The B side of rheumatoid arthritis pathogenesis

The B side of rheumatoid arthritis pathogenesis

Journal Pre-proof The B side of Rheumatoid Arthritis pathogenesis Stefano Alivernini, Barbara Tolusso, Anna Laura Fedele, Clara Di Mario, Gianfranco F...

3MB Sizes 0 Downloads 87 Views

Journal Pre-proof The B side of Rheumatoid Arthritis pathogenesis Stefano Alivernini, Barbara Tolusso, Anna Laura Fedele, Clara Di Mario, Gianfranco Ferraccioli, Elisa Gremese

PII:

S1043-6618(19)31356-8

DOI:

https://doi.org/10.1016/j.phrs.2019.104465

Reference:

YPHRS 104465

To appear in:

Pharmacological Research

Received Date:

11 July 2019

Revised Date:

19 September 2019

Accepted Date:

20 September 2019

Please cite this article as: Alivernini S, Tolusso B, Fedele AL, Mario CD, Ferraccioli G, Gremese E, The B side of Rheumatoid Arthritis pathogenesis, Pharmacological Research (2019), doi: https://doi.org/10.1016/j.phrs.2019.104465

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. 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. © 2019 Published by Elsevier.

The B side of Rheumatoid Arthritis pathogenesis Stefano Alivernini1,2^, Barbara Tolusso1^, Anna Laura Fedele1, Clara Di Mario2, Gianfranco Ferraccioli2 and Elisa Gremese1,2*.

Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli – IRCCS, Rome, Italy

2

Institute of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy.

ro of

1

-p

Words count: 4137 Figures: 1

re

Tables: 1

^equally contributed to the work; *Corresponding author:

lP

References: 133

Jo

ur na

Elisa Gremese, MD Associate Professor of Rheumatology Division of Rheumatology Fondazione Policlinico Universitario A. Gemelli – IRCCS Università Cattolica del Sacro Cuore Via Giuseppe Moscati 31, 00168 – Rome, Italy Tel: +39 06 30159659 Email address: [email protected]

lP

re

-p

ro of

Graphical

ur na

Abstract

In the last years, a dramatic amount of research has been performed increasing the knowledge about the biological mechanism underpinning Rheumatoid Arthritis (RA) inflammation, putting B lymphocytes in the center of RA pathogenesis. Nowadays, B cell phenotypes and autoantibodies

Jo

positivity arose as important biomarkers in early and long-standing disease. Moreover, comparative analysis of peripheral blood and synovial tissue compartments enables the identification of novel physiopathological mechanisms promoting inflammation. In this narrative review we will discuss the biological relevance of B cell derived autoimmunity and in RA course, from disease onset to remission achievement.

2

1. Introduction Rheumatoid Arthritis (RA) is a chronic disease in which synovial tissue inflammation causes articular pain and progressive joint damage, leading to deformities and loss of function if not treated. In the last years, a dramatic amount of research has been performed increasing the knowledge about disease pathogenesis and course. Nowadays it is known that in RA course, a preclinical phase exists, before the onset of any joint symptoms. Multiple trigger factors have been implicated in the complex biological process of the loss of immunological tolerance during RA. In

ro of

particular, complex mechanisms regulate the cross-talk between innate and adaptive immune cells triggering the onset and maintenance of synovial tissue inflammation in RA [1]. In this narrative review we will dissect the biological relevance of B cell derived autoimmunity in the pathogenesis

-p

of RA.

The detection of autoantibodies in peripheral blood of asymptomatic subjects up to 10 years before

re

the clinical manifestation of the disease strongly suggests that the loss of tolerance to autoantigens

lP

occurs much time before the clinical disease onset and that, at clinical onset, the pathology is at an already advanced step. In particular, the availability of genetic markers which enable to predict RA onset or severity supports the notion that such mechanisms are effective over the whole life of a

ur na

person [2,3]. Moreover, genetic studies performed on multiple populations showed that the socalled ”shared epitope” [human leukocyte antigen (HLA)-DRB1 locus] is strongly linked with seropositivity for Rheumatoid Factor (RF) or Anti-Citrullinated Peptide Antibodies (ACPA) [4], in

Jo

addition with other risk alleles (i.e. PTPN22 and CTLA4) which have been demonstrated to be associated with T-cell stimulation, activation and functional differentiation [5-8]. Furthermore, the tight synergistic link between genetic and environmental parameters was widely demonstrated for smoking and other forms of bronchial stress able to increase the risk of RA development in genetically susceptible individuals [9,10]. Among other environmental factors, an increasingly consolidated role is attributed to obesity, that seems to influence the outcomes and prognosis of RA, but also contribute to the raising in disease 3

incidence [11,12], suggesting a role of fat tissue also in the disease pathogenesis [13]. In particular, data from three different RA cohorts indicate that obesity is associated with a likelihood of developing a seronegative RA, mostly in female (OR 1.6-3.5) [14-16].

1.1 Clinical phenotypes of RA Due to the wide heterogeneity in clinical presentation, evolution and response to treatments, several efforts have employed in defining subtypes of RA, but to date the main clinical and biological

ro of

distinction is still based on autoantibody status. It is now recognized that the presence or absence of RF and/or ACPA identify two different disease phenotypes, also from a pathogenetic point of view, as evidenced by the finding of autoantibodies years before the clinical onset of the disease. Studies

-p

mainly based on the 1987 RA classification criteria [17], supported the hypothesis that seropositive RA also underpinned a more severe prognosis of the disease, as regards radiographic progression

re

and response to therapies. More recent studies, mainly based on RA cohorts diagnosed according to

lP

the new 2010 RA classification criteria [18], show that seronegative RA have more inflammation and higher disease activity at baseline than seropositive patients, probably due to the greater weight given to the presence of autoantibodies in the 2010 criteria, having similar results in terms of

ur na

outcomes in both subtypes of RA [19-21].

On the other hand, the long-term extension analysis of the BeST study, that enrolled early RA patients (disease duration <3 years) with high disease activity, diagnosed according to the 1987

Jo

criteria, showed similar course in seronegative than in seropositive patients at 10 years of follow-up [22]. These findings indicate the need to adopt, in clinical practice, comparable approaches in ACPAneg as well as ACPApos RA patients but taking into account differential physio-pathological underlining mechanisms [23].

1.2 Autoimmunity against citrullinated antigens in RA

4

The detection of autoantibodies against citrullinated antigens is the main immunological hallmark of RA. These autoantibodies react against citrullinated peptides derived from arginine after posttranslational modification by the enzyme Peptidyl Arginine Deiminases (PADs) [24], being found in nearly two thirds of RA patients [25-29]. It has been demonstrated that the immune response against citrullinated peptides is initially restricted and increases over time as suggested by autoantibodies levels increase during the predating time period closer to the onset of clinical symptoms [30]. Moreover, a time-dependent expansion of ACPA specificity and subtypes suggests

ro of

the production of different autoantibody specificities mirroring the epitope spreading phenomenon [31]. In addition, the loss of tolerance leading to clinical manifestation of arthritis is tightly associated with the aberrant deregulation of cytokines release [32,33], including TNF- IL-6, IL-

-p

12p70, IL17 and IFN-, already years before RA onset. Recently, it was demonstrated that the inflammatory activity of ACPA IgG is promoted by Fc fragment glycosylation which is increased

re

before RA onset [34], in parallel with a galactosylation decrease and core fucosylation increase

lP

[35].

However, ACPA seropositivity in RA is not a matter only of specificity but their presence is

ur na

associated with a severe and erosive phenotype of the disease [36-39]. In this contest, ACPA may behave as osteoclasts activator [40], providing the biological basis for the detection of bone erosive features in ACPApos asymptomatic individuals at risk of RA development [41], as suggested by the periarticular bone changes in terms of thinning and fenestration of the cortical bone together with

Jo

milder changes in the trabecular bone fund in such population [41]. These findings challenged the concept that bone damage in RA is exclusively led by inflammation. Moreover, considering that ACPA production is not merely limited to joint environment but may occur at different anatomical sites as lung epithelium [42] and gingival mucosa [43], to date it is still unclear where and how the loss of immunological tolerance takes place and where the ACPA production by autoreactive plasmacells firstly occurs [44].

5

However, in daily clinical practice only 2/3 of RA are positive for ACPA and/or RF. Sieghart et al. recently investigated the distribution of different isoforms of ACPA and RF in a wide RA cohort finding that the contemporary analysis of multiple antibody isoforms allows to reduce the definition as seronegative to nearly 20%, suggesting that the determination of a wider range of autoantibody isotype is recommended to reduce not only the diagnostic but even the sensitivity gap left by the routine determination limited to IgM-RF and IgG-ACPA only [45]. Moreover, the combination of ACPA and IgM-RF was demonstrated to exert a direct action, in vitro, on the inflammatory status.

ro of

In particular, ACPA/IgM-RF/citrullinated fibrinogen immune complexes were found to increase, in vitro, cytokines release by macrophages (i.e. TNF-, IL-1, IL-6, IL-17), influencing cellular polarization towards pro-inflammatory (M1) phenotype and suggesting a synergistic biological

-p

effect [46]. Finally, the determination of multiple autoantibody isoforms plays a prognostic role in the earliest phase of RA since the combined seropositivity of IgM-RF and IgG-ACPA characterizes

re

early RA patients with high likelihood chance of radiographic progression during the “treat-to-

lP

target” follow-up [47].

1.3 Additional autoantibody positivities in RA

ur na

The aberrant autoimmune response in RA, may be directed against modified antigens others than the ones daily used in clinical practice. Vimentin is an intermediate filament expressed in mesenchymal cells and macrophages of synovial tissues involved in adhesion, migration and

Jo

survival as well as cells signaling during stress and autoantibodies against modified vimentin (MCV) have higher sensitivity (69%) but less specificity (94%) compared to ACPA (62% of sensitivity and 97% of specificity respectively) in RA [48,49]. However, despite limitations of the available studies investigating this issue, the specificity of anti-MCV decreases to 86% if other rheumatic diseases are used as comparison group, being detectable in other conditions [49]. In particular, the combined analysis of anti-MCV, IgA/IgM-RF and ACPA in a wide early-RA cohort (n=370) showed the importance of anti-MCV detection being able to reduce the gap of 6

“seronegative” patients of nearly 10% (personal data). The utility of anti-MCV detection in RA is supported by the evidence that anti-MCV positive RA patients show a higher prevalence of bone erosions regardless to ACPA positivity not only at disease onset but even after one year follow-up [50], maybe due to the induction of osteoclastogenesis and bone loss promoted by anti-MCV presence leading to cell activation and TNF release [39,51]. Interestingly, anti-MCV autoimmunity was demonstrated being associated with periarticular bone loss, synovitis and bone erosions in animal model leading to the decrease of trabecular bone and to the increase of osteoclasts [52].

ro of

These findings support the concept that the presence of autoantibodies against modified antigens before the clinical disease onset, may play an active role on bone biology. However, despite the data presented in multiple studies suggest that autoantibodies can mediate pathological events in

-p

RA through multiple mechanisms, as immune-complexes formation and direct agonistic activity,

extensive experimental validation [53].

re

the activation role of ACPA on cells involved in the disease pathogenesis is going to require more

lP

To date, other mechanisms of antigen modification have been identified not directly enzymatically mediated. Carbamilation is a chemical modification mediated by cyanide in which a lysine residue is converted into homocitrulline, resulting in a self-reacting antigen. Similarly to ACPA, anti-CarP

ur na

antibodies alone are not sufficient to break immune tolerance and induce autoimmunity [54]. However, anti-CarP antibodies are detectable during the pre-symptomatic phase of RA and even in unaffected first-degree relatives, despite their lower sensitivity (42%) and similar specificity (96%)

Jo

compared to ACPA, increase the diagnostic accuracy even in case of RF/ACPA negativity [55-58]. Moreover, the concomitant presence of anti-CarP, RF and ACPA increases the diagnostic power for RA of three folds than the routinely used RF/ACPA combination (OR from 36.7 to 112.2) [59]. Interestingly, despite their strict association with RF/ACPA positivity, anti-CarP antibodies seem not to be dependent to known RA risk factors (i.e. share epitope and smoking) suggesting the need to be investigated to uncover possible novel pathogenetic mechanisms involved in RA [60]. Recently, Truchetet et al. showed that anti-CarP positivity is associated with a more severe clinical 7

and radiographic disease in a cohort of early arthritis patients (ESPOIR cohort). In particular, antiCarPpos arthritis patients had a higher chance of ACR/EULAR 2010 criteria fulfillment among eight years and were more frequently treated with at least one biological DMARD compared to antiCarPneg arthritis patients despite RF/ACPA status [61]. Finally, a new class of autoantibodies against PAD enzymes has been discovered in RA patients. Five different isoforms belonging to PAD enzymes class (PAD1-4 and 6) are known and anti-PAD4 was the first to be discovered [62]. It is well known that PAD4 plays an important role in RA

ro of

pathogenesis contributing to autoantigen citrullination, even if only recently it has been shown to be tightly related to the inflammatory process. In particular, the association between PAD4 and its substrates may result with the loss of tolerance since dying cells or netting neutrophils in RA joint

-p

are able to release enzymatically active PAD4 within the extracellular space where the latter can modify arginine containing proteins in the nearby as collagen or fibrinogen [63]. Anti-PAD4

re

antibodies are detectable in the pre-clinical phase of RA being strongly associated with ACPA and

lP

with radiographic damage and progression [64-66]. Despite similar specificity for RA (96%), antiPAD4 autoantibodies show lower sensitivity (35-38%) than ACPA [67,68]. It was proposed that anti-PAD4 are able to interfere with PAD4 unfolding or directly affecting antigen binding due to

ur na

the modification of the number of citrulline residues derived by PAD4. Controversial data have been produced on the action exerted by anti-PAD4 antibodies on the enzymatic activity of PAD4 itself. Darra et al. demonstrated that a subset of anti-PAD4 antibodies cross-reacting with anti-

Jo

PAD3 are able to increase PAD4 catalytic efficacy through a decrease of enzyme’s calcium requirement [69]. Based on the upcoming data, further investigations are needed to better define the pathogenetic role of such autoimmune response products, mainly in ACPA/RFneg RA.

1.4 Aberrant B lymphocyte activation in RA The presence of autoantibodies and the successful effect of B cell targeted therapy clearly place the adaptive immune cells at the center of RA pathogenesis [70,71]. Specifically, RA patients show, at 8

synovial tissue level, abundant myeloid and plasmacytoid dendritic cells that express cytokines and costimulatory molecules which are involved in antigen presentation, loss of immunological tolerance and T cell activation [72]. Moreover, B lymphocytes exert multiple role in the pathogenesis of RA not limited only to autoantibodies production but also modulating both T and dendritic cells function, promoting ectopic lymphoid neogenesis development as well as the release of inflammatory mediators [73]. Moreover, the presence of T cell oligoclonality and B cell hypermutation, in synovial tissue, already detectable in preclinical phases increasing overtime

ro of

closer to disease onset, suggest the aberrant activation of the adaptive immune system as early event promoting disease development [74-77].

Differential synovial tissue composition characterizes RA patients, from a pauci-immune phenotype

-p

to a follicular well-structured synovitis. Synovial B cells are located in the context of lymphoid aggregates where they undergo to clonal expansion and support the production of autoantibodies

re

[75-77].

lP

Recently, Gerlag and colleagues reported the results of the PRAIRI study, in which, ACPA/RF positive asymptomatic individuals were included in a randomized, double-blind, placebo-controlled study to receive a single infusion of 1000 mg of Rituximab or placebo showing that B cell targeted

ur na

therapy is able to significantly delay the development of arthritis, providing evidence for the pathogenetic role of B cells in the earliest, pre-arthritis stage of autoantibody positive RA [78]. Despite ACPA positivity in the preclinical phase, their presence does not cause apparent pathology,

Jo

since ACPA and/or RF positive individuals do not show proven synovitis, in terms of CD3pos, CD22pos, CD55pos, CD68pos or CD138pos cells [79]. Multiple evidences suggested that alteration of B cells subsets distribution is present at the beginning of RA in peripheral blood compartment [80]. In particular, RA patients with disease duration less than 6 weeks showed lower percentages of memory B cells and pre-switched antigen experienced memory B cells (IgDposCD27pos) than healthy controls [81]. Considering comparative studies enrolling RA patients with different disease durations, higher percentages of switched9

memory B cells and plasmablasts characterized long-standing RA with a strong direct correlation with disease duration [80,82]. It has been proposed that synovial tissue becomes enriched of memory B cells under the influence TNF during the disease course [80,82,83]. Moura RA et al. showed that IgDnegCD27neg memory B cells are suppressed by TNF inhibitor and Tocilizumab treatment in RA supporting the notion that TNF and IL-6 represent crucial stimuli for B cell activation and survival [84]. Recently, a new previously unknown mechanism through which B cells may act on bone damage

ro of

was demonstrated to be mediated by CCL3-TNF signaling that inhibits osteoblast differentiation by the activation of ERK and NF-kB signaling pathways. These findings demonstrated the effect of B lymphocytes on bone formation inhibition through the direct production of multiple osteoblasts

-p

inhibitors in addition to the direct effect on bone exerted by autoantibodies themselves [85].

re

1.5 Synovial tissue features based on the autoimmune profile in RA

lP

During RA synovial tissue inflammation may display various degrees of synovial lining layer hyperplasia, mononuclear cells infiltrates within the sublining areas, stromal cells hyperplasia and vascularity, leading to an extreme heterogeneity among RA patients from pauci-immune to diffuse

ur na

synovial lymphoid neogenesis (follicular synovitis) [76]. synovitis development caused by leukocytes homing from peripheral blood towards synovial tissue is mediated by endothelial cells activation in synovial vessels, whose number is increased and the expression of adhesion molecules

Jo

(as integrins and selectins) is enhanced [86]. Multiple studies have been conducted to dissect the possible differences in terms of synovial tissue inflammation features among RA patients stratified based on the autoantibody positivity with controversial results. Van Oosterhout and colleagues, found that ACPApos long-standing RA patients showed, at synovial tissue level, higher rate of infiltrating lymphocytes, less extent of fibrosis and a thinner lining layer than ACPAneg RA. Moreover, the authors hypothesized that the attraction of leukocytes within the synovial tissue of ACPApos RA may be mediated through CXCL12 [87]. On the contrary, Gómez-Puerta et al. did not 10

report a significant difference in terms of synovial infiltrates and lymphoneogenesis enrolling RA patients with long-standing disease (disease duration more than 40 months). However, the analysis of cytokines milieu in the synovial fluid of RA patients stratified based on ACPA positivity revealed that the synovial fluid of ACPApos RA was enriched of IL-1, IL-10, CCL20, IL-17 and IL-10 compared to ACPAneg RA [88]. Similar results have been obtained stratifying RA patients based on RF positivity [89]. Investigating the synovial features of early RA patients based on ACPA/RF positivity, a higher rate of follicular synovitis was found in seropositive RA patients than

ro of

seronegative ones [90]. In this contest, mast cells (CD117+ cells) can be detected in the synovial tissue of ACPA/RFneg RA patients as recently found to characterize ACPA/RFpos synovitis

-p

promoting autoantibodies production [91,92].

1.6 Epigenetic mechanisms underpinning B cell abnormal function in RA

re

The genetic concordance between monozygotic twins was estimated to be nearly 15%, suggesting

lP

the additional contribution exerted by other factors, as epigenetic changes in the regulation of cellular homeostasis [93]. Epigenetic mechanisms, including covalent modification of DNA (Methylation of cytosine at position C5 in CpG pairs mainly in regulatory regions) and histones

ur na

modifications (methylation and acetylation) able to remodel chromatin leading to activation or silencing of gene transcription are potentially heritable and able to regulate gene expression without DNA sequence changes [93]. In particular, Julià and colleagues performed, in a limited cohort of

Jo

RA patients, an epigenome-wide association study (EWAS) to identify disease-specific alterations in DNA methylation in B cells finding that differential degree of methylation was observed in 64 CpG sites leading to the identification of multiple biological pathways differentially expressed in RA B cells [94]. Recently, microRNA (miRNA) acting as post-transcriptional regulators of gene expression through repression of target mRNA [95], arose as fundamental in the regulation of the innate and the adaptive immune system in RA [96,97]. In particular, miR-155 was confirmed as fine tuner of 11

macrophages/dendritic cells as well as T and B lymphocytes biology [96]. Anaparti and colleagues investigated the peripheral blood microRNA expression signature showing that patients with RA and their seropositive first-degree relatives have similar expression of miR-155 significantly higher than healthy unrelated controls suggesting that the abnormal epigenetic regulation of the autoimmune cascade is an early phenomenon within RA course [98]. Moreover, miR-155 knock out mice are resistant to collagen induced arthritis, bone damage and autoantibody production by B cells [95]. These findings are due to the repressive activity exerted by miR-155 on

ro of

Phosphatidylinositol-3,4,5-trisphosphate 5-phosphatase 1 (SHIP-1) which is an anti-inflammatory molecule in macrophages [99] and on PU.1 that is a transcription factor need for B cell maturation and antibody production [90]. Moreover, at synovial tissue level, RA patients with synovial ectopic

-p

follicular structures show the highest rate of miR-155 compared to RA patients with diffuse/pauciimmune synovitis suggesting that the miR-155/PU.1 axis can be added to the molecular pathways

re

involved in lymphoneogenesis promotion in inflamed tissues in RA. Moreover, considering the B

lP

cell subpopulations in relation to the epigenetic signature, IgDnegCD27neg memory B cells were shown to have the highest endogenous expression of miR-155 compared to other B cell subsets in

ur na

RA patients [90].

1.7 Autoantibodies in clinical remission in RA As stated above, the seropositivity for ACPA is a useful biomarker not only for diagnostic purpose

Jo

but even for prediction of a worse prognosis concerning joint damage [100]. In particular, memory B-cell subset(s) distribution correlates with worse long-term clinical outcomes at disease onset in RA [101]. Importantly, it has been demonstrated that, despite the achievement of clinical remission, through the “treat to target” strategy, ACPA seropositivity generally persists. The seroconversion for IgG-ACPA determining the so called “immunological remission” is a rare phenomenon in clinical practice compared to IgM/IgA isotypes [102]. This phenomenon, over the first year of pharmacological treatment does not translate into a higher chance of drug-free remission in RA 12

[103]. The immune-biologic basis for this persistence is still obscure as ACPA production in ectopic synovial lymphoid tissue is presumably decreased with disease remission. In fact, the analysis of synovial tissue histological composition in RA patients in sustained clinical and ultrasound remission revealed that the rate of synovial lymphoid aggregates is significantly less compared to RA patients at disease onset but not abrogated [104]. Therefore, it can be postulated that ACPA continue to be spontaneously produced by long-lived plasma cells in the synovial tissue, bone marrow or other sites. Pelzek and colleagues have recently investigated the behavior of

ro of

disease-associated autoreactive peripheral B cells in treatment induced clinical remission in RA patients, showing that recirculating ACPA memory B cells generally persist despite DMARD or TNF-inhibition, rationalizing why therapeutic cessation most often results in disease reactivation

-p

and clinical flare [105]. Haschka and colleagues investigated the predictive power of multiple factors associated with disease flare occurrence in RA patients after treatment discontinuation once

re

disease remission is achieved finding that ACPApos RA patients show significant higher change of

lP

disease flare compared to ACPAneg ones [106]. Moreover, ACPA positivity significantly enhances the disease flare risk of more than 3 times, after treatment discontinuation, in RA patients in disease remission if combined with aberrant Multi-Biomarker Disease Activity (MDBA) panel [107].

ur na

Based on these findings, B cell abnormalities in RA should be considered at the time of remission achievement to foresee the risk of disease flare after treatment modifications.

Jo

1.8 Pharmacological effects on B cells in peripheral blood and synovial compartments of RA Based on their crucial effects on cytokines production and antigen presentation, B cells targeted therapy, as Rituximab, is included in the therapeutic armamentarium to treat RA [108]. Rituximab treatment enables a significant reduction of serum levels of IgM/IgA-RF, ACPA and Anti-MCV autoantibodies [109,110] as well as reduction of BCR clones in the peripheral blood of RA [111]. Considering the synovial tissue environment, Rituximab causes a significant reduction in B cells number [112] but not a complete depletion [109,111]. 13

Moreover, other therapies that are not strictly targeting B-cells, showed a significant effect on B cells in terms of cellular maturation and autoantibody production. Among them, conventional Disease Modifying Anti-Rheumatic Drugs (c-DMARDs) as Methotrexate and Sulphasalazine are able to significantly inhibit clonal growth of B cells in vitro and antibody production [113-115]. Moreover, Methotrexate significantly increases global DNA methylation of B cells in RA [116] and their maturation [117] and Methotrexate/Hydroxychloroquine/Sulfasalazine triple therapy significantly represses genes related to plasmablasts/plasma cells differentiation in synovial tissue

ro of

of RA patients [118]. Moura and co-workers showed that TNF-inhibition enables the reduction of IgDneg/CD27neg B cells in the peripheral blood of RA patients [84] despite no significant change in BCR gene rearrangements [119] whereas Bankò and colleagues demonstrated that Etanercept

-p

induces significant increase of IL-10 producing B cells (B10) [120]. Considering the synovial tissue compartment, TNF-inhibitor represses memory B cell network despite the persistence of residual B

re

lymphocytes in the synovial tissue in RA patients in sustained clinical and ultrasound sustained

lP

remission [104,121]. The effects of IL-6R inhibition on B cells biology confirmed the crucial role of IL-6 in the maturation and activation of B cells in RA pathogenesis. In particular, a significant reduction of mutational frequency in immunoglobulin receptor and BCR gene rearrangements were

ur na

found in RA patients treated with IL-6R inhibitor together with a significant decrease of peripheral blood IgDneg/CD27neg B cells [84,119,122] together with a significant reduction of mutational frequency in immunoglobulin receptor and ACPA titer [123,124]. Moreover, IL-6R inhibition was

Jo

demonstrated to significantly reduce B cell count and the expression of immunoglobulin genes in the synovial tissue of RA patients [125,126]. Interestingly, CTLA4-Ig therapy causes a significant reduction of post-switch memory B cells and a decrease of Syk phosphorylation in B cells within the peripheral blood compartment of RA patients with a parallel decrease of ACPA-specific B cell clones and IgG/IgA-ACPA and IgM/IgA/IgG-RF serum levels [127-129]. Finally, multiple research groups recently demonstrated that JAK1/JAK3 blockade causes a strong inhibition of plasmablasts development and immunoglobulin secretion in vitro [130] and a significant decrease of synovial 14

tissue expression of CXCL13 suggesting a possible mechanism by which B cells homing within the target tissue may be reduced in RA [131]. In addition, JAK2/JAK3 inhibition was found to significantly suppresses the differentiation of B cells in plasmablasts [132] despite an early increase of peripheral blood B cell count [133]. Therefore, based on these findings, B cells-derived biomarkers may be useful in the assessment of therapeutic response to different conventional and biological therapies in RA management (Table 1).

ro of

2. Conclusions The terrific progress of research on B cells led new knowledge about the multiple functions exerted by B lymphocytes and the molecular pathways underpinning their abnormal function in RA.

-p

Moreover, the investigation of multiple body compartments (i.e. peripheral blood and synovial tissue) allowed the identification of additional biomarkers associated with the clinical phase and

re

disease phenotype (Figure 1). Based on this, we believe that the use of new advanced technologies

Jo

ur na

lP

will reveal, in the next future, their utility in a deeper personalized management of RA patients.

15

References 1. McInnes IB, Schett G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet 2017;389:2328-2337. 2. Raychaudhuri, S. Recent advances in the genetics of rheumatoid arthritis. Curr Opin Rheumatol 2010;22:109-118. 3. MacGregor AJ, Snieder H, Rigby AS, et al. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum 2000;43:30-7. 4. Gregersen PK, Silver J, Winchester RJ. The shared epitope hypothesis: an approach to understanding the molecular genetics of susceptibility to rheumatoid arthritis. Arthritis Rheum 1987;30:1205-13.

ro of

5. Begovich AB, Carlton VE, Honigberg LA, et al. A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase (PTPN22) is associated with rheumatoid arthritis. Am J Hum Genet 2004;75:330-7. 6. Kurreeman FA, Padyukov L, Marques RB, et al. A candidate gene approach identifies the TRAF1/C5 region as a risk factor for rheumatoid arthritis. PLoS Med 2007;4(9):e278. [Erratum, PLoS Med 2007;4(12):e358]

-p

7. Plenge RM, Cotsapas C, Davies L, et al. Two independent alleles at 6q23 associated with risk of rheumatoid arthritis. Nat Genet 2007;39:1477-82.

re

8. Totaro MC, Tolusso B, Napolioni V, et al. PTPN22 1858C>T polymorphism distribution in Europe and association with rheumatoid arthritis: case-control study and meta-analysis. PLoS One. 2011;6(9):e24292

lP

9. Symmons DP, Bankhead CR, Harrison BJ, et al. Blood transfusion, smoking, and obesity as risk factors for the development of rheumatoid arthritis: results from a primary care-based incident case-control study in Norfolk, England. Arthritis Rheum 1997;40:1955-61.

ur na

10. Klareskog L, Stolt P, Lundberg K, et al. A new model for an etiology of rheumatoid arthritis: smoking may trigger HLA-DR (shared epitope)-restricted immune reactions to autoantigens modified by citrullination. Arthritis Rheum 2006;54:38-46. 11. Gremese E, Carletto A, Padovan M, et al. Obesity and reduction of the response rate to anti-tumor necrosis factor α in rheumatoid arthritis: An approach to a personalized medicine. Arthritis Care Res (Hoboken). 2013;65:94-100. 12. Gremese E, Tolusso B, Gigante MR, et al. Obesity as a risk and severity factor in rheumatic diseases (autoimmune chronic inflammatory diseases). Front Immunol. 2014;5:576.

Jo

13. Tolusso B, Gigante MR, Alivernini S, et al. Chemerin and PEDF Are Metaflammation-Related Biomarkers of Disease Activity and Obesity in Rheumatoid Arthritis. Front Med (Lausanne). 2018;5:207. 14. Crowson CS, Matteson EL, Davis JM III, et al. Contribution of obesity to the rise in incidence of rheumatoid arthritis. Arthritis Care Res 2013;65:71–7. 15. Lahiri M, Luben RN, Morgan C, et al. Using lifestyle factors to identify individuals at higher risk of inflammatory polyarthritis (results from the European Prospective Investigation of Cancer- Norfolk and the Norfolk Arthritis Register-the EPIC-2-NOAR Study). Ann Rheum Dis 2014;73:219–26. 16. Wesley A, Bengtsson C, Elkan AC, et al. Association between body mass index and anti-citrullinated protein antibody- positive and anti-citrullinated protein antibody-negative rheumatoid arthritis: results from a population-based case-control study. Arthritis Care Res (Hoboken) 2013;65:107–12.

16

17. Arnett FC, Edworthy SM, Bloch DA, et al. The American Rheumatism Association 1987

revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum.1988;31:315-24. 18. Aletaha D, Neogi T, Silman AJ, et al. 2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Ann Rheum Dis. 2010;69:1580-8. 19. Barra L, Pope JE, Orav JE, et al. Prognosis of seronegative patients in a large prospective cohort of patients with early inflammatory arthritis. J Rheumatol. 2014;41:2361-9. 20. Nordberg LB, Lillegraven S, Lie E, et al. Patients with seronegative RA have more inflammatory activity compared with patients with seropositive RA in an inception cohort of DMARD-naïve patients classified according to the 2010 ACR/EULAR criteria. Ann Rheum Dis. 2017;76:341-345.

ro of

21. Nordberg LB, Lillegraven S, Aga AB, et al. Comparing the disease course of patients with seronegative and seropositive rheumatoid arthritis fulfilling the 2010 ACR/EULAR classification criteria in a treat-totarget setting: 2-year data from the ARCTIC trial. RMD Open 2018;4:e000752. 22. Akdemir G, Markusse I, Dirven L, et al. Effectiveness of four dynamic treatment strategies in patients with anti-citrullinated antibody-negative rheumatoid arthritis-a randomized trial. RMD Open 2016;25;2:e000143.

-p

23. Ferraccioli G, Tolusso B, Fedele AL, Gremese E. Do we need to apply a T2Tstrategy even in ACPAnegative early rheumatoid arthritis? YES. RMD Open 2016;25;2:e000263.

re

24. Catrina AI, Svensson CI, Malmström V, et al. Mechanisms leading from systemic autoimmunity to jointspecific disease in rheumatoid arthritis. Nat Rev Rheumatol 2017;13:79-86.

lP

25. Schellekens GA, Visser H, de Jong BA, et al. The diagnostic properties of rheumatoid arthritis antibodies recognizing a cyclic citrullinated peptide. Arthritis Rheum 2000;43:155-163.

ur na

26. Nielen MM, van der Horst AR, van Schaardenburg D, et al. Antibodies to citrullinated human fibrinogen (ACF) have diagnostic and prognostic value in early arthritis. Ann Rheum Dis 2005;64:1199-204. 27. Rantapaa-Dahlqvist S, de Jong BA, Berglin E, et al. Antibodies against cyclic citrullinated peptide and IgA rheumatoid factor predict the development of rheumatoid arthritis. Arthritis Rheum 2003: 48:2741-2749. 28. Majka DS, Deane KD, Parrish LA, et al. Duration of preclinical rheumatoid arthritis-related autoantibody positivity increases in subjects with older age at time of disease diagnosis. Ann Rheum Dis 2008:67:801807.

Jo

29. Pietrapertosa D, Tolusso B, Gremese E, et al. Diagnostic performance of anti-citrullinated peptide antibodies for the diagnosis of rheumatoid arthritis: the relevance of likelihood ratios. Clin Chem Lab Med 2010;48:829-34. 30. Brink M, Hansson M, Mathsson L, et al. Multiplex analyses of antibodies against citrullinated peptides in individuals prior to development of rheumatoid arthritis. Arthritis Rheum 2013;65:899-910. 31. Sokolove J, Bromberg R, Deane KD, et al. Autoantibody epitope spreading in the pre-clinical phase predicts progression to rheumatoid arthritis. PLoS One 2012;7:e35296. 32. Deane KD, O’Donnell CI, Hueber W, et al. The number of elevated cytokines/chemokines in pre-clinical seropositive rheumatoid arthritis predicts time to diagnosis in an age-dependent manner. Arthritis Rheum 2010;62:3161-728.

17

33. Kokkonen H, Soderstrom I, Rocklov J, et al. Up-regulation of cytokines and chemokines predates the onset of rheumatoid arthritis. Arthritis Rheum 2010;62:383-391. 34. Arnold JN, Wormald MR, Sim RB, et al. The impact of glycosylation on the biological function and structure of human immunoglobulins. Annu Rev Immunol 2007;25:21-50. 35. Rombouts Y, Ewing E, van de Stadt LA, et al. Anti-citrullinated protein antibodies acquire a proinflammatory Fc glycosylation phenotype prior to the onset of rheumatoid arthritis. Ann Rheum Dis 2015;74:234-41. 36. Clavel C, Nogueira L, Laurent L, et al. Induction of macrophage secretion of tumor necrosis factor alpha through Fcgamma receptor IIa engagement by rheumatoid arthritis-specific autoantibodies to citrullinated proteins complexed with fibrinogen. Arthritis Rheum 2008;58:678-88.

ro of

37. Sokolove J, Zhao X, Chandra PE, et al. Immune complexes containing citrullinated fibrinogen costimulate macrophages via Toll-like receptor 4 and Fcgamma receptor. Arthritis Rheum 2011;63:53-62. 38. Trouw LA, Haisma EM, Levarht EW, et al. Anti-cyclic citrullinated peptide antibodies from rheumatoid arthritis patients activate complement via both the classical and alternative pathways. Arthritis Rheum 2009;60:1923-31.

-p

39. Harre U, Georgess D, Bang H, et al. Induction of osteoclastogenesis and bone loss by human autoantibodies against citrullinated vimentin. J Clin Invest 2012;122:1791-802.

re

40. Krishnamurthy A, Joshua V, Haj Hensvold A, et al. Identification of a novel chemokine-dependent molecular mechanism underlying rheumatoid arthritis-associated autoantibody-mediated bone loss. Ann Rheum Dis 2016;75:721-9.

lP

41. Kleyer A, Finzel S, Rech J, et al. Bone loss before the clinical onset of rheumatoid arthritis in subjects with anticitrullinated protein antibodies. Ann Rheum Dis 2014;73:854-60. 42. Malmström V, Catrina AI, Klareskog L. The immunopathogenesis of Seropositive rheumatoid arthritis: from triggering to targeting Nature Reviews Immunology 2017;17:60-75.

ur na

43. Li S, Yu Y, Yue Y, et al. Autoantibodies From Single Circulating Plasmablasts React With Citrullinated Antigens and Porphyromonas gingivalis in Rheumatoid Arthritis. Arthritis Rheumatol 2016;68:614-26. 44. Jimenez-Boj E, Redlich K, Türk B, et al. Interaction between synovial inflammatory tissue and bone marrow in rheumatoid arthritis. J Immunol 2005;175:2579-88 45. Sieghart D, Platzer A, Studenic P, et al. Determination of autoantibody isotypes increases the sensitivity of serodiagnostics in Rheumatoid Arthritis. Front Immunol 2018;9:876

Jo

46. Sokolove J, Johnson DS, Lahey LJ, et al. Rheumatoid factor as a potentiator of anti-citrullinated protein antibody-mediated inflammation in rheumatoid arthritis. Arthritis Rheumatol 2014;66:813-21. 47. Fedele AL, Petricca L, Tolusso B, et al. Interleukin-6 and IgA-rheumatoid factor are crucial for baseline erosiveness, and anti-citrullinated peptide antibodies for radiographic progression in early rheumatoid arthritis treated according to a treat-to-target strategy. Scand J Rheumatol 2018;47:351-359. 48. Musaelyan A, Lapin S, Nazarov V, et al. Vimentin as antigenic target in autoimmunity: A comprehensive review. Autoimm Rev 2018;17:926-934. 49. Lee YH, Bae SC, Song GG. Diagnostic accuracy of anti-MCV and anti-CCP antibodies in rheumatoid arthritis: A meta-analysis. Z Rheumatol 2015;74:911-8.

18

50. Montes A, Perez-Pampin E, Calaza M, et al. Association of anti-citrullinated vimentin and anticitrullinated α-enolase antibodies with subsets of rheumatoid arthritis. Arthritis Rheum 2012;64:3102-10. 51. Degboé Y, Constantin A, Nigon D, et al. Predictive value of autoantibodies from anti-CCP2, anti-MCV and anti-human citrullinated fibrinogen tests, in early rheumatoid arthritis patients with rapid radiographic progression at 1 year: results from the ESPOIR cohort. RMD Open 2015;1:e000180. 52. Engdahl C, Bang H, Dietel K, et al. Periarticular bone loss in arthritis is induced by Autoantibodies Against Citrullinated Vimentin. J Bone and Mineral Res 2015;32:1681-91. 53. Toes R, Pisetsky DS. Pathogenic effector functions of ACPA: where do we stand? Ann Rheum

Dis 2019;78:716-21. 54. Alivernini S, Galeazzi M, Peleg H, et al. Is ACPA positivity the main driver for rheumatoid arthritis treatment? Pros and cons. Autoimmun Rev 2017;16:1096-1102.

ro of

55. Shi J, van de Stadt LA, Levarht EW, et al. Anti-carbamylated protein antibodies are present in arthralgia patients and predict the development of rheumatoid arthritis. Ann Rheum Dis 2013;65:911-5. 56. Shi J, van Steenbergen HW, van Nies JA, et al. The specificity of anti-carbamylated protein antibodies for rheumatoid arthritis in a setting of early arthritis. Arthritis Res Ther 2015;17:339.

-p

57. Li L, Deng C, Chen S, et al. Meta-Analysis: Diagnostic accuracy anti-Carbamylated Protein Antibody for Rheumatoid Arthritis. PloS One 2016;11:e0159000.

re

58. Alessandri C, Bartosiewicz I, Pendolino M, et al. Anti-carbamylated protein antibodies in unaffected first-degree relatives of rheumatoid arthritis patients: lack of correlation with anti-cyclic citrullinated protein antibodies and rheumatoid factor. Clin Exp Rheumatol 2015;33:824-30.

lP

59. Mahler M, Fritzler MJ. Diagnostic Utility of Anticarbamylated Protein Antibodies as Measured Using Carbamylated Fetal Calf Serum. J Rheumatol 2018;45:444-445.

ur na

60. Jiang X, Trouw LA, van Wesemael TJ, et al. Anti-CarP antibodies in two large cohorts of patients with rheumatoid arthritis and their relationship to genetic risk factors, cigarette smoking and other autoantibodies. Ann Rheum Dis 2014;73:1761-8. 61. Truchetet ME, Dublanc S, Barnetche T, et al. Association of the Presence of Anti-Carbamylated Protein Antibodies in Early Arthritis With a Poorer Clinical and Radiologic Outcome: Data From the French ESPOIR Cohort. Arthritis Rheum 2017;69:2292-2302.

Jo

62. Nissinen R, Paimela L, Julkunen H, et al. Peptidylarginine deiminase, the arginine to citrulline converting enzyme, is frequently recognized by sera of patients with rheumatoid arthritis, systemic lupus erythematosus and primary Sjögren syndrome. Scand J Rheumatol 2003;32:337-42. 63. Spengler J, Lugonja B, Ytterberg AJ, et al. Release of Active Peptidyl Arginine Deiminases by Neutrophils Can Explain Production of Extracellular Citrullinated Autoantigens in Rheumatoid Arthritis Synovial Fluid. Arthritis Rheumatol 2015;67:3135-45. 64. Harris ML, Darrah E, Lam GK, et al. Association of autoimmunity to peptidyl arginine deiminase type 4 with genotype and disease severity in rheumatoid arthritis. Arthritis Rheumatol 2008;58:1958-67. 65. Halvorsen EH, Pollmann S, Gilboe IM, et al. Serum IgG antibodies to peptidylarginine deiminase 4 in rheumatoid arthritis and associations with disease severity. Ann Rheum Dis 2008;67:414-7. 66. Kolfenbach JR, Deane KD, Derber LA, et al. Autoimmunity to peptidyl arginine deiminase type 4 precedes clinical onset of rheumatoid arthritis. Arthritis Rheumatol 2010;62:2633-9.

19

67. Reyes-Castillo Z, Muñoz-Valle JF, Llamas-Covarrubias MA. Clinical and immunological aspects of antipeptidylarginine deiminase type 4 (anti-PAD4) autoantibodies in rheumatoid arthritis. Autoimmunity Review 2018;17:94-102. 68. Martinez-Prat L, Lucia D, Ibarra C, et al. Antibodies targeting protein-arginine deiminase 4 (PAD4) demonstrate diagnostic value in rheumatoid arthritis. Ann Rheum Dis 2019;78:434-436. 69. Darrah E, Giles JT, Ols ML, et al. Erosive rheumatoid arthritis is associated with antibodies that activate PAD4 by increasing calcium sensitivity. Sci Transl Med 2013;5:185ra65. 70. Ferraccioli G, Tolusso B, Gremese E. Could we use a lower dose of rituximab to treat rheumatoid arthritis in clinical practice: pros and cons? Arthritis Res Ther. 2016;18:126.

ro of

71. Ferraccioli G, Tolusso B, Bobbio-Pallavicini F, et al. Biomarkers of good EULAR response to the B cell depletion therapy in all seropositive rheumatoid arthritis patients: clues for the pathogenesis. PLoS One. 2012;7:e40362. 72. Lebre MC, Jongbloed SL, Tas SW, et al. Rheumatoid arthritis synovium contains two sub- sets of CD83DC-LAMP- dendritic cells with distinct cytokine profiles. Am J Pathol 2008;172:940-50.

-p

73. Martin F, Chan AC. B cell immunobiology in disease: evolving concepts from the clinic. Annu Rev Immunol 2006;24:467-496.

re

74. Schröder AE, Greiner A, Seyfert C, et al. Differentiation of B cells in the nonlymphoid tissue of the synovial membrane of patients with rheumatoid arthritis. Proc Natl Acad Sci U S A 1996;93:221-5. 75. Cantaert T, Brouard S, Thurlings RM, et al. Alterations of the synovial T cell repertoire in anticitrullinated protein anti- body-positive rheumatoid arthritis. Arthritis Rheum 2009;60:1944-56.

lP

76. Humby F, Bombardieri M, Manzo A, et al. Ectopic lymphoid structures support ongoing production of class-switched autoantibodies in rheumatoid synovium. PLoS Med 2009;6:e1.

ur na

77. Tak PP, Doorenspleet ME, de Hair MJH, et al. Dominant B cell receptor clones in peripheral blood predict onset of arthritis in individuals at risk for rheumatoid arthritis. Ann Rheum Dis 2017;76:1924-1930. 78. Gerlag DM, Safy M, Maijer KI, et al. Effects of B-cell directed therapy on the preclinical stage of rheumatoid arthritis: the PRAIRI study. Ann Rheum Dis 2019;78:179-185. 79. van de Sande MG, de Hair MJ, van der Leij C, et al. Different stages of rheumatoid arthritis: features of the synovium in the preclinical phase. Ann Rheum Dis 2011;70:772-7.

Jo

80. Fedele AL, Tolusso B, Gremese E, et al. Memory B cell subsets and plasmablasts are lower in early than in long-standing rheumatoid arthritis. BMC Immunol 2014;15:28. 81. Moura RA, Weinmann P, Pereira PA, et al. Alterations on peripheral blood B-cell subpopulations in very early arthritis patients. Rheumatology (Oxford) 2010;49:1082-92. 82. Fekete A, Soos L, Szekanecz Z, et al. Disturbances in B- and T-cell homeostasis in rheumatoid arthritis: suggested relationships with antigen-driven immune responses. J Autoimmunity 2007;29:154-63. 83. Souto-Carneiro MM, Mahadevan V, Takada K, et al. Alterations in peripheral blood memory B cells in patients with active rheumatoid arthritis are dependent on the action of tumour necrosis factor. Arthritis Res Ther 2009;11:R84.

20

84. Moura RA, Quaresma C, Vieira AR, et al. B-cell phenotype and IgD-CD27- memory B cells are affected by TNF-inhibitors and tocilizumab treatment in rheumatoid arthritis. PloS One 2017;12:e0182927. 85. Sun W, Meednu N, Rosenberg A, et al. B cells inhibit bone formation in rheumatoid arthritis by suppressing osteoblast differentiation. Nat Commun 2018;9:5127. 86. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011;365:2205-19. 87. van Oosterhout M, Bajema I, Levarht EW, et al. Differences in synovial tissue infiltrates between anticyclic citrullinated peptide-positive rheumatoid arthritis and anti-cyclic citrullinated peptide-negative rheumatoid arthritis. Arthritis Rheumatol 2008;58:53-60. 88. Gómez-Puerta JA, Celis R, Hernández MV, et al. Differences in synovial fluid cytokine levels but not in synovial tissue cell infiltrate between anti-citrullinated peptide/protein antibody-positive and -negative rheumatoid arthritis patients. Arthritis Res Ther 2013;15:R182.

ro of

89. Fujinami M, Sato K, Kashiwazaki S, et al. Comparable histological appearance of synovitis in seropositive and seronegative rheumatoid arthritis. Clin Exp Rheumatol 1997;15:11-7. 90. Alivernini S, Kurowska-Stolarska M, Tolusso B, et al. MicroRNA-155 influences B-cell function through PU.1 in rheumatoid arthritis.Nat Commun 2016 27;7:1297.

-p

91. Rivellese F, Mauro D, Nerviani A, et al. Mast cells in early rheumatoid arthritis associate with disease severity and support B cell autoantibody production. Ann Rheum Dis 2018;77:1773-1781.

re

92. Alivernini S, Pugliese D, Tolusso B, et al. Paradoxical arthritis occurring during anti-TNF in patients with inflammatory bowel disease: histological and immunological features of a complex synovitis. RMD Open. 2018;4:e000667.

lP

93. van der Woude, D, Houwing-Duistermaat JJ, Toes RE, et al. Quantitative heritability of anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis. Arthritis Rheum 2009;60:916-923.

ur na

94. Julià A, Absher D, López-Lasanta M et al. Epigenome-wide association study of rheumatoid arthritis identifies differentially methylated loci in B cells. Hum Mol Genet 2017;26:2803-2811. 95. Filipowicz W, Bhattacharyya SN, Sonenberg, N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet 2008;9:102-114. 96. Alivernini S, Gremese E, McSharry C, et al. MicroRNA-155-at the Critical Interface of Innate and Adaptive Immunity in Arthritis. Front Immunol 2018;8:1932.

Jo

97. Elmesmari A, Fraser AR, Wood C, et al. MicroRNA-155 regulates monocyte chemokine and chemokine receptor expression in Rheumatoid Arthritis. Rheumatology (Oxford) 2016;55:2056-2065. 98. Anaparti V, Smolik I, Meng X, et al. Whole blood microRNA expression pattern differentiates patients with rheumatoid arthritis, their seropositive first-degree relatives, and healthy unrelated control subjects. Arthritis Res Ther. 2017;19:249. 99. Kurowska-Stolarska M, Alivernini S, Ballantine LE, et al. MicroRNA-155 as a proinflammatory regulator in clinical and experimental arthritis. Proc Natl Acad Sci U S A 2011;108:11193-8. 100. Singh JA, Furst DE, Bharat A, et al. 2012 update of the 2008 American College of Rheumatology recommendations for the use of disease-modifying antirheumatic drugs and biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res (Hoboken) 2012; 64:625-639.

21

101. Silverman GJ, Pelzek A. Rheumatoid arthritis clinical benefits from abatacept, cytokine blockers, and rituximab are all linked to modulation of memory B cell responses. J Rheumatol. 2014; 41:825-828. 102. Schett G, Emery P, Tanaka Y, et al. Tapering biologic and conventional DMARD therapy in rheumatoid arthritis: current evidence and future directions. Ann Rheum Dis 2016;75:1428–37. 103. de Moel EC, Derksen VFAM, Trouw LA, et al. In RA, becoming seronegative over the first year of treatment does not translate to better chances of drug-free remission. Ann Rheum Dis. 2018 Dec;77(12):1836-1838. 104. Alivernini S, Tolusso B, Petricca L, et al. Synovial features of patients with rheumatoid arthritis and psoriatic arthritis in clinical and ultrasound remission differ under anti-TNF therapy: a clue to interpret different chances of relapse after clinical remission? Ann Rheum Dis 2017;76:1228-1236.

ro of

105. Pelzek AJ, Grönwall C, Rosenthal P, et al. Persistence of Disease-Associated Anti-Citrullinated Protein Antibody-Expressing Memory B Cells in Rheumatoid Arthritis in Clinical Remission. Arthritis Rheumatol. 2017;69:1176-1186. 106. Haschka J, Englbrecht M, Hueber AJ, et al. Relapse rates in patients with rheumatoid arthritis in stable remission tapering or stopping antirheumatic therapy: interim results from the prospective randomised controlled RETRO study. Ann Rheum Dis 2016;75:45-51.

-p

107. Rech J, Hueber AJ, Finzel S,et al. Prediction of disease relapses by multibiomarker disease activity and autoantibody status in patients with rheumatoid arthritis on tapering DMARD treatment. Ann Rheum Dis 2016;75:1637-44.

re

108. Smolen JS, Landewè R, Bijlsma J, et al. EULAR Recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann Rheum Dis 2017;76:960-977.

lP

109. Thurlings RM, Vos K, Wijbrandts CA, et al. Synovial tissue response to rituximab: mechanism of action and identification of biomarkers of response. Ann Rheum Dis 2008;67:917-25.

ur na

110. Fabris M, De Vita S, Blasone N, et al. Serum level of anti-CCp antibodies, anti-MCV andibodies and RF IgA in the follow-up of patients with rheumatoid arthritis treated with rituximab. Auto Immun Highlights 2010;1:87-94. 111. Pollastro S, Klarenbeek PL, Doorenspleet ME, et al. Non-response to rituximab therapy in rheumatoid arthritis is associated with incomplete disruption of the B cell receptor repertoire. Ann Rheum Dis 2019;78:1339-1345.

Jo

112. Vos K, Thurlings RM, Wijbrandts CA, et al. Early effects of rituximab on the synovial cell infiltrate in patients with rheumatoid arthritis. Arthritis Rheumatol 2007;56:772-8. 113. Hirohata S, Ohshima N, Yanagida T, et al. Regulation of human B cell function by sulfasalazine and its metabolites. Int Immunopharmacol 2002;2:631-40. 114. Imai F, Suzuki T, Ishibashi T, et al. Effect of sulfasalazine on B cell hyperactivity in patients with rheumatoid arthritis. J Rheumatol 1994;21:612-5. 115. Nakajima A, hakoda M, Yamanaka H, et al. Divergent effect of methotrexate on the clonal growth of T and B lymphocytes and synovial adherent cells from patients with rheumatoid artrhritis. Ann Rheum Dis 1996;55:237-42.

22

116. de Andres MC, Perez-Pampin F, Calaza M, et al. Assessment of global DNA methylation in peripheral blood cell aubpopulations of early rheumatoid arthritis before and after methotrexate. Arthritis Res Therapy 2015;17:233. 117. McComish J, Mundy J, Sullivan T, et al. Changes in peripheral blood B cell subsets at diagnosis and after treatment with disease-modifying anti-rheumatic drugs in patients with rheumatoid arthritis: correlation with clinical and laboratory parameters. Int J Rheum Dis 2015;18:421-32. 118. Walsh AM, Wechalekar MD, Guo Y, et al. Triple DMARD treatment in early rheumatoid arthritis modulates synovial T cell activation and plasmablast/plasma cell differentiation pathways. PLoS One 2017;12:e0183928. 119. Mahmood Z, Muhammad K, Schmalzing M, et al. CD27-IgD- memory B cells are modulated by in vivo interleukin-6 receptor (IL-6R) blockade in rheumatoid arthritis. Arthritis Res Therapy 2015; 17:61.

ro of

120. Bankò Z, pozsgay J, Gáti T, Rojkovich B, et al. Regulatory B cells in rheumatoid arthritis: Alterations in patients receiving anti-TNF therapy. Clin Immunol 2017;184:63-69. 121. Anolik JH, Ravikumar R, Barnard J, et al. Cutting edge: anti-tumor necrosis factor therapy in rheumatoid arthritis inhibits memory B lymphocytes via effects on lymphoid germinal centers and follicular dendritic cell networks. J Immunol 2008; 180:688-92.

-p

122. Nakayamada S, Kubo S, Yoshikawa M, et al. Differential effect of biological DMARDs on peripheral immune cell phenotypes in patients with rheumatoid arthritis. Rheumatology (Oxford) 2018;57:164-174.

re

123. Muhammad K, Roll P, Seibold T, et al. Impact of IL-6 receptor inhibition on human memory B cells in vivo: impaired somatic hypermutation in preswitch memory B cells and modulation targeting in memory B cells. Ann Rheum Dis 2011;70:1507-10.

lP

124. Noguchi A, yasuda S, Hisada R, et al. Anti-cyclic citrullinated peptide antibody titers decrease in rheumatoid arthritis patients treated with tocilizumab: A pilot study. Mod Rheumatology 2019;21:1-6. 125. Kanbe K, Chen Q, Nakamura A, Hobo K. Inhibition of MAP kinase in synovium by treatment with tocilizumab in rheumatoid arthritis. Clin Rheumatology 2011;30:1407-13

ur na

126. Ducreux J, Durez P, Galant C, et al. Global molecular effect of tocilizumab therapy in rheumatoid arthritis synovium. Arthritis Rheumatol 2014;66:15-23. 127. Scarsi M, Paolini L, Ricotta D, et al. Abatacept reduces levels of switched memory B cells, autoantibodies, and immunoglobulins in patients with rheumatoid arthritis. J Rheumatol 2014;41:666-72.

Jo

128. Iwata S, Nakayamada S, Fukuyo S, et al. Activation of Syk in peripheral blood B cells in patients with rheumatoid arthritis: a potential target for abatacept therapy. Arthritis and Rheumatol 2015;67:63-73. 129. Lorenzetti R, janowska I, Smulski CR, et al. Abatacept modulates CD80 and CD86 expression and memory formation in human B-cells. J Autoimmun 2019;101:145-152. 130. Rizzi M, Lorenzetti R, Fischer K, et al. Impact of tofacitinib treatment on human B-cells in vitro and in vivo. J Autoimmunity 2017;77:55-66. 131. Boyle DL, Soma K, Hodge J, et al. The JAK inhibitor tofacitinib suppresses synovial JAK1-STAT signaling in rheumatoid arthritis. Ann Rheum Dis 2015;74:1311-6. 132. Kubo S, Nakayamada S, Sakata K, et al. Janus kinase inhibitor baricitinib modulates human innate and adaptive immune system. Front Immunol 2018;9:1510.

23

Jo

ur na

lP

re

-p

ro of

133. Tanaka Y, McInnes IB, Taylor PC, et al. Characterization and changes of lymphocyte subsets in baricitinib-treated patients with rheumatoid arthritis: An integrated analysis. Arthritis Rheumatol 2018;70:1923-1932.

24

Figure and figure legend.

lP

re

-p

ro of

Figure 1. B cell aberrant activation in peripheral blood and synovial compartments in RA.

ur na

Figure 1 legend. The B cell phenotype analysis in the peripheral blood compartment in RA patients revealed the dynamics of B cell subpopulations distribution consistently with disease phase and treatment response. Autoantibodies as ACPA and RF may be detected years before the onset of clinical symptoms and may decrease based on the different isotypes at the time of remission achievement (immunological remission).

Jo

Moreover, the test of additional autoantibody specificities (i.e. anti-MCV, anti-CarP and anti-PAD4), other than ACPA and RF, allows the reduction of the seronegative immunological gap in RA. The new advances in synovial tissue collection, through mini-invasive techniques, allowed the study of synovial tissue inflammation. Different synovitis pathotypes have been found in RA with a higher rate of follicular synovitis in ACPApos compared to ACPAneg RA patients, with a differential rate of response to targeted therapies and a negative impact on bone damage in the former group. In particular, resident synovial B cells, within ectopic lymphoid structures, are enriched of miR-155, whose increase promotes cellular activation, maturation and

25

autoantibody release. RA: Rheumatoid Arthritis; ACPA: Anti-Citrullinated Peptide Antibody; miR: microRNA; MCV: Modified Citrullinated Vimentin; CarP: Carbamylated Protein; PAD: Peptidyl Arginine

Jo

ur na

lP

re

-p

ro of

Deiminase; RF: Rheumatoid Factor; CD: Cluster Designation.

26

ro

of

Table 1. Pharmacological effects on B cells compartment and autoantibodies of conventional and biological therapies used for RA.

PERIPHERAL BLOOD

SYNOVIAL TISSUE

Conventional-DISEASE MODIFYING ANTI-RHEUMATIC DRUGS (c-DMARDs)

re

-p

Sulfasalazine suppresses the production of IgM and IgG, represses B cell hyperactivity [113,114] Methotrexate reduces B cells clonal growth and increases global DNA methylation in B cells [115,116] DMARDs reduce the absolute numbers of total B cells, switched memory B cells, CD21low cells, transitional B cells and plasmablasts [117]

Methotrexate/Hydroxychloroquine/Sulphasalazine triple therapy significantly represses genes related to plasmablasts/plasma cells differentiation [118]

TNF-INHIBITOR

al P

Reduction of IgDneg/CD27neg B cells [84] No effect on mutational frequency of BCR gene rearrangements at week 12, 24 and 52 in RA patients [119] Significant increase of IL-10 productive B cells after 6 months without difference in ACPA serum levels [120]

IL-6R INHIBITOR

Significant reduction of mutational frequency in Immunoglobulin Receptor [123] Significant reduction in mutational frequency of BCR gene rearrangements [119] Significant reduction of IgDnegCD27neg B cells [84,119,122] Significant reduction of ACPA titer in RA after 24 weeks treatment [124]

ur n

Persistence of B cells in the sublining of RA in sustained clinical and ultrasound remission [104] Inhibits memory B lymphocytes via effects on lymphoid germinal centers and follicular cell network [121] Reduction of immunoglobulin genes and CD20 [125,126]

CTLA4-Ig

Jo

Significant reduction of IgG/IgA/IgM and free light chains plasma serum levels in RA. Significant reduction of IgG/IgA-ACPA and IgM/IgA and IgG-RF in RA. Significant reduction of post-switch memory B cells in RA [127] Significant reduction of phosphorylation of Syk in B cells of RA [128] Significant decrease of ACPA-specific switched memory B cells after treatment [129]

RITUXIMAB

Serum levels of IgM- RF and ACPA decreases significantly at 24 and 36 weeks respectively [109] Significant reduction of ACPA, anti-MCV and IgA-RF [110] After 4 weeks, the peripheral blood BCR repertoire of treated patients consisted of fewer, more dominant and more mutated BCR clones [111]

Synovial B cells were significantly decreased, but were not completely depleted in all patients [109,112] No significant changes in the synovial tissue BCR repertoire until week 16, when a reduced clonal overlap with baseline and an increased mutation load is observed [111]

JAK-inhibitor Tofacitinib strongly inhibits plasmablasts development and immunoglobulin secretion in vitro [130] B cells increase after 4 weeks of Baricitinib [133]

Tofacitinib significantly reduced synovial mRNA expression of MMP-1, MMP-3 and chemokines CCL2, CXCL10 and

CXCL13 [131]

of

Baricitinib suppresses the differentiation of human B cells into plasmablasts by B cell receptor and type-I IFN stimuli and inhibits the production of IL-6 from B cells [132]

ro

Table 1 legend. BCR: B cell Receptor; ACPA: Anti-Citrullinated Peptide Antibodies; RF: Rheumatoid Factor; MCV: Mutated Citrullinated Vimentin; MMP:

Jo

ur n

al P

re

-p

metalloproteinases; RA: Rheumatoid Arthritis; JAK: Janus Kinase.

28