bowel disease, J. Pediatr.
903–911. Gastroenterol. Nutr. 70 (4) (2020) 497–502, https://doi.org/10.1097/MPG.
[124] B.P. Vaughn, T. Vatanen, J.R. Allegretti, et al., Increased intestinal microbial di- 0000000000002611.
versity following fecal microbiota transplant for active Crohn's disease, Inflamm. [139] D. Turner, J. Bishai, L. Reshef, et al., Antibiotic cocktail for pediatric acute severe
Bowel Dis. 22 (2016) 2182–2190. colitis and the microbiome: the PRASCO randomized controlled trial, Inflamm.
[125] L. Gogokhia, K. Buhrke, R. Bell, et al., Expansion of bacteriophages is linked to Bowel Dis. (2019), https://doi.org/10.1093/ibd/izz298 (pii: izz298, Epub ahead
aggravated intestinal inflammation and colitis, Cell Host Microbe 25 (2019) of print).
285–299.e8. [140] M.L. Stoll, R. Kumar, C.D. Morrow, et al., Altered microbiota associated with
[126] M.N. Quraishi, W. Shaheen, Y.H. Oo, et al., Immunological mechanisms under- abnormal humoral immune responses to commensal organisms in enthesitis-re-
pinning faecal microbiota transplantation for the treatment of inflammatory bowel lated arthritis, Arthritis. Res. Ther. 16 (2014) 486.
disease, Clin. Exp. Immunol. 199 (2019) 24–38. [141] M.V. Tejesvi, M. Arvonen, S.M. Kangas, et al., Faecal microbiome in new-onset
[127] S. Paramsothy, S. Nielsen, M.A. Kamm, et al., Specific bacteria and metabolites juvenile idiopathic arthritis, Eur. J. Clin. Microbiol. Infect. Dis. 35 (2016)
associated with response to fecal microbiota transplantation in patients with ul- 363–370.
cerative colitis, Gastroenterology 156 (2019) 1440–1454.e2. [142] A. Aggarwal, A.N. Sarangi, P. Gaur, et al., Gut microbiome in children with en-
[128] S. Paramsothy, R. Paramsothy, D.T. Rubin, et al., Faecal microbiota transplanta- thesitis-related arthritis in a developing country and the effect of probiotic ad-
tion for inflammatory bowel disease: a systematic review and meta-analysis, J. ministration, Clin. Exp. Immunol. 187 (2017) 480–489.
Crohns. Colitis. 11 (2017) 1180–1199. [143] M. Di Paola, D. Cavalieri, D. Albanese, et al., Alteration of fecal microbiota profiles
[129] T. Aomatsu, H. Imaeda, T. Fujimoto, et al., Terminal restriction fragment length in juvenile idiopathic arthritis. Associations with HLA-B27 allele and disease
polymorphism analysis of the gut microbiota profiles of pediatric patients with status, Front. Microbiol. 7 (2016) 1703.
inflammatory bowel disease, Digestion 86 (2012) 129–135. [144] P. Hissink Muller, T. Meij, M. Westedt, et al., Disturbance of microbial Core species
[130] U.Z. Ijaz, C. Quince, L. Hanske, et al., The distinct features of microbial 'dysbiosis' in new-onset juvenile idiopathic arthritis, J. Pediatr. Infect. Dis. 12 (2017).
of Crohn's disease do not occur to the same extent in their unaffected, genetically- [145] E.H.P. van Dijkhuizen, F. Del Chierico, C. Malattia, et al., Microbiome analytics of
linked kindred, PLoS One 12 (2017) e0172605. the gut microbiota in patients with juvenile idiopathic arthritis: a longitudinal
[131] N.O. Kaakoush, A.S. Day, S.T. Leach, et al., Effect of exclusive enteral nutrition on observational cohort study, Arthritis Rheum. 71 (2019) 1000–1010.

13

You might also like