Nature versus nurture in the spectrum of rheumatic diseases: Classification of spondyloarthritis as autoimmune or autoinflammatory

Nature versus nurture in the spectrum of rheumatic diseases: Classification of spondyloarthritis as autoimmune or autoinflammatory

Accepted Manuscript Nature versus nurture in the spectrum of rheumatic diseases: Classification of spondyloarthritis as autoimmune or autoinflammatory...

2MB Sizes 0 Downloads 13 Views

Accepted Manuscript Nature versus nurture in the spectrum of rheumatic diseases: Classification of spondyloarthritis as autoimmune or autoinflammatory

Elena Generali, Tanima Bose, Carlo Selmi, J. Willem Voncken, Jan G.M.C. Damoiseaux PII: DOI: Reference:

S1568-9972(18)30163-0 doi:10.1016/j.autrev.2018.04.002 AUTREV 2194

To appear in:

Autoimmunity Reviews

Received date: Accepted date:

5 April 2018 10 April 2018

Please cite this article as: Elena Generali, Tanima Bose, Carlo Selmi, J. Willem Voncken, Jan G.M.C. Damoiseaux , Nature versus nurture in the spectrum of rheumatic diseases: Classification of spondyloarthritis as autoimmune or autoinflammatory. Autrev (2018), doi:10.1016/j.autrev.2018.04.002

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Nature versus nurture in the spectrum of rheumatic diseases: Classification of spondyloarthritis as autoimmune or autoinflammatory

Running title: Is spondyloarthritis an autoimmune or autoinflammatory disease?

SC

RI

and Jan G.M.C. Damoiseaux6*

PT

Elena Generali1, Tanima Bose2,3*, Carlo Selmi1,4, J. Willem Voncken5,

1

Rheumatology and Clinical Immunology, Humanitas Research Hospital, Rozzano, Italy, 2

3

NU

Leibniz Institute for Neurobiology, Magdeburg, Germany; Institute for Clinical

Neuroimmunology, Ludwigs-Maximilian-University, Munich; 4 BIOMETRA Department,

MA

University of Milan, Italy; 5 Molecular Genetics, Maastricht University Medical Center, Maastricht, The Netherlands; 6 Cellular Diagnostics & Cell Therapy, Central Diagnostic

PT E

D

Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands

CE

*Corresponding authors: E-mail: [email protected]; Tel: +31 43-3876655.

Abbreviations: AAV, ANCA-associated vasculitis; ACPA, anti-citrullinated protein

AC

antibodies; AID, autoimmune disease; AIM, autoimmune myopathies; AInfD, autoinflammatory disease; AS, ankylosing spondylitis; DZ, dizygotic; ER, endoplasmic reticulum; FMF, familial Mediterranean fever; GWAS, genome wide association study; HLA, human leukocyte antigen; HR, hazard ratio; IBD, inflammatory bowel disease; MZ, monozygotic; PsA, psoriatic arthritis; RA, rheumatoid arthritis; ReA, reactive arthritis; SLE, systemic lupus erythematosus; SpA, spondyloarthritides; TRAPS: tumor necrosis factor receptor associated autoinflammatory periodic syndrome

1

ACCEPTED MANUSCRIPT Abstract Spondyloarthritides (SpA) include inflammatory joint diseases with various clinical phenotypes that may also include the axial skeleton and/or entheses. SpA include psoriatic arthritis, reactive arthritis, enteropathic arthritis and ankylosing spondylitis; the latter is frequently associated with extra-articular manifestations, such as uveitis, psoriasis, and

PT

inflammatory bowel disease. SpA are associated with the HLA-B27 allele and recognize T cells as key pathogenetic players. In contrast to other rheumatic diseases, SpA affect women

RI

and men equally and are not associated with detectable serum autoantibodies. In addition,

SC

but opposite to rheumatoid arthritis, SpA are responsive to treatment regimens including IL23 or IL-17-targeting biologics, yet are virtually unresponsive to steroid treatment. Based on

NU

these differences with prototypical autoimmune diseases, such as rheumatoid arthritis or connective tissue diseases, SpA may be better classified among autoinflammatory diseases,

MA

with a predominant innate immunity involvement. This would rank SpA closer to gouty arthritis and periodic fevers in the spectrum of rheumatic diseases, as opposed to

D

autoimmune-predominant diseases. We herein provide available literature on risk factors

environmental factors.

PT E

associated with SpA in support of this hypothesis with a specific focus on genetic and

CE

Keywords: genetics; rheumatoid arthritis; ankylosing spondylitis; inflammasome;

AC

autoantibody; psoriatic arthritis.

Take home messages: 

Spondyloarthritis are “mixed-pattern diseases”, ranked in between autoimmune and autoinflammatory diseases.



Genetics, in particular HLA-B27, play a major role in spondyloarthritis pathogenesis, however, contrarily to autoinflammatory diseases, no IL-1 gene cluster has been associated with spondyloarthritis.

2

ACCEPTED MANUSCRIPT Environmental factors and microbiota play a crucial role in spondyloarthritis

CE

PT E

D

MA

NU

SC

RI

PT

development, however no single pathogenic factor has been identified.

AC



3

ACCEPTED MANUSCRIPT Introduction Systemic rheumatic diseases comprise a group of heterogeneous conditions affecting a wide range of tissues and organs, i.e. joints, eyes, skin, vascular system of lung and/or kidney, and present with significant healthcare costs and increased mortality and disability. Among such diverse clinical manifestations, symptoms and serological profiles of the rheumatic

PT

diseases, not all conditions can be classified as autoimmune [1]. Autoimmune diseases (AID) are characterized by a striking female preponderance, by the presence of serum

RI

autoantibodies detected sometimes years before the development of clinical manifestations

SC

[2], and by B or T cell selection defects characterized by aberrant responses to autoantigens [3]. Also, AID fulfill Koch’s postulates adapted for autoimmunity [4], borrowed from the field of

NU

microbiology, suggesting that autoantibodies and/or autoreactive T cells are the drivers of autoimmunity: passive transfer of these autoimmune factors to susceptible animals can result

MA

in the induction of the AID [5]. Rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) represent the paradigms of rheumatic AID. Conversely, autoinflammatory diseases

D

(AInfD) have no detectable autoantibodies, are characterized by a strong activation of the

PT E

innate immune system, and fever is the most common symptom [6]. Gout and Behçet disease are paradigmatic AInfD, along with periodic fevers [7]. However, a substantial number of systemic rheumatic diseases neither show full-overlap with

CE

common features of AID nor of AInfD, as in the case for spondyloarthritis (SpA) (Figure 1).

AC

SpA comprise a heterogeneous group of inflammatory joint diseases, including psoriatic arthritis (PsA), ankylosing spondylitis (AS), reactive arthritis (ReA), arthritis associated with inflammatory bowel disease (IBD) and undifferentiated SpA. While PsA can be considered an extended variant of psoriasis [8], also undifferentiated SpA is debated with respect to being a discrete entity [9].SpA affects both sexes with equal frequency, albeit with different clinical manifestations: e.g. axial involvement being more frequent in men [10]. The incidence rates of SpA vary between 0.48 to 63 per 100,000 individuals, whereas the prevalence ranges from 0.01 to 2.5%. SpA is not considered to be typically associated with specific autoantibodies, although several studies report on the presence of autoantibodies [11]. In

4

ACCEPTED MANUSCRIPT PsA, for instance, autoantibodies to carbamylated proteins (anti-CarP) and to anti-PsA peptides have been reported [12-14]. Pathogenecity of these autoantibodies has not been established, but they may add up to the few non-imaging biomarkers that are available [15] [16, 17]. Importantly, SpA was recently suggested to be a “mixed-pattern diseases”, ranked in between AID and AInfD along a continuum of immunological diseases, classified on the

PT

basis of clinical prominence and characteristics of the pathology [18]. In contrast, SpA has been considered as an AInfD by others, based on the underlying mechanisms and response

RI

to therapy [19]. Indeed, although both RA as well as SpA have a good response to anti-TNF

SC

drugs, there seem to be some apparent differences in terms of adverse events and drug survival [20] In this review, we aim to discuss the most recent insights regarding the

NU

pathogenesis of SpA in light of the ongoing discussion whether SpA should be considered as

MA

AID or AInfD.

D

The genetic perspective

PT E

Genetics plays an important role in SpA pathogenesis, as exemplified by the strong association of SpA with the human leukocyte antigen (HLA)-B27 allele, which is present in 85-90% of AS cases [21, 22]. Moreover, other HLA alleles, especially HLA-C*06, B*39 and

CE

B*07 have been associated with PsA. HLA-B27 is thought to have a direct pathogenic role in SpA development and various molecular mechanisms have been proposed, mostly related to

AC

intracellular protein misfolding of this class I surface antigen [23]. Accumulation of nonconventional forms of HLA-B27, such as free heavy chains, was recently reported in the gut and synovial tissue of SpA patients, as well as in HLA-B27 transgenic rats [24]. In vitro and in vivo animal studies show that slow folding of HLA-B27 and its association with ß2microglobulin causes accumulation of misfolded protein in the endoplasmic reticulum (ER) which triggers the unfolded protein response, a pro-inflammatory ER stress response [2527], which induces the expression of IL-23 [28]. In human dendritic cells, ER stress was also shown to increase IL-23 production after toll-like receptor stimulation [29].

5

ACCEPTED MANUSCRIPT Nonetheless, HLA accounts only for 40% of the genetic risk for SpA; other polymorphisms in non-HLA genes, involved in innate immune recognition and cytokine signaling pathways, are linked with SpA [30]. Such genes include tumor necrosis factor (TNF) and IL-23 [19], which are shared with IBD and psoriasis [31]. On the other hand, thus far no IL-1 gene cluster mutations have been associated with SpA [32, 33], whereas this locus is clearly implicated in

PT

AInfD [34]; this observation argues against the definition of SpA as an AInfD. Albeit that genetics plays a major role in SpA development, additional factors are required to

RI

trigger the disease. In this context, twin studies represent an excellent model to study the

SC

role of environmental factors. In particular, as monozygotic (MZ) twins share an identical genetic background, diseases with high concordance rates between MZ twins suggest a

NU

genetic predisposition, while low concordance rates support the role of environmental factors as part of a multifactorial origin of disease [35]. The caveat of this assumption, however, is

MA

that generation of the T cell repertoire is, to a certain extent, a stochastic process. As such, MZ twins may develop with distinct T cell repertoires. In SpA, relatively few twin-based

D

observations are available. Nevertheless, near identical concordance rates for PsA in MZ

PT E

and dizygotic (DZ) twins stress the importance to continue the search for non-genetic effectors in PsA [36]. In AS, conversely, genetic factors seem to exert a more prominent role, as MZ twins have higher concordance rates (±75%), compared to DZ twins (±15%) [37, 38].

CE

Several candidate gene variants have been identified by genetic mapping in RA. A

AC

predominant association with the HLA-DRB1 locus was identified decades ago, however recent advances in genome-wide association studies (GWAS) and subsequent metaanalyses have led to the identification of numerous alleles that govern RA susceptibility, with variations according to the population evaluated [39]. In addition to HLA loci, more than 100 common variants in non-HLA loci have been implicated in RA susceptibility [39]. Several polymorphisms identified in RA are involved in immunoregulatory mechanisms, for instance polymorphisms in signaling transducers and activators of transcription-4 (STAT4) and IL-10 genes, and are shared with other prototypic AID [40]. Concordance rates for RA in MZ twins, however, are lower than observed in other AID, approximately around 15% in MZ and 4% in

6

ACCEPTED MANUSCRIPT DZ twins [35]. Obviously, RA as well as most other AID cannot be explained by genetic susceptibility alone. With regard to AInfD, we can distinguish rare monogenic diseases, like familial Mediterranean fever (FMF) and tumor necrosis factor receptor associated autoinflammatory periodic syndrome (TRAPS), and polygenic diseases, such as gout and IBD. In gout, serum

PT

urate levels are influenced by a combination of inherited genetic variants and the environment [41]. The largest GWAS thus far among Europeans involved 110,000 individuals

RI

and uncovered 28 urate-associated loci [42], of which 10 were previously identified in smaller

SC

studies [43]. These polymorphisms are mainly located in genes encoding renal and gut uric acid transporters (SLC2A9/GLUT9, ABCG2, SLC22A11/OAT4, SLC22A12/URAT1,

NU

SLC17A1/NPT1 and the auxiliary molecule PDZK1) [43]. Twin studies have estimated the

MA

heritability of urate levels to be between 45% and 73% [44].

Is it a matter of sex?

D

Sex is genetically determined and is a biologic variable influencing both innate and adaptive

PT E

immune responses. Men and women also differ in their immunological responses to infections and self-antigens [45]. In general terms, women have enhanced antibody production and increased cell-mediated responses following immunization, while men

CE

produce a more vigorous immune response to infectious organisms [46]. Furthermore,

AC

women show higher CD4+ T cell counts than men, which contributes to an increased CD4/CD8 ratio, higher levels of plasma IgM and greater T-helper 1 (Th1) cytokine production [47]. Predictably, women and men not only differ in their normal immune response, but there are also differences in the prevalence, clinical presentation and severity of systemic rheumatic diseases [48]. This had fed the notion that an altered immune responses may correlate with increased susceptibility to AID in women [49, 50]. With regard to SpA, men and women are equally affected, but men are 2-3 times more susceptible than women to all forms of axial SpA [51, 52]. With a similar ratio between men and women in gout and Behcet’s disease [53], men are more affected than women at all

7

ACCEPTED MANUSCRIPT ages, despite an increase of gout in women following menopause [54]. As mentioned above, the sex bias is quite clear for AID as nearly all conditions are largely more prevalent in women [55, 56], as represented by women being 2-3 times more likely to develop RA compared to men [57]. The lack of a female preponderance in SpA argues against a

PT

classification as AID and is in favor of SpA being an AInfD (Figure 1).

The role of the environment

RI

As the development of SpA cannot be fully explained by genetic factors, it has been

SC

proposed that many environmental components, including smoking, trauma, infections and microbiota are critically involved in the induction of the disease in genetically predisposed

NU

individuals. Epigenetic regulation of gene activity represents an important operational link between cells and their internal and/or external environment, as it allows them to adapt to

MA

(physiological) change [58]. Molecular epigenetic regulation occurs at three macromolecular levels, i.e. DNA, RNA and histone proteins, that collectively control gene expression at the

D

transcriptional and post-transcriptional levels [59, 60]. The most-studied epigenetic

PT E

mechanisms include DNA cytosine-(hydroxy)methylation and many biochemically distinct combinatorial post-translational modifications to histones (i.a. methyl-, acetyl-, phosphoryl-, ubiquityl-, sumoylation and citrullination), the core components of nucleosomal modules in

CE

eukaryotic chromatin. In conjunction with various types of non-coding RNAs, DNA and

AC

histone modifications constitute the nuclear epigenome: nucleosome positioning and chromatin accessibility of effector complexes control e.g. transcription, replication, recombination and repair [61]. Multiple studies have revealed a correlation between gene expression and epigenetic regulation in the context of immune cell development and function, and, of relevance in this review, the involvement of epigenetic deregulation in AInfD and AID [62-64]. Despite these advances, the origin of such epigenetic deregulation often remains obscure. Genetic defects in epigenetic regulatory factors are implicated in various rare syndromes; one of the most prominent examples thereof is the direct connection between intellectual disabilities and

8

ACCEPTED MANUSCRIPT genetic mutations in a variety of epigenetic regulators [65, 66]. In the context of immunedysfunction, mutant forms of the DNMT3B (DNA de novo methyltransferase 3B) and AICDA (5-methyl cytidine deaminase) genes have been directly connected to immunodeficiency, centromeric instability and facial anomalies (ICF) and hyper-IgM 2 (HIGM2) syndrome, respectively [67, 68]. Although numerous transcription factors have been implicated in

the involvement of non-coding RNAs is lagging behind [69-71].

PT

genetic predisposition to immune-disorders, including AInfD and AID, similar knowledge on

RI

Although immune-(dys)regulation is clearly associated with environmental exposure,

SC

however, exactly how environmental factors affect epigenetic regulation and may contribute to disease is much less clear [72-75]. The link between nutrition, metabolic status and

NU

epigenetic transcriptional regulation illustrates a direct functional interaction between genes and their microenvironment, especially in the context of immune function [76-79]. Although

MA

the underlying etiology is often far from being completely understood, multiple environmental exposures that adversely affect health (i.a. Western type diet, tobacco smoke, particulate

D

matter) are known to affect the immune system [80-82].

PT E

It is widely accepted that smoking is one of the factors that contributes to the pathogenesis of AS, while being also associated with disease severity [83]. Smoking may also influence the development of psoriasis and PsA: a recent report showed that childhood exposure to

CE

environmental tobacco smoke is significantly associated with psoriasis (OR 1.28, 95% CI

AC

1.10-1.49, p=0.002). Moreover, smokers with a history of >5 pack-years manifest a higher risk for psoriasis (OR 2.18, 95% CI 1.82-2.61, p<0.001). Twin studies have shown that psoriasis risk is 1.23 in the ever-smoking twin among MZ twins (95% CI 0.59-2.56; p=0.578) compared to same-sex DZ twins (OR 2.21, 95% CI 1.36-3.58; p=0.001). The results support that 20% (14-25%; p<0.001) of the correlation between psoriasis and smoking is hereditary, whereas non-shared environmental factors seem to account for an additional 8% (0-22%; p=0.504) [84]. The exact mechanism behind this gene-environment interaction is not known but it may be related to overproduction of pro-inflammatory factors like C-reactive protein, interleukin-6, matrix metalloproteinases [85]. In RA the role of smoking is mechanistically

9

ACCEPTED MANUSCRIPT better understood. Smoking appears important throughout the preclinical phases of RA development, as it may, in the context of HLA DR-shared epitope proteins, trigger RAspecific immune reactions to citrullinated proteins [86], particularly for seropositive RA [87] and in men [88]. Trauma, injury and heavy lifting have been associated with SpA development, in particular for PsA [89, 90]. Physical trauma is a potential trigger for PsA, as

PT

evidenced by the long-standing observed Koebner phenomenon in psoriasis [91], where psoriatic plaques develop in areas of physical trauma [92]. A case report of MZ twins

RI

developing PsA after trauma substantiates this notion [93]. These findings have recently

SC

been confirmed by a large real-life observational study, in which physical trauma was associated with PsA development in psoriasis patients, with a hazard ratio (HR) of 1.32 (95%

NU

CI 1.13-1.54). Among traumas displaying a significant association with PsA development were bone and joint traumas, while skin and nerve traumas were not associated [94]. In this

MA

view, enthesitis, the inflammation of fibrocartilaginous entheses (ligaments), represents the common denominator between SpA, and subclinical enthesitis may be the primary trigger of

D

secondary joint synovitis through the release of proinflammatory mediators [95]. Recent MRI

PT E

analyses have enabled important in-depth characterization and showed that enthesitis is a very common feature (30-50%) in distal interphalangeal joints in PsA patients [96, 97]. Enthesitis envelops the nail matrix [98], and high mechanical stress and chronic micro-

CE

trauma may be the underlying trigger for this inflammatory reaction [95].

AC

Infections have been recognized as triggers for SpA since a long time, as ReA (previously including Reiter syndrome, with the classic triad of symptoms in a soldier following a digestive infection, and attributed to Treponema pallidum infection [99]. However, ReA is a disease with an ancient history, as Hippocrates around 460 BC may have been the first to postulate the association between arthritis and genitourinary infections, writing that “a youth does not suffer from gout until sexual intercourse” [100]. Even in ancient Egypt, ReA was reported in a medical papyrus [101]. ReA follows an infection of the digestive or genitourinary tract caused by different microorganisms, including: Chlamydia trachomatis, Shigella flexneri, Salmonella

enteritidis,

Salmonella

typhimurium,

Salmonella

muenchen,

Yersinia

10

ACCEPTED MANUSCRIPT enterocolitica, Yersinia pseudotubercolosis, Campylobacter jejuni, Campylobacter fetus, Ureaplasma urealyticum, and Clostridium difficile. Other less common microorganisms involved in ReA include Neisseria gonorrhoeae, Borrelia burgdorferi, Chlamydia pneumoniae and Escherichia coli [102]. The exact mechanism by which these bacteria trigger ReA is currently unknown. Cross-reactivity between the pathogen and self-components has been

PT

hypothesized as a factor in the onset of ReA, because of the association or ReA with HLAB27 [102]. Interestingly, antibodies directed to microorganism antigens were reported to also

RI

display affinity for HLA-B27 [102]. However, this argument fails if the association with HLA-

SC

B27 is attributed to intracellular protein misfolding (vide supra). More studies are needed to test the hypothesis that molecular mimicry plays a role in the development of ReA. This is

NU

important information for the classification of SpA as AInfD or AID because molecular mimicry implicitly implicates that the adaptive immune system is involved, which is the typical

MA

hallmark of AID.

Infections have also been related to the pathogenesis of RA. In particular Porphyromonus

D

gingivalis, the main cause of periodontitis, is a well-known risk factor. It is the only human

PT E

pathogen known to express peptidylarginine deiminase, the enzyme that generates citrullinated epitopes that are recognized by anti-citrullinated protein antibodies (ACPA) [103]. These enzymes, however, are also abundantly expressed by inflammatory cells, including

CE

macrophages, that infiltrate the affected synovial tissues.

AC

Overall, environmental factors seem to play a crucial role in determining SpA, however current research has not identified a single pathogenic trigger. Infections, in particular genitourinary and gastro-intestinal, represent a risk factor for ReA, while mechanical stress for PsA.

The hidden microbial self Besides the effect of external exposure to microbial organisms on immune reactivity, it is becoming increasingly clear that commensal bacteria (microbiota) have a significant impact on our immunological health. The microbiota, or the ecosystem of microorganisms living on

11

ACCEPTED MANUSCRIPT mucosal surfaces and the skin in mutually beneficial coexistence with the host, influences numerous physiological processes, including longevity, metabolism and, relevant to the present discussion, the immune system development and function [104, 105]. Our resident bacterial community should be considered an acquired organ with its own plasticity, organized with stratification and hierarchy and not assembled randomly [106]. It is

PT

established that disturbances in the microbiota mainly in the gut, or dysbiosis, not only correlates with, but may cause a number of pathological processes, including AID and AInfD

RI

[107]. The microbiota is in strict relationship with the mucosal immune system, which evolved

SC

to prevent invasion of resident and pathogenic microbes. As such, the gut microbiota is essential for oral tolerance, the capacity of the enteric immune system to acquire non-

NU

responsiveness to food and dietary antigens [108].

MA

Crohn’s disease and ulcerative colitis were, for obvious reasons, the first diseases to be associated with gut dysbiosis, with initial studies showing substantial taxonomic shifts with a reduction in beneficial Clostridia and Bacteroides fragilis subtypes and enrichment in

D

Escherichia coli [109]. The interaction at mucosal sites between the microbiota and the

PT E

epithelial cells shapes the adaptive immune system: particular Clostridia clusters allow the fermentation of dietary fibers, leading to the production of short-chain fatty acids, like

CE

butyrate, which exert anti-inflammatory effects by promoting T regulatory (Treg) cell development [110-112]. Of note, butyrate is a known histone deactetylase inhibitor, and thus

AC

a potential epigenetic modulator in the context of immune function [113]. Moreover, B. fragilis produces capsular polysaccharide A which induces Tregs, while the exogenous administration of polysaccharide A restores the immunological balance in germ-free mice and has protective effects towards colitis [104, 114]. Additional beneficial effects of the gut microbiota include the induction of IL-22 by bacteria-derived indole metabolites, which promotes the expression of antimicrobial proteins, limits the expansion of commensal segmented filamentous bacteria and inhibits Th17 cell activation [115]. The long-established link between IBD and SpA suggests and involvement of the microbiota in systemic rheumatic disease [116]. Early evidence from animal studies revealed a relevant interaction between

12

ACCEPTED MANUSCRIPT HLA surface antigens and microbiota: in HLA-B27 transgenic rats, the presence of the HLAB27 antigen was associated with an altered microbial composition of the gut; the relative abundance of Prevotella spp. and Bacteroides vulgatus was increased, whereas that of Rikenellaceae was decreased [117]. Moreover, it has been demonstrated that prebiotics reduce the severity of colitis and prevent the development of arthritis in these rats [118].

PT

Human observational studies have assessed whether the composition of the faecal microbiome differs between SpA and controls. Sequence analysis of 16S ribosomal RNA

RI

genes, whole-microbial genomes and metabolomic analysis, pointed to a decreased

SC

microbial diversity in PsA patients compared to controls: Akkermansia, Ruminococcus, and Pseudobutyrivibrio populations were significantly reduced [119]. It has also been

NU

demonstrated that dysbiosis in SpA is disease specific, as there is an abundance of Ruminococcus gnavus compared with both RA and controls [120]. The dysbiosis could be

MA

influenced by breastfeeding, as AS risk is reduced in subjects that were breastfed, in line with a potentially important involvement of gut microbiota in the onset of the disease [121].

D

Complex disturbances in the microbiota have also been associated with RA: whereas germ-

PT E

free mice are protected against experimental arthritis, the reestablishment of intestinal microbiota restores the production of autoantibodies and arthritis [122, 123]. The underlying mechanisms likely involve induction of Th17 cells in the intestinal lamina propria [123],

CE

possibly via dendritic cell and macrophage activation, production of pro-inflammatory

AC

cytokines, as IL-1β and IL-6 [124], and induction of IL-10-producing B regulatory cells [125]. Interestingly, in RA the microbiota changes during the disease course, with enrichment in Prevotella copri in the early phases and reduction in long-standing RA [126, 127]. It is currently unclear whether this may reflect a common disease-related evolution in the microbiota composition, or whether this relates to chronic treatment. In addition to changes in gut microbiota RA also correlates with altered oral microbiota, as suggested by its association with P. gingivalis [128]. The gut microbiota was also shown to modulate arthritis in experimental models of gout. In germ-free mice, the production of short-chain fatty acids that are necessary for adequate inflammasome assembly and IL-1β production is decreased,

13

ACCEPTED MANUSCRIPT due to the metabolite-sensing receptor free fatty acid receptor 2 (also known as G-protein coupled receptor 43) [129]. Moreover, observations in some experimental models of arthritis show that microbial triggers by commensal bacteria are required to promote disease in genetically predisposed animals [125]. Also in PsA it has been postulated that host genetics may be involved in the balance between microbiome and the inflammatory disease [130].

PT

Overall, the role of microbiota in inflammatory arthritis has been established, as for instance in the HLA-B27/human beta-2 microglobulin transgenic rat model. However, several studies

RI

reported gut dysbiosis in different arthritides, with few differences but this could be due to the

SC

limited number of patients and microbial species examined. Furthermore, the impact of

NU

epigenetics will need further investigation.

Current status and perspectives

MA

The available evidence with regards to genetic and environmental involvement show that SpA are chronic inflammatory diseases associated with a strong genetic predisposition, i.e.

D

HLA-B27, and with a variable environmental component, which typically induce development

PT E

of the disease. Different from classical AID, such as RA, there is no female predominance in SpA, autoantibodies are undetectable and there are thus far no known shared genetic predispositions. Moreover, whereas in RA environmental agents, among which smoking,

CE

appear to induce pathogenesis, in SpA the evidence for an etiological involvement of e.g.

AC

smoking is less robust. In contrast physical trauma and infections appear to trigger SpA development in genetically predisposed individuals. Considering that it is part of human nature to cluster and distinguish disease entities based on discernable criteria, it should be recognized that boundaries between such clusters are often subjective and may even be based on incorrect assumptions. By analogy, several animal species have been clustered in genus and/or families because of morphological characteristics, while genomic analyses eventually pointed towards another direction. Similarly, in the era of omics and big data analyses, a reclassification of immune mediated inflammatory diseases may be revealed in the near future. As for now, AInfD and AID are

14

ACCEPTED MANUSCRIPT distinguished primarily based on the involvement of the innate and adaptive immune system, respectively (Figure 1). These immune responses are the consequence of geneenvironment interactions. Obviously, some AInfD, for instance the monogenetic periodic fever syndromes, are exclusively driven by innate immune responses, whereas other AInfD, like psoriasis and IBD, are additionally characterized by T cell involvement next to clear-cut

PT

innate mechanisms. As such, SpA seems to be more closely related to the typical AID than other polygenic AInfD. Nevertheless SpA display prominent features of autoinflammatory

RI

conditions and, in our opinion, should, perhaps more justifiably, be classified within the

SC

autoinflammatory category. This is further underscored by the response to immunemodulating therapies, in particular biologics. As has been shown for neutralization of IL-17

NU

and IL-23, effective future therapies for AInfD are likely to be also effective for SpA.

MA

Conclusion

The combined data presented herein suggests that the induced inflammatory reaction is out

D

of control in SpA, either due to over-reactivity of the innate immune system, or due to

PT E

insufficient control mechanisms. In addition, the microbiota differentially shape the immune system in SpA compared to RA; conclusive evidence for microbiota involvement for other

CE

conditions, like gout, is currently not available. The exact etiological involvement and mechanistic underpinnings of epigenetic dysregulation in SpA and other conditions

AC

discussed throughout this review, is expected to become increasingly clear as technology (acquisition and analysis) required to study chromatin-structure biology and its interaction with the environment further develops.

15

ACCEPTED MANUSCRIPT References

1.

Doria, A., et al., Autoinflammation and autoimmunity: bridging the divide.

Autoimmun Rev, 2012. 12(1): p. 22-30. 2.

Arbuckle, M.R., et al., Development of autoantibodies before the clinical onset of

systemic lupus erythematosus. N Engl J Med, 2003. 349(16): p. 1526-33. Davidson, A. and B. Diamond, Autoimmune diseases. N Engl J Med, 2001. 345(5): p.

PT

3.

Damoiseaux, J.G. and J.W. Tervaert, The definition of autoimmune disease: are

SC

4.

RI

340-50.

Koch's postulates applicable? Neth J Med, 2002. 60(7): p. 266-8. Putterman, C. and Y. Naparstek, Koch's postulates and autoimmunity: an opposing

NU

5.

viewpoint. Arch Immunol Ther Exp (Warsz), 1999. 47(1): p. 25-32. Rigante, D., et al., The hereditary autoinflammatory disorders uncovered. Autoimmun

MA

6.

Rev, 2014. 13(9): p. 892-900.

Ciccarelli, F., M. De Martinis, and L. Ginaldi, An update on autoinflammatory

D

7.

8.

PT E

diseases. Curr Med Chem, 2014. 21(3): p. 261-9. Sakkas, L.I. and D.P. Bogdanos, Are psoriasis and psoriatic arthritis the same

Deodhar, A., P. Miossec, and X. Baraliakos, Is undifferentiated spondyloarthritis a

AC

9.

CE

disease? The IL-23/IL-17 axis data. Autoimmun Rev, 2017. 16(1): p. 10-15.

discrete entity? A debate. Autoimmun Rev, 2018. 17(1): p. 29-32. 10.

Eder, L., et al., Gender difference in disease expression, radiographic damage and

disability among patients with psoriatic arthritis. Ann Rheum Dis, 2013. 72(4): p. 578-82. 11.

Quaden, D.H., L.M. De Winter, and V. Somers, Detection of novel diagnostic

antibodies in ankylosing spondylitis: An overview. Autoimmun Rev, 2016. 15(8): p. 820-32. 12.

Dolcino, M., et al., Crossreactive autoantibodies directed against cutaneous and joint

antigens are present in psoriatic arthritis. PLoS One, 2014. 9(12): p. e115424. 16

ACCEPTED MANUSCRIPT 13.

Chimenti, M.S., et al., Auto-reactions, autoimmunity and psoriatic arthritis.

Autoimmun Rev, 2015. 14(12): p. 1142-6. 14.

Conigliaro, P., et al., Autoantibodies in inflammatory arthritis. Autoimmun Rev, 2016.

15(7): p. 673-83. 15.

Prajzlerova, K., et al., An update on biomarkers in axial spondyloarthritis.

Maksymowych, W.P., An update on biomarker discovery and use in axial

RI

16.

PT

Autoimmun Rev, 2016. 15(6): p. 501-9.

17.

Generali, E., et al., Biomarkers in psoriatic arthritis: a systematic literature review.

McGonagle, D. and M.F. McDermott, A proposed classification of the immunological

MA

diseases. PLoS Med, 2006. 3(8): p. e297. 19.

NU

Expert Rev Clin Immunol, 2016. 12(6): p. 651-60. 18.

SC

spondyloarthritis. Expert Rev Mol Diagn, 2017. 17(11): p. 965-974.

Ambarus, C., et al., Pathogenesis of spondyloarthritis: autoimmune or

Rubbert-Roth, A., et al., TNF inhibitors in rheumatoid arthritis and spondyloarthritis:

PT E

20.

D

autoinflammatory? Curr Opin Rheumatol, 2012. 24(4): p. 351-8.

Are they the same? Autoimmun Rev, 2018. 17(1): p. 24-28. Ranganathan, V., et al., Pathogenesis of ankylosing spondylitis - recent advances and

CE

21.

future directions. Nat Rev Rheumatol, 2017. 13(6): p. 359-367. Queiro, R., et al., HLA-B27 and psoriatic disease: a modern view of an old

AC

22.

relationship. Rheumatology (Oxford), 2016. 55(2): p. 221-9. 23.

Colbert, R.A., et al., HLA-B27 misfolding and spondyloarthropathies. Prion, 2009.

3(1): p. 15-26. 24.

Turner, M.J., et al., HLA-B27 up-regulation causes accumulation of misfolded heavy

chains and correlates with the magnitude of the unfolded protein response in transgenic rats: Implications for the pathogenesis of spondylarthritis-like disease. Arthritis Rheum, 2007. 56(1): p. 215-23. 17

ACCEPTED MANUSCRIPT 25.

Colbert, R.A., et al., From HLA-B27 to spondyloarthritis: a journey through the ER.

Immunol Rev, 2010. 233(1): p. 181-202. 26.

Turner, M.J., et al., HLA-B27 misfolding in transgenic rats is associated with

activation of the unfolded protein response. J Immunol, 2005. 175(4): p. 2438-48. Bowness, P., Hla-B27. Annu Rev Immunol, 2015. 33: p. 29-48.

28.

DeLay, M.L., et al., HLA-B27 misfolding and the unfolded protein response augment

PT

27.

RI

interleukin-23 production and are associated with Th17 activation in transgenic rats.

29.

SC

Arthritis Rheum, 2009. 60(9): p. 2633-43.

Goodall, J.C., et al., Endoplasmic reticulum stress-induced transcription factor,

NU

CHOP, is crucial for dendritic cell IL-23 expression. Proc Natl Acad Sci U S A, 2010. 107(41): p. 17698-703.

Vitulano, C., et al., The interplay between HLA-B27 and ERAP1/ERAP2

MA

30.

aminopeptidases: from anti-viral protection to spondyloarthritis. Clin Exp Immunol, 2017.

Nestle, F.O., D.H. Kaplan, and J. Barker, Psoriasis. N Engl J Med, 2009. 361(5): p.

PT E

31.

D

190(3): p. 281-290.

496-509.

Bowes, J., et al., Investigation of IL1, VEGF, PPARG and MEFV genes in psoriatic

CE

32.

arthritis susceptibility. Ann Rheum Dis, 2012. 71(2): p. 313-4. Brown, M.A., T. Kenna, and B.P. Wordsworth, Genetics of ankylosing spondylitis--

AC

33.

insights into pathogenesis. Nat Rev Rheumatol, 2016. 12(2): p. 81-91. 34.

Lopalco, G., et al., Interleukin-1 as a common denominator from autoinflammatory to

autoimmune disorders: premises, perils, and perspectives. Mediators Inflamm, 2015. 2015: p. 194864. 35.

Generali, E., et al., Lessons learned from twins in autoimmune and chronic

inflammatory diseases. J Autoimmun, 2017. 83: p. 51-61.

18

ACCEPTED MANUSCRIPT 36.

Pedersen, O.B., et al., On the heritability of psoriatic arthritis. Disease concordance

among monozygotic and dizygotic twins. Ann Rheum Dis, 2008. 67(10): p. 1417-21. 37.

Pedersen, O.B., et al., Ankylosing spondylitis in Danish and Norwegian twins:

occurrence and the relative importance of genetic vs. environmental effectors in disease causation. Scand J Rheumatol, 2008. 37(2): p. 120-6. Brown, M.A., et al., Susceptibility to ankylosing spondylitis in twins: the role of genes,

PT

38.

Angelotti, F., et al., One year in review 2017: pathogenesis of rheumatoid arthritis.

SC

39.

RI

HLA, and the environment. Arthritis Rheum, 1997. 40(10): p. 1823-8.

Clin Exp Rheumatol, 2017. 35(3): p. 368-378.

Ciccacci, C., et al., Polymorphisms in STAT-4, IL-10, PSORS1C1, PTPN2 and

NU

40.

MIR146A genes are associated differently with prognostic factors in Italian patients affected

41.

MA

by rheumatoid arthritis. Clin Exp Immunol, 2016. 186(2): p. 157-163. So, A.K. and F. Martinon, Inflammation in gout: mechanisms and therapeutic targets.

Kottgen, A., et al., Genome-wide association analyses identify 18 new loci associated

PT E

42.

D

Nat Rev Rheumatol, 2017. 13(11): p. 639-647.

with serum urate concentrations. Nat Genet, 2013. 45(2): p. 145-54. Dalbeth, N., L.K. Stamp, and T.R. Merriman, The genetics of gout: towards

CE

43.

personalised medicine? BMC Med, 2017. 15(1): p. 108. Krishnan, E., et al., Nature versus nurture in gout: a twin study. Am J Med, 2012.

AC

44.

125(5): p. 499-504. 45.

Klein, S.L. and K.L. Flanagan, Sex differences in immune responses. Nat Rev

Immunol, 2016. 16(10): p. 626-38. 46.

Roved, J., H. Westerdahl, and D. Hasselquist, Sex differences in immune responses:

Hormonal effects, antagonistic selection, and evolutionary consequences. Horm Behav, 2017. 88: p. 95-105.

19

ACCEPTED MANUSCRIPT 47.

Lleo, A., et al., Is autoimmunity a matter of sex? Autoimmun Rev, 2008. 7(8): p. 626-

30. 48.

Invernizzi, P., et al., Female predominance and X chromosome defects in autoimmune

diseases. J Autoimmun, 2009. 33(1): p. 12-6. 49.

Selmi, C., The X in sex: how autoimmune diseases revolve around sex chromosomes.

Selmi, C., et al., The X chromosome and the sex ratio of autoimmunity. Autoimmun

RI

50.

SC

Rev, 2012. 11(6-7): p. A531-7. 51.

PT

Best Pract Res Clin Rheumatol, 2008. 22(5): p. 913-22.

Lubrano, E., et al., The Sex Influence on Response to Tumor Necrosis Factor-alpha

52.

NU

Inhibitors and Remission in Axial Spondyloarthritis. J Rheumatol, 2017. Landi, M., et al., Gender differences among patients with primary ankylosing

MA

spondylitis and spondylitis associated with psoriasis and inflammatory bowel disease in an iberoamerican spondyloarthritis cohort. Medicine (Baltimore), 2016. 95(51): p. e5652. Hatemi, G., et al., One year in review 2016: Behcet's syndrome. Clin Exp Rheumatol,

54.

PT E

2016. 34(6 Suppl 102): p. 10-22.

D

53.

Dalbeth, N., T.R. Merriman, and L.K. Stamp, Gout. Lancet, 2016. 388(10055): p.

CE

2039-2052.

Voskuhl, R., Sex differences in autoimmune diseases. Biol Sex Differ, 2011. 2(1): p. 1.

56.

Margery-Muir, A.A., et al., Gender balance in patients with systemic lupus

AC

55.

erythematosus. Autoimmun Rev, 2017. 16(3): p. 258-268. 57.

Deane, K.D., et al., Genetic and environmental risk factors for rheumatoid arthritis.

Best Pract Res Clin Rheumatol, 2017. 31(1): p. 3-18. 58.

Tammen, S.A., S. Friso, and S.W. Choi, Epigenetics: the link between nature and

nurture. Mol Aspects Med, 2013. 34(4): p. 753-64. 59.

Guil, S. and M. Esteller, DNA methylomes, histone codes and miRNAs: tying it all

together. Int J Biochem Cell Biol, 2009. 41(1): p. 87-95. 20

ACCEPTED MANUSCRIPT 60.

Chen, B.S. and C.W. Li, Constructing an integrated genetic and epigenetic cellular

network for whole cellular mechanism using high-throughput next-generation sequencing data. BMC Syst Biol, 2016. 10: p. 18. 61.

Musselman, C.A., et al., Perceiving the epigenetic landscape through histone readers.

Nat Struct Mol Biol, 2012. 19(12): p. 1218-27. Saito, Y., et al., Epigenetic alterations and microRNA misexpression in cancer and

PT

62.

Gupta, B. and R.D. Hawkins, Epigenomics of autoimmune diseases. Immunol Cell

SC

63.

RI

autoimmune diseases: a critical review. Clin Rev Allergy Immunol, 2014. 47(2): p. 128-35.

Biol, 2015. 93(3): p. 271-6.

Wu, H., et al., Critical Link Between Epigenetics and Transcription Factors in the

NU

64.

Induction of Autoimmunity: a Comprehensive Review. Clin Rev Allergy Immunol, 2016.

65.

MA

50(3): p. 333-44.

Kleefstra, T., et al., The genetics of cognitive epigenetics. Neuropharmacology, 2014.

Huidobro, C., A.F. Fernandez, and M.F. Fraga, The role of genetics in the

PT E

66.

D

80: p. 83-94.

establishment and maintenance of the epigenome. Cell Mol Life Sci, 2013. 70(9): p. 1543-73. Rodriguez-Cortez, V.C., et al., Epigenomic deregulation in the immune system.

CE

67.

Epigenomics, 2011. 3(6): p. 697-713. Langlais, D., N. Fodil, and P. Gros, Genetics of Infectious and Inflammatory Diseases:

AC

68.

Overlapping Discoveries from Association and Exome-Sequencing Studies. Annu Rev Immunol, 2017. 35: p. 1-30. 69.

Hu, H. and R.A. Gatti, MicroRNAs: new players in the DNA damage response. J Mol

Cell Biol, 2011. 3(3): p. 151-8. 70.

John, L.B. and A.C. Ward, The Ikaros gene family: transcriptional regulators of

hematopoiesis and immunity. Mol Immunol, 2011. 48(9-10): p. 1272-8.

21

ACCEPTED MANUSCRIPT 71.

Lambert, S.A., et al., The Human Transcription Factors. Cell, 2018. 172(4): p. 650-

665. 72.

Lisciandro, J.G. and A.H. van den Biggelaar, Neonatal immune function and

inflammatory illnesses in later life: lessons to be learnt from the developing world? Clin Exp Allergy, 2010. 40(12): p. 1719-31. Wlasiuk, G. and D. Vercelli, The farm effect, or: when, what and how a farming

PT

73.

RI

environment protects from asthma and allergic disease. Curr Opin Allergy Clin Immunol,

74.

SC

2012. 12(5): p. 461-6.

Blumberg, R.S., Environment and Genes: What Is the Interaction? Dig Dis, 2016.

75.

Ege, M.J., The Hygiene Hypothesis in the Age of the Microbiome. Ann Am Thorac

MA

Soc, 2017. 14(Supplement_5): p. S348-S353. 76.

NU

34(1-2): p. 20-6.

Wallace, D.C., Bioenergetics and the epigenome: interface between the environment

Salminen, A., et al., Epigenetic regulation of ASC/TMS1 expression: potential role in

PT E

77.

D

and genes in common diseases. Dev Disabil Res Rev, 2010. 16(2): p. 114-9.

apoptosis and inflammasome function. Cell Mol Life Sci, 2014. 71(10): p. 1855-64. Nieborak, A. and R. Schneider, Metabolic intermediates - Cellular messengers talking

CE

78.

to chromatin modifiers. Mol Metab, 2018. Rapozo, D.C., C. Bernardazzi, and H.S. de Souza, Diet and microbiota in

AC

79.

inflammatory bowel disease: The gut in disharmony. World J Gastroenterol, 2017. 23(12): p. 2124-2140. 80.

Tilg, H. and A.R. Moschen, Food, immunity, and the microbiome. Gastroenterology,

2015. 148(6): p. 1107-19. 81.

De Rosa, V., et al., Nutritional control of immunity: Balancing the metabolic

requirements with an appropriate immune function. Semin Immunol, 2015. 27(5): p. 300-9.

22

ACCEPTED MANUSCRIPT 82.

Garn, H., et al., Current concepts in chronic inflammatory diseases: Interactions

between microbes, cellular metabolism, and inflammation. J Allergy Clin Immunol, 2016. 138(1): p. 47-56. 83.

Villaverde-Garcia, V., et al., The effect of smoking on clinical and structural damage

in patients with axial spondyloarthritis: A systematic literature review. Semin Arthritis

Lonnberg, A.S., et al., Smoking and risk for psoriasis: a population-based twin study.

RI

84.

PT

Rheum, 2017. 46(5): p. 569-583.

85.

SC

Int J Dermatol, 2016. 55(2): p. e72-8.

Eder, L., et al., The association between smoking and the development of psoriatic

86.

NU

arthritis among psoriasis patients. Ann Rheum Dis, 2012. 71(2): p. 219-24. Klareskog, L., et al., A new model for an etiology of rheumatoid arthritis: smoking

MA

may trigger HLA-DR (shared epitope)-restricted immune reactions to autoantigens modified by citrullination. Arthritis Rheum, 2006. 54(1): p. 38-46. Sparks, J.A. and E.W. Karlson, The Roles of Cigarette Smoking and the Lung in the

D

87.

PT E

Transitions Between Phases of Preclinical Rheumatoid Arthritis. Curr Rheumatol Rep, 2016. 18(3): p. 15.

Sugiyama, D., et al., Impact of smoking as a risk factor for developing rheumatoid

CE

88.

arthritis: a meta-analysis of observational studies. Ann Rheum Dis, 2010. 69(1): p. 70-81. Love, T.J., et al., Obesity and the risk of psoriatic arthritis: a population-based study.

AC

89.

Ann Rheum Dis, 2012. 71(8): p. 1273-7. 90.

Van Mechelen, M. and R.J. Lories, Microtrauma: no longer to be ignored in

spondyloarthritis? Curr Opin Rheumatol, 2016. 28(2): p. 176-80. 91.

Liang, Y., et al., Psoriasis: a mixed autoimmune and autoinflammatory disease. Curr

Opin Immunol, 2017. 49: p. 1-8. 92.

Veale, D.J. and O. FitzGerald, Psoriatic arthritis--pathogenesis and epidemiology.

Clin Exp Rheumatol, 2002. 20(6 Suppl 28): p. S27-33. 23

ACCEPTED MANUSCRIPT 93.

Ng, J., A.L. Tan, and D. McGonagle, Unifocal psoriatic arthritis development in

identical twins following site specific injury: evidence supporting biomechanical triggering events in genetically susceptible hosts. Ann Rheum Dis, 2015. 74(5): p. 948-9. 94.

Thorarensen, S.M., et al., Physical trauma recorded in primary care is associated with

the onset of psoriatic arthritis among patients with psoriasis. Ann Rheum Dis, 2017. 76(3): p.

McGonagle, D., Enthesitis: an autoinflammatory lesion linking nail and joint

RI

95.

PT

521-525.

96.

SC

involvement in psoriatic disease. J Eur Acad Dermatol Venereol, 2009. 23 Suppl 1: p. 9-13. Tan, A.L., et al., High-resolution magnetic resonance imaging for the assessment of

97.

NU

hand osteoarthritis. Arthritis Rheum, 2005. 52(8): p. 2355-65.

Kaeley, G.S., et al., Enthesitis: A hallmark of psoriatic arthritis. Semin Arthritis

98.

MA

Rheum, 2018.

Tan, A.L., et al., The relationship between the extensor tendon enthesis and the nail in

D

distal interphalangeal joint disease in psoriatic arthritis--a high-resolution MRI and

99.

PT E

histological study. Rheumatology (Oxford), 2007. 46(2): p. 253-6. Wu, I.B. and R.A. Schwartz, Reiter's syndrome: the classic triad and more. J Am

100.

CE

Acad Dermatol, 2008. 59(1): p. 113-21. Carter, J.D. and A.P. Hudson, Reactive arthritis: clinical aspects and medical

101.

AC

management. Rheum Dis Clin North Am, 2009. 35(1): p. 21-44. Kwiecinski, J., Reactive arthritis in ancient Egypt: a possible description in medical

papyri. J Rheumatol, 2014. 41(3): p. 556-7. 102.

Generali, E., et al., Seronegative reactive spondyloarthritis and the skin. Clin

Dermatol, 2015. 33(5): p. 531-7. 103.

Potempa, J., P. Mydel, and J. Koziel, The case for periodontitis in the pathogenesis of

rheumatoid arthritis. Nat Rev Rheumatol, 2017. 13(10): p. 606-620.

24

ACCEPTED MANUSCRIPT 104.

Theofilopoulos, A.N., D.H. Kono, and R. Baccala, The multiple pathways to

autoimmunity. Nat Immunol, 2017. 18(7): p. 716-724. 105.

Van de Wiele, T., et al., How the microbiota shapes rheumatic diseases. Nat Rev

Rheumatol, 2016. 12(7): p. 398-411. 106.

Barin, J.G., L.D. Tobias, and D.A. Peterson, The microbiome and autoimmune

Opazo, M.C., et al., Intestinal Microbiota Influences Non-intestinal Related

108.

SC

Autoimmune Diseases. Front Microbiol, 2018. 9: p. 432.

RI

107.

PT

disease: Report from a Noel R. Rose Colloquium. Clin Immunol, 2015. 159(2): p. 183-8.

Huttenhower, C., A.D. Kostic, and R.J. Xavier, Inflammatory bowel disease as a

109.

NU

model for translating the microbiome. Immunity, 2014. 40(6): p. 843-54. Ruff, W.E. and M.A. Kriegel, Autoimmune host-microbiota interactions at barrier

110.

MA

sites and beyond. Trends Mol Med, 2015. 21(4): p. 233-44. Smith, P.M., et al., The microbial metabolites, short-chain fatty acids, regulate

Arpaia, N., et al., Metabolites produced by commensal bacteria promote peripheral

PT E

111.

D

colonic Treg cell homeostasis. Science, 2013. 341(6145): p. 569-73.

regulatory T-cell generation. Nature, 2013. 504(7480): p. 451-5. Koh, A., et al., From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as

CE

112.

Key Bacterial Metabolites. Cell, 2016. 165(6): p. 1332-1345. O'Keefe, S.J., Diet, microorganisms and their metabolites, and colon cancer. Nat Rev

AC

113.

Gastroenterol Hepatol, 2016. 13(12): p. 691-706. 114.

Neff, C.P., et al., Diverse Intestinal Bacteria Contain Putative Zwitterionic Capsular

Polysaccharides with Anti-inflammatory Properties. Cell Host Microbe, 2016. 20(4): p. 535547. 115.

Zelante, T., et al., Tryptophan catabolites from microbiota engage aryl hydrocarbon

receptor and balance mucosal reactivity via interleukin-22. Immunity, 2013. 39(2): p. 372-85.

25

ACCEPTED MANUSCRIPT 116.

Picchianti-Diamanti, A., M.M. Rosado, and R. D'Amelio, Infectious Agents and

Inflammation: The Role of Microbiota in Autoimmune Arthritis. Front Microbiol, 2017. 8: p. 2696. 117.

Lin, P., et al., HLA-B27 and human beta2-microglobulin affect the gut microbiota of

transgenic rats. PLoS One, 2014. 9(8): p. e105684. Hoentjen, F., et al., Reduction of colitis by prebiotics in HLA-B27 transgenic rats is

PT

118.

RI

associated with microflora changes and immunomodulation. Inflamm Bowel Dis, 2005.

119.

SC

11(11): p. 977-85.

Scher, J.U., et al., Decreased bacterial diversity characterizes the altered gut

NU

microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease. Arthritis Rheumatol, 2015. 67(1): p. 128-39.

Breban, M., et al., Faecal microbiota study reveals specific dysbiosis in

MA

120.

spondyloarthritis. Ann Rheum Dis, 2017. 76(9): p. 1614-1622. Ahsan, T., et al., Ankylosing Spondylitis: A rheumatology clinic experience. Pak J Med

122.

PT E

Sci, 2016. 32(2): p. 365-8.

D

121.

Carter, N.A., E.C. Rosser, and C. Mauri, Interleukin-10 produced by B cells is crucial

CE

for the suppression of Th17/Th1 responses, induction of T regulatory type 1 cells and reduction of collagen-induced arthritis. Arthritis Res Ther, 2012. 14(1): p. R32. Wu, H.J., et al., Gut-residing segmented filamentous bacteria drive autoimmune

AC

123.

arthritis via T helper 17 cells. Immunity, 2010. 32(6): p. 815-27. 124.

Messemaker, T.C., T.W. Huizinga, and F. Kurreeman, Immunogenetics of rheumatoid

arthritis: Understanding functional implications. J Autoimmun, 2015. 64: p. 74-81. 125.

Rosser, E.C., et al., Regulatory B cells are induced by gut microbiota-driven

interleukin-1beta and interleukin-6 production. Nat Med, 2014. 20(11): p. 1334-9.

26

ACCEPTED MANUSCRIPT 126.

Pianta, A., et al., Evidence of the Immune Relevance of Prevotella copri, a Gut

Microbe, in Patients With Rheumatoid Arthritis. Arthritis Rheumatol, 2017. 69(5): p. 964975. 127.

Scher, J.U., et al., Expansion of intestinal Prevotella copri correlates with enhanced

susceptibility to arthritis. Elife, 2013. 2: p. e01202. Montgomery, A.B., et al., Crystal structure of Porphyromonas gingivalis

PT

128.

RI

peptidylarginine deiminase: implications for autoimmunity in rheumatoid arthritis. Ann

129.

SC

Rheum Dis, 2016. 75(6): p. 1255-61.

Vieira, A.T., et al., A Role for Gut Microbiota and the Metabolite-Sensing Receptor

130.

NU

GPR43 in a Murine Model of Gout. Arthritis Rheumatol, 2015. 67(6): p. 1646-56. Chimenti, M.S., et al., Interaction between microbiome and host genetics in psoriatic

AC

CE

PT E

D

MA

arthritis. Autoimmun Rev, 2018. 17(3): p. 276-283.

27

ACCEPTED MANUSCRIPT Legend to figure 1:

Figure 1: The spectrum of systemic rheumatic diseases. Based on gene-environment interactions, the immune response will be directed towards predominant innate or adaptive immune system involvement; as such immune-mediated diseases can be separated in

PT

autoinflammatory (red box) and autoimmune diseases (blue box). Within the category of autoinflammatory diseases the predominance of the innate immunity is strongest on the left

RI

side and weakest on the right side of the red box. Spondyloarthritis is currently allocated to

SC

the autoimmune diseases, but classifies as an autoinflammatory disease based on a strong inflammatory component and lack of a female preponderance. The latter is supported by

NU

effectiveness of therapeutic approaches applied in the autoinflammatory diseases. The distribution of the distinct entities within the category of autoimmune diseases is rather

MA

arbitrary.

Abbreviations: AAV, ANCA-associated vasculitis; AIM, autoimmune myopathies; IBD,

AC

CE

PT E

D

inflammatory bowel disease; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus.

28

Figure 1