Next Article in Journal
Protective Effects of Lactobacillus plantarum Lac16 on Clostridium perfringens Infection-Associated Injury in IPEC-J2 Cells
Next Article in Special Issue
Rheumatoid Arthritis: Pathogenic Roles of Diverse Immune Cells
Previous Article in Journal
Molecular Mechanisms of Possible Action of Phenolic Compounds in COVID-19 Protection and Prevention
Previous Article in Special Issue
Epigenetic Regulation (Including Micro-RNAs, DNA Methylation and Histone Modifications) of Rheumatoid Arthritis: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Genetic, Environmental, and Immunopathological Complexity of Autoantibody-Negative Rheumatoid Arthritis

by
Ludovico De Stefano
1,2,
Bernardo D’Onofrio
1,2,
Antonio Manzo
1,2,
Carlomaurizio Montecucco
1,2 and
Serena Bugatti
1,2,*
1
Division of Rheumatology, IRCCS Policlinico San Matteo Foundation, Viale Golgi 19, 27100 Pavia, Italy
2
Department of Internal Medicine and Therapeutics, University of Pavia, Via Aselli 43/45, 27100 Pavia, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(22), 12386; https://doi.org/10.3390/ijms222212386
Submission received: 13 October 2021 / Revised: 15 November 2021 / Accepted: 15 November 2021 / Published: 17 November 2021
(This article belongs to the Special Issue Molecular Mechanisms of Rheumatoid Arthritis)

Abstract

:
Differences in clinical presentation, response to treatment, and long-term outcomes between autoantibody-positive and -negative rheumatoid arthritis (RA) highlight the need for a better comprehension of the immunopathogenic events underlying the two disease subtypes. Whilst the drivers and perpetuators of autoimmunity in autoantibody-positive RA have started to be disclosed, autoantibody-negative RA remains puzzling, also due its wide phenotypic heterogeneity and its possible misdiagnosis. Genetic susceptibility appears to mostly rely on class I HLA genes and a number of yet unidentified non-HLA loci. On the background of such variable genetic predisposition, multiple exogeneous, endogenous, and stochastic factors, some of which are not shared with autoantibody-positive RA, contribute to the onset of the inflammatory cascade. In a proportion of the patients, the immunopathology of synovitis, at least in the initial stages, appears largely myeloid driven, with abundant production of proinflammatory cytokines and only minor involvement of cells of the adaptive immune system. Better understanding of the complexity of autoantibody-negative RA is still needed in order to open new avenues for targeted intervention and improve clinical outcomes.

1. Introduction

Rheumatoid arthritis (RA) is a multifactorial chronic immune-inflammatory disease characterized by significant heterogeneity in clinical presentation and outcomes among different individuals with the same formal diagnosis. A major subclassification is based on the presence or absence of classical RA-associated autoantibodies, such as rheumatoid factor (RF) and anti-citrullinated protein antibodies (ACPAs). Traditionally, autoantibody-positive RA, if left untreated, is associated with worst outcomes in terms of high disease activity, rapidly progressive joint damage, and increased mortality [1,2]. In the past 10 years, however, earlier diagnosis and more intensive management with conventional synthetic (cs), biologic (b), and targeted synthetic (ts) disease-modifying anti-rheumatic drugs (DMARDs) have considerably improved the prognosis of RA, with milder disease course achieved in particular in autoantibody-positive patients [3]. In contrast, patients with autoantibody-negative RA still experience delayed diagnosis [4] and highly heterogeneous response to therapy [5], and some continue to develop extensive joint destruction and disability. Indeed, over the years, it has become increasingly apparent that autoantibody-negative RA is a more heterogeneous entity than autoantibody-positive RA, and that the current classification criteria and treatment approaches may still be insufficient at conveying favorable outcomes in a proportion of the patients.
The pathophysiological basis underlying the clinical diversity of RA is only partially understood. In particular, it is at present poorly defined whether autoantibody-positive and -negative RA are sustained by heterogeneous immune mechanisms operating variably, albeit with some overlap, in individual patients. Intensive research into the etiopathogenesis of ACPA-positive RA over the last years has substantially clarified the complex interplay between genetic risk factors and environmental susceptibility resulting in dysregulated adaptive immunity with the generation of pathogenic autoantibodies [6,7]. In contrast, the genetic architecture and the predisposing factors of autoantibody-negative RA remain an area of uncertainty, and the relative contribution of innate over adaptive immune pathways has been hypothesized but not formally proven [8,9]. Although RA is currently managed the same way irrespective of the autoantibody profile, a better understanding of the pathophysiological heterogeneity of the disease would definitively improve personalized approaches more focused on specific risk factors and immune pathways. Here, we will revise current advancements in the understanding of the genetic, environmental, and immunopatogenetic complexity of autoantobody-negative RA.

2. Genetic Susceptibility

The contribution of genetic factors to the susceptibility of ACPA-positive and ACPA-negative RA was initially estimated to be equivalent in two small twin studies [10,11]. However, in a recent study using large population-representative samples, the heritability calculation was revised and reported to be 50% for ACPA-positive and 20% for ACPA-negative RA [12]. Despite the lowest susceptibility, autoantibody-negative RA has still been reported to significantly co-aggregate with both autoantibody-positive RA and with spondyloarthropathies in first-degree relatives [13]. This finding highlights the complexity of autoantibody-negative RA, with some cases genetically similar to autoantibody-positive RA but possibly with undetected antibodies, and some others etiologically more related to classical seronegative arthritides.
HLA genes typically play the largest contribution to inherited disease susceptibility [14]. The strongest genetic loci reported for autoantibody-positive RA, specifically the shared epitope-containing HLA-DRB1 alleles, are minimally or not associated with autoantibody-negative RA [15]. Rather, HLA associations appear mostly located within the class I region [14] (Table 1). In particular, studies have consistently reported polymorphic aspartate at position 9 of the class I HLA-B protein, which is strongly correlated with the expression of HLA-B*08 [16,17,18,19]. The same studies also identified serine at position 11 of the class II HLA-DRB1 protein. However, this association is partially explained by the existence of a common extended haplotype, the ancestral haplotype 8.1, which contains HLA-B*08 with aspartate at position 9 and DRB1*03 with serine at position 11. The existence of such linkage disequilibrium might justify previously reported DR3 associations with autoantibody-negative RA [20,21]. From a pathophysiological perspective, variations in HLA class I alleles would underscore the possible predominant role of CD8+ over CD4+ T cells in autoantibody-negative RA [22,23]. Still, HLA-DRB1 haplotypes would appear to play a role also in autoantibody-negative RA by shaping the autoantibody response. The presence of basic aminoacids, such as histidine, at position 13 indeed strongly confers risk of ACPA production [24], whilst not charged or negatively charged aminoacids have been reported to be associated with autoantibody-negative juvenile and adult polyarthritis [25]. Genetic polymorphisms within the HLA region, however, explain only a modest proportion of autoantibody-negative RA heritability. Genome-wide association studies have identified a number of non-HLA determinants at confirmed levels of statistical significance. However, most of the genetic associations have been reported in single studies and have not been independently replicated. Overall, it would appear that a number of non-HLA susceptibility loci are more specific of ACPA-positive RA (AFF3, CCR6, CCL21, IL2RA, and CD28), some are shared irrespective of the autoantibody serotype (TNFAIP3, C5orf30, STAT4, ANKRD55, BLK, and PTPN22), and others are more associated with autoantibody-negative RA (CLYBL, SMIM2, SPP1, CLEC16A, IRF5, and DCIR) [16,26,27,28,29]. Of the markers reported in ACPA-negative RA, ANKRD55 is the only locus to be associated at genome-wide significance levels [16,29]. Although the biologic function of ANKRD55 gene products remains unclarified, single nucleotide polymorphisms in ANKRD55 are risk factors for multiple immune diseases, including multiple sclerosis, Crohn’s Disease, diabetes, and inflammatory myopathies [30]. Recently, novel non-additive loci related to the metabolism of vitamin D, such as DHCR7 and IFR4 mutations, have been proposed [31], but their association with autoantibody-negative RA remains to be replicated. It is also worth mentioning that some forms of autoantibody-negative polyarthritis classified as RA may in fact share genetic similarities with autoinflammatory diseases, including mutations in NLRP3, MEFV, NOD2, or TNFAIP3 genes [8].

3. Environmental and Lifestyle Factors

Environmental susceptibility factors in autoantibody-negative RA are thought to be diverse and variably present among patients. Numerous pieces of evidence support the precipitating role of environmental and internal factors, such as respiratory diseases, exposure to toxins, dysbiosis, and biological responses to stress, hormones, and drugs, on the development of autoantibody-negative RA in genetically susceptible patients (Table 1). However, established non-genetic risk factors only provide an explanation for risk of RA in a minor part, suggesting that many (familial) risk factors remain to be identified.

3.1. Smoking, Other Pulmonary Irritants, and Respiratory Diseases

Numerous studies have established a clear relationship between smoking and the occurrence of autoantibody-positive RA [32,33]. The risk is significantly higher among RF-positive cases irrespective of the genetic background, whilst the association between smoking and ACPA appears most strongly related to HLA-DRB1 alleles [34,35,36]. Accordingly, it has been reported that smoking increases RA susceptibility in individuals carrying the HLA-DRB1 shared epitope regardless of the autoantibody status [37]. However, outside of genetic studies, the association between smoking and autoantibody-negative RA appears either absent [32] or low [33]. Indirectly supporting the non-causative role of smoking on autoantibody-negative RA, recent studies have indicated a temporal trend for increased incidence of RF-negative RA along with reduced smoking habits [38].
In contrast to cigarette smoking, other pulmonary irritants like air pollution [39,40], organic dusts [41], asbestos [42,43], and silica [44] have been observed to be associated with increased risk not only of autoantibody-positive but also of autoantibody-negative RA, with only silica showing a strong association with ACPA-positive RA alone. In keeping with the aforementioned studies on the different gene–environment interactions in RF-positive and ACPA-positive patients [34,35,36], these results would suggest that pulmonary inflammation could favor RA development not only through the formation of neo-antigens, but also via a number of other mechanisms, such as cytokine production, T cell polarization, epigenetic changes, modulation of the local microbiome, and others [45].
A recent epidemiological study in the Swedish Epidemiological Investigation of Rheumatoid Arthritis (EIRA) cohort helps clarifying the specific relationships between pulmonary inflammation, respiratory irritants such as smoking, and serostatus [46]. Pre-existing respiratory diseases were associated with both autoantibody-positive and -negative incident RA, with only small differences between the two. Acute lower airway exposure indeed interacted with smoking only in ACPA-positive subjects. In contrast, upper and lower chronic respiratory diseases, such as rhinitis, asthma, chronic obstructive pulmonary disease, and interstitial lung disease, were associated with risk of RA irrespective of RF and ACPA status, and more strongly in non-smokers. Based on these findings, it would appear that pulmonary inflammation is associated with RA through different, non mutually exclusive, and possibly synergic pathogenetic mechanisms. On the one side, smoking would predominantly drive autoantibody production, amplified in the context of a predisposing genetic background. Other pulmonary irritants and respiratory diseases would instead promote more generalized immune activation favoring the development of both autoantibody-positive and -negative RA.

3.2. Microbial Dysbiosis and Mucosal Inflammation

Accumulating evidence from epidemiological and translational studies indicates that alterations in the composition of the microbial flora at mucosal sites may contribute to the development and perpetuation of many immune-mediated inflammatory diseases [47]. The presence of dysbiosis has been demonstrated in the oral cavity, lungs, and gut of patients with RA, and is thought to have effects on inflammation and activation of autoreactive T and B cells at multiple time-points [48]. Of particular relevance to disease pathogenesis, specific bacteria have been associated with citrullinated protein production [49], and perturbations of the mucosal microbiome are already seen in ACPA-positive individuals at risk of developing RA [50,51]. At present, no specific studies have addressed the possible impact of dysbiosis in autoantibody-negative RA. However, interactions of the microbiota with the human immune system occur at multiple levels beyond autoreactive B cell activation, with effects on toll-like receptor (TLR) signaling, neutrophil extracellular traps formation, expansion of mucosa-derived innate immune cells, and polarization of T cells [47,52]. Supporting a role of microbial dysbiosis also in autoantibody-negative RA, mice deficient in interleukin (IL)-1 receptor antagonist show reduced intestinal microbial diversity and develop spontaneous arthritis through TLR activation and T helper 17 induction irrespective of autoantibodies [53]. Furthermore, respiratory diseases associated with chronic bacterial infection have been linked to RA development irrespective of RF and ACPA [46], and treatment-induced modifications of the circulating microbiome have recently been reported to differ between autoantibody-positive and -negative patients [54].

3.3. Body Weight and Diet

An association between excess body weight measured by body mass index (BMI) and RA has been suggested in many observational studies with conflicting results, especially in subgroups of different sex, age, or serological status [55,56,57,58,59]. In a small case-control study of 515 RA cases, Pedersen M [55] first reported that the association of being overweight or obese was confined to ACPA-negative RA. This observation was later confirmed by other independent large cohort studies. In the European Prospective Investigation of Cancer, Norfolk (EPIC-Norfolk) study, a BMI ≥30 was associated with a nearly threefold increase in the risk of developing autoantibody-negative inflammatory polyarthritis, with no significant effects on autoantibody-positive cases (HR 1.05, 95%CI 0.61 to 1.79, age and gender adjusted) [56]. A statistically significant association between obesity and risk of ACPA-negative RA was also found in the EIRA cohort but only among women (OR 1.6, 95%CI 1.0–3.3) [57]. These data are in line with a dose–response meta-analysis, in which an increase in RA risk was observed in overweight and obese women (RR 1.11, 95%CI 1.00–1.23, and RR 1.26, 95%CI 1.12–1.40, respectively) and in obese autoantibody-negative subjects (RR 1.47, 95%CI 1.11–1.96) [58]. In these sub-populations, the likelihood of developing RA increased linearly with the increase of BMI. However, differences related to the serological status were not confirmed in two large prospective cohorts, the Nurses’ Health Study (NHS) and Nurses’ Health Study II (NHSII), where the association between being overweight and obese and RA was borderline significant in both autoantibody-negative and -positive subjects (HR 1.37, 95%CI 0.95–1.98, and HR 1.37, 95%CI 0.91–2.09) [59]. Interestingly, risk was stronger and statistically significant among women diagnosed at age 55 years or younger. Although BMI is a poor measure of adiposity, studies on body composition assessed through whole-body dual energy X-ray absorptiometry have shown lower lean mass and higher fat mass in RA patients both in early and established disease [60,61]. Overall, the association between excess body weight and RA is not surprising, given the proinflammatory role of metabolic factors produced by adipose tissue and adipocytes [62]. The selective risk reported for autoantibody-negative RA should instead be interpreted with caution, as pain and inflammatory markers may be increased in obese subjects irrespective of swollen joints [63], with possible misclassification of a proportion of non-RA subjects.
In addition to body weight, many food components and beverages have been investigated in relation to RA risk, with inconsistent results [64,65]. Larger evidence exists for omega-3 fatty acids, which have been associated with lower risk of developing ACPA [66] and lower odds of developing inflammatory arthritis in ACPA-positive healthy subjects [67]. Despite such promising data, however, individual food and nutrients are unlikely to confer strong effects compared with the overall diet. Accordingly, in the NHS and NHSII cohorts, despite the lack of significant relationships between intake of omega-3 fatty acids and incident RA, healthy diet calculated according to the Alternative Healthy Eating Index (AHEI) score was shown to be protective for autoantibody-positive RA in women ≤55 years of age [68]. Among the individual components of the AHEI score, lower red meat intake and moderate alcohol consumption were found to be mostly associated with decreased early onset RA risk. Interestingly, the protective dose-dependent effect of alcohol on risk for RA has been confirmed by several studies [64,65] and is similar for ACPA-positive and -negative RA [69].

3.4. Hormonal Factors

Adult women experience three phases of endogenous hormonal shifts in life: pregnancy, postpartum, and menopause. It has been observed that the incidence of RA is lower during pregnancy [70,71], which might be explained by the complex mechanisms of immune tolerance and activation of immunoregulatory pathways [72]. Interestingly, although RA overall ameliorates in the course of pregnancy [73], pregnancy-related improvements of the disease are more evident in ACPA-negative RA [74]. Three to 24 months after delivery, the risk of RA seems to be increased [70,75], possibly due to the drastic fall in hormonal levels and increased prolactin during breastfeeding. Although detailed stratification based on the autoantibody serotype is not available, one study has suggested that the increased postpartum risk might be restricted to ACPA-negative RA [76]. The relation between parity and risk of incident RA is more difficult to establish, with recent meta-analyses providing conflicting results [77,78]. Previous pregnancy and childbirth appear overall irrelevant [78] or only slightly protective, with a J-shaped association between the number of pregnancies and RA risk [77]. However, the effect of parity might vary according to the patient’s HLA genotype. In particular, it has been reported that the protective effect of previous pregnancies is in fact restricted to RA women who possess the shared epitope [79]. Although these data need to be confirmed, it would thus appear that the positive impact of parity, if any, is more related to autoantibody-positive RA.
The third hormonal shift is during menopause, when estrogen levels decrease. In the large NHS and NHSII prospective cohorts, menopausal factors were only marginally associated with the risk of autoantibody-positive RA, whilst postmenopausal women had more than a doubled risk of autoantibody-negative disease compared with premenopausal women [80]. These results are in agreement with another smaller case-control study showing an association between early menopause and increased risk of RF-negative RA [81]. However, in the NHS and NHSII studies, the peak risk of developing autoantibody-negative RA was mostly observed after the menopausal transition [80], suggesting the contribution of additional yet unidentified factors acting together with hormonal shifts. Accordingly, the association between menopause and different RA serotypes remains controversial, with some studies rather reporting an increased risk of RF-positive RA in association with early age at menopause [82]. Ovarian failure has indeed been suggested to impact on RA-specific autoimmunity. In a female population of first-degree relatives of patients with RA, ACPA positivity was associated with menopause [83]; mechanistically, a decrease in estrogens may create a proinflammatory state characterized by low antibody sialylation as a consequence of downregulation of the enzyme β-galactoside α2,6-sialyltransferases 1 in plasmablasts [84].

3.5. Mental Health, Post-Traumatic Stress Disorders, and Depression

In longitudinal studies, depression has been associated with future risk of several chronic inflammatory diseases, including psoriasis and psoriatic arthritis [85,86] and inflammatory bowel diseases [87]. Two retrospective cohort studies suggested that depression may increase the risk for developing RA [88,89]. However, conclusions were limited due to possible confounding from lifestyle factors as well as possible reverse causation bias where early RA symptoms may have worsened mood prior to clinical arthritis. A recent and large nationwide longitudinal study of nearly 200,000 women with up to 22 years of follow-up found that indicators of depression (measured as a composite of self-reported clinician-diagnosed depression, regular antidepressant use, or a 5-question Mental Health Inventory score of <60) were associated with increased risk of developing incident RA [90]. Of note, the composite measure of depression was specifically associated with 63% increased risk for autoantibody-negative RA irrespective of measured factors including smoking pack-years, BMI, dietary intake, menopausal status, or physical activity. This study adds to the growing literature implicating a possible connection between RA risk and mental health in general. In the NHSII, an increasing number of symptoms of post-traumatic stress disorder (PTSD) increased RA risk [91]. Similarly, among military members, PTSD was associated with a 58% increased risk for any incident autoimmune disease, the most common being RA [92]. A significant association between PTSD and increased risk for incident RA has also been found among individuals exposed to the terrorist attack on the World Trade Center [93]. Mechanistically, patients with depression as well as individuals with post-traumatic stress disorder have increased levels of systemic inflammatory markers, such as IL-6 and C-reactive protein (CRP), compared to healthy controls, which could trigger inflammatory arthritis in predisposed subjects [94]. The reasons why such proinflammatory milieu may specifically favor the development of autoantibody-negative RA remain, however, unexplained.

3.6. Drugs

Studies focusing on immune-related adverse events (irAEs) of patients with cancer treated with immune checkpoint inhibitors (ICIs) may provide a good opportunity to unravel the characteristics and underlying immunological mechanisms of early stage autoantibody-negative RA. Immune checkpoint inhibitors target co-inhibitory pathways that normally function to downregulate T cell activation, such as cytotoxic T-lymphocyte antigen-4 CTLA-4, programmed death 1 (PD-1), or its ligand PD-L1. By blocking these co-inhibitory pathways, ICIs promote T cell-mediated antitumor immunity but may lead to a break in self-tolerance [95], manifesting as systemic or organ-specific autoimmunity. Inflammatory arthritis (IA) is the most common rheumatic irAE and has the potential to persist even after ICI cessation. IA-irAE is characteristically polyarticular and autoantibody negative [96]. A retrospective, cross-sectional comparative study on 20 cancer patients with de novo IA of the peripheral joints after the initiation of ICI therapy (ipilimumab, nivolumab, pembrolizumab, or combination) found that IA-irAE resembled autoantibody-negative RA in certain immunological characteristics, including negativity for RF and ACPA and a speckled pattern of anti-nuclear antibodies [97]. The low prevalence or very low titers of RF and ACPA in patients with IA-irAE has also been reported by others [98,99,100]. Based on these findings, one could hypothesize that IA-irAE is a disease process more likely dependent on autoreactive T cells rather than on B cell-based autoantibody production, and that dysregulation of PD-1 signaling may play an important role in autoantibody-negative RA.

4. Immunopathogenesis

4.1. Autoantibodies

The progressive improvement in autoantibody laboratory assays, together with the recognition of other possible autoantibody specificities beyond RF and ACPA, have led some authors to hypothesize that at least a proportion of autoantibody-negative RA is in fact incorrectly denominated seronegative [9]. Apart from non-canonical ACPAs, which can be detected in as many as 16% of ACPA-negative patients by multiplex citrullinated peptide arrays [101], the occurrence of autoantibody responses to other post-translational modified proteins is a well-known phenomenon in RA (Table 1). This is the case, for instance, of autoantibodies against carbamylated proteins (anti-CarP), reported to be present in 10–15% of RF and ACPA double-negative patients [102,103]. In contrast with the well-established clinical and pathogenetic role of ACPA [104,105], the significance of anti-CarP in the context of ACPA negativity remains controversial [106,107,108]. Still, the strong and specific association of HLA-B*08 carrying aspartate at position 9 with anti-CarP but not ACPA [109] reinforces the concept that the spectrum of autoimmunity in RA is broader than that traditionally recognized. The discovery of additional reactivities outside the spectrum of anti-modified protein autoantibodies, such as those against peptidylarginine deiminases (PADs), adds a further layer of complexity to the understanding of the immunological nature of autoantibody-negative RA. Both anti-PAD4 and anti-PAD3 may indeed be found in a variable proportion of 3–19% of ACPA-negative patients, and appear associated with worst disease prognosis in terms of joint damage and extra-articular involvement [110]. More sophisticated techniques, such as high-density protein microarrays, have very recently identified a number of other possible autoantibody specifities in approximatively 35% of ACPA-negative patients, with specificity of >90% for RA [111]. Of these, the combination of anti-PTX3 and anti-DUSP11 was found to have optimal diagnostic performance regardless of ACPA status. Whilst the exact role of DUSP11 is poorly studied, PTX3 is an essential component of innate immunity [112], underscoring the possible differential contribution of different branches of the immune system in autoantibody-negative compared to -positive RA.

4.2. Cytokine Networks

Cytokine-mediated pathways are at the center of the immunopathogenesis of RA, as demonstrated by animal models of arthritis and, more importantly, as confirmed by the dramatic improvement of patients’ prognosis upon the introduction of anti-cytokine therapies [113]. The pleiotropy, redundancy, synergy, and antagonism of the cytokine system hamper the establishment of linear models of cytokine hierarchy across multiple stages or phenotypes of the disease. However, the partially different response to therapies among individual patients with the same formal diagnosis of RA indicates that a molecular taxonomy based on cytokine ‘hubs’ could better address possible pathophysiological differences within the disease [114]. The efficacy of tumor necrosis factor (TNF)-α inhibition irrespective of the autoantibody status [115] indicates that this cytokine likely represents a common effector pathway that acts downstream of many inflammatory processes. Still, the selective benefit of concomitant methotrexate only in patients who are autoantibody positive [116] raises the possibility that the sources and effector functions of TNF-α may differ between the two disease subtypes, with autoantibody-negative RA more dependent on myeloid production, and less in need of simultaneous inhibition of IL-6-mediated B cell activation [117,118]. In line with this concept, myeloid-driven synovial and systemic inflammation appear more susceptible to TNF-α rather than IL-6R antagonism [119], and even less responsive to B cell-depleting agents [120]. Equally important, in a proportion of patients with abrupt onset polyarticular synovitis and systemic symptoms, as often occurs in autoantibody-negative RA, the pathogenetic events might be more centered on activation of the inflammasome and hyperproduction of IL-1β [8,121]. Accordingly, similarly to systemic adult-onset Still’s disease, these patients may exhibit a response to IL-1 blockers [122], which in contrast have limited efficacy in classical autoimmune RA. Further complicating the puzzle of cytokine taxonomy in RA, response has also been noted with IL-17A inhibition [123], suggesting that some forms of (autoantibody-negative) RA may share similarities with psoriatic arthritis [8].

4.3. Synovial Pathology

Irrespective of the clinical diagnosis, the synovial membrane in the course of chronic inflammatory arthritis is characterized by a number of common histopathological changes, including neoangiogenesis, proliferation and activation of the tissue stroma, and infiltration of the sublining layer by cells of the innate and adaptive immune system [124,125,126,127]. Notwithstanding these general similarities, synovial inflammatory features greatly vary in terms of cellular composition and reciprocal spatial arrangements across different disease entities and within a same disease, possibly reflecting divergent pathogenetic pathways [128,129]. One of the best characterized immunopathological and molecular feature is the relative balance between infiltrating macrophages and lymphoid cells, which helps to distinguish among myeloid, lympho-myeloid, and pauci-immune synovial pathotypes [130]. Relevantly, the lympho-myeloid pathotype is enriched in B cells and in genes of the adaptive immune response both locally [130,131] and systemically [119,132,133], and associates with a poorer response to non-targeted treatments [119,120,132,134] and more severe radiographic damage [130,131,133,134,135,136].
Despite B cell lineage synovial and systemic signatures predominating in patients with positive autoantibodies, particularly ACPA [130,131,132,133,135], autoantibody-negative RA can still display features indicative of adaptive immune responses at the site of inflammation in a variable but significant proportion of the cases [130,131,133,137] (Table 1) and, at least in long-standing disease, B cell-rich synovitis can be detected in as many as 45% of patients, without significant differences compared with autoantibody-positive RA [135]. Importantly, this finding has also been confirmed at the single-cell level, where different B cell subpopulations including memory and plasma cells appear mostly comparable irrespective of the autoantibody status [138,139]. It is therefore possible that, although the initial pathogenetic pathways of autoantibody-positive and -negative RA may differ, the two disease subsets eventually convey on shared inflammatory mechanisms. Confirming this notion, psoriatic arthritis, which is traditionally recognized as a non-autoimmune condition, may show some aspects of B cell activation in the synovium [140], and does not significantly differ from autoantibody-negative RA in the extent of B cell infiltration [141]. Single-cell sequencing of synovial immune cells has, however, very recently revealed key differences in local gene expression, with lower antigen processing and presentation activity and lower cytotoxicity and exhaustion in B and T lymphocytes but increased pro-inflammatory activity in macrophages from ACPA-negative compared to ACPA-positive RA [139] (Table 1). If confirmed, such pathogenetic diversity could help to explain the different response to targeted drugs with various mechanisms of action [104] and fuel precision therapy based on the autoantibody status.

4.4. Extra-Articular Involvement

Patients with RA suffer from multiple comorbidities, including cardiovascular disease (CVD), interstitial lung disease, and accelerated systemic bone loss [142]. Although chronic inflammation represents the major pathogenetic process of RA extra-articular involvements [143], the higher comorbid burden recognized in autoantibody-positive patients since the very beginning of the disease [144,145,146] and also beyond inflammation [2,147,148] underlines the importance of additional factors, including adaptive immune mechanisms and autoantibodies. However, at least in the case of CVD, incident cardiovascular events may occur early during the course of the disease also in autoantibody-negative RA [149]. Whether such an association generally arises from traditional risk factors or might be more specific of proinflammatory cytokines and inflammasome-driven pathways [150] remains to be established. Of particular relevance to autoantibody-negative RA, depression not only increases the risk of disease development, but also complicates its course in a significant proportion of the patients [151]. From a pathophysiologic perspective, TNF-driven arthritis, which is a model of myeloid cell-mediated disease, is associated with activation of microglia and neuroinflammation of certain brain regions, a pattern that is not seen in lymphoid cell-based arthritis [152]. A better understanding of the pathogenetic pathways underlying the development of different comorbidities in different subtypes of RA will certainly translate into more personalized medical approaches.

5. Conclusions

The mechanisms lying behind the phenotypic heterogeneity of RA remain largely unidentified, but possible genetic, environmental, and pathogenetic differences in relation to the autoantibody status are starting to emerge (Table 1). The timeline of disease development is remarkably less characterized in autoantibody-negative RA, but genetic susceptibility (mostly outside the HLA region) and environmental and endogenous triggers contribute to the development of an immune-inflammatory cascade that appears, at least in a proportion of the patients, largely driven by innate immune pathways and myeloid cytokines (Figure 1). A better definition of the pathogenic taxonomy of autoantibody-negative RA is needed to help improve the clinical management of this underlooked disease subtype, which remains challenging even in the modern treatment era.
The genetic susceptibility risk of autoantibody-negative rheumatoid arthritis mostly relies on class I HLA genes and a number of yet unidentified non-HLA loci. On the background of variable genetic predisposition, multiple exogeneous, endogenous, and stochastic factors contribute to the onset of the inflammatory cascade. These include, but are not limited to, inflammatory and microbial stimuli at mucosal sites, dietary factors and excess body weight, hormonal factors, mental health, and medications. In a proportion of the patients, the immunopathology of synovitis, at least in the initial stages, appears largely myeloid driven, with an abundant production of proinflammatory cytokines and only minor involvement of cells of the adaptive immune system. However, T and B cells can infiltrate the synovial tissue, and autoantibody production may also occur.

Author Contributions

L.D.S. and S.B. conceived the study, were involved in the literature search and drafted the manuscript. B.D. participated in the literature search and revised the manuscript critically. A.M. interpreted the literature critically and revised the manuscript for important intellectual content. C.M. conceived the study and revised the manuscript critically for important intellectual content. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported in part by funding from the IRCCS Policlinico San Matteo Foundation, Pavia, Italy.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

S.B. reports grant/research support from: Pfizer, and personal fees from: AbbVie, Bristol-Myers Squibb, Eli Lilly, Gilead, Pfizer, Sanofi. C.M. reports personal fees from: AbbVie, Bristol-Myers Squibb, Eli Lilly, Gilead, Pfizer, Roche, Sanofi.

References

  1. Van der Helm-van Mil, A.H.; Verpoort, K.N.; Breedveld, F.C.; Toes, R.E.; Huizinga, T.W. Antibodies to citrullinated proteins and differences in clinical progression of rheumatoid arthritis. Arthritis Res. Ther. 2005, 7, R949–R958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Westerlind, H.; Rönnelid, J.; Hansson, M.; Alfredsson, L.; Mathsson-Alm, L.; Serre, G.; Cornillet, M.; Holmdahl, R.; Jakobsson, P.; Skriner, K.; et al. Anti–Citrullinated Protein Antibody Specificities, Rheumatoid Factor Isotypes, and Incident Cardiovascular Events in Patients with Rheumatoid Arthritis. Arthritis Rheumatol. 2020, 72, 1658–1667. [Google Scholar] [CrossRef]
  3. Matthijssen, X.M.E.; Niemantsverdriet, E.; Huizinga, T.W.J.; van der Helm-van Mil, A.H.M. Enhanced treatment strategies and distinct disease outcomes among autoantibody-positive and -negative rheumatoid arthritis patients over 25 years: A longitudinal cohort study in the Netherlands. PLoS Med. 2020, 17, e1003296. [Google Scholar] [CrossRef]
  4. Boeters, D.M.; Gaujoux-Viala, C.; Constantin, A.; van der Helm-van Mil, A.H. The 2010 ACR/EULAR criteria are not sufficiently accurate in the early identification of autoantibody-negative rheumatoid arthritis: Results from the Leiden-EAC and ESPOIR cohorts. Semin. Arthritis Rheum. 2017, 47, 170–174. [Google Scholar] [CrossRef] [PubMed]
  5. Courvoisier, D.S.; Chatzidionysiou, K.; Mongin, D.; Lauper, K.; Mariette, X.; Morel, J.; Gottenberg, J.-E.; Bergstra, S.A.; Suarez, M.P.; Codreanu, C.; et al. The impact of seropositivity on the effectiveness of biologic anti-rheumatic agents: Results from a collaboration of 16 registries. Rheumatology 2020, 60, 820–828. [Google Scholar] [CrossRef] [PubMed]
  6. Malmström, V.; Catrina, A.I.; Klareskog, L. The immunopathogenesis of seropositive rheumatoid arthritis: From triggering to targeting. Nat. Rev. Immunol. 2017, 17, 60–75. [Google Scholar] [CrossRef]
  7. Scherer, H.U.; Häupl, T.; Burmester, G.R. The etiology of rheumatoid arthritis. J. Autoimmun. 2020, 110, 102400. [Google Scholar] [CrossRef]
  8. McGonagle, D.; Watad, A.; Savic, S. Mechanistic immunological based classification of rheumatoid arthritis. Autoimmun. Rev. 2018, 17, 1115–1123. [Google Scholar] [CrossRef]
  9. Pouw, J.; Leijten, E.; Van Laar, J.; Boes, M. Revisiting B cell tolerance and autoantibodies in seropositive and seronegative autoimmune rheumatic disease (AIRD). Clin. Exp. Immunol. 2021, 203, 160–173. [Google Scholar] [CrossRef]
  10. Van der Woude, D.; Houwing-Duistermaat, J.J.; Toes, R.E.M.; Huizinga, T.W.J.; Thomson, W.; Worthinton, J.; van der Helm-van Mil, A.H.M.; de Vries, R.R.P. Quantitative heritability of anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis. Arthritis Rheum. 2009, 60, 916–923. [Google Scholar] [CrossRef]
  11. Hensvold, A.H.; Magnusson, P.; Joshua, V.; Hansson, M.; Israelsson, L.; Ferreira, R.C.; Jakobsson, P.-J.; Holmdahl, R.; Hammarström, L.; Malmström, V.; et al. Environmental and genetic factors in the development of anticitrullinated protein antibodies (ACPAs) and ACPA-positive rheumatoid arthritis: An epidemiological investigation in twins. Ann. Rheum. Dis. 2015, 74, 375–380. [Google Scholar] [CrossRef] [PubMed]
  12. Frisell, T.; Holmqvist, M.; Källberg, H.; Klareskog, L.; Alfredsson, L.; Askling, J. Familial Risks and Heritability of Rheumatoid Arthritis: Role of Rheumatoid Factor/Anti-Citrullinated Protein Antibody Status, Number and Type of Affected Relatives, Sex, and Age. Arthritis Rheum. 2013, 65, 2773–2782. [Google Scholar] [CrossRef] [PubMed]
  13. Frisell, T.; Hellgren, K.; Alfredsson, L.; Raychaudhuri, S.; Klareskog, L.; Askling, J. Familial aggregation of arthritis-related diseases in seropositive and seronegative rheumatoid arthritis: A register-based case-control study in Sweden. Ann. Rheum. Dis. 2016, 75, 183–189. [Google Scholar] [CrossRef] [Green Version]
  14. Kirino, Y.; Remmers, E.F. Genetic architectures of seropositive and seronegative rheumatic diseases. Nat. Rev. Rheumatol. 2015, 11, 401–414. [Google Scholar] [CrossRef]
  15. Padyukov, L.; Seielstad, M.; Ong, R.T.H.; Ding, B.; Ronnelid, J.; Seddighzadeh, M.; Alfredsson, L.; Klareskog, L.; Epidemiological Investigation of Rheumatoid Arthritis (EIRA) Study Group. A genome-wide association study suggests contrasting associations in ACPA-positive versus ACPA-negative rheumatoid arthritis. Ann. Rheum. Dis. 2011, 70, 259–265. [Google Scholar] [CrossRef]
  16. Eyre, S.; Bowes, J.; Diogo, D.; Lee, A.; Barton, A.; Martin, P.; Zhernakova, A.; Stahl, E.; Viatte, S.; McAllister, K.; et al. High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis. Nat. Genet. 2012, 44, 1336–1440. [Google Scholar] [CrossRef] [PubMed]
  17. Han, B.; Diogo, D.; Eyre, S.; Kallberg, H.; Zhernakova, A.; Bowes, J.; Padyukov, L.; Okada, Y.; González-Gay, M.A.; Dahlqvist, S.R.; et al. Fine Mapping Seronegative and Seropositive Rheumatoid Arthritis to Shared and Distinct HLA Alleles by Adjusting for the Effects of Heterogeneity. Am. J. Hum. Genet. 2014, 94, 522–532. [Google Scholar] [CrossRef] [Green Version]
  18. Bossini-Castillo, L.; de Kovel, C.; Kallberg, H.; van ‘t Slot, R.; Italiaander, A.; Coenen, M.; Tak, P.P.; Posthumus, M.D.; Wijmenga, C.; Huizinga, T.; et al. A genome-wide association study of rheumatoid arthritis without antibodies against citrullinated peptides. Ann. Rheum. Dis. 2015, 74, e15. [Google Scholar] [CrossRef] [Green Version]
  19. Terao, C.; Brynedal, B.; Chen, Z.; Jiang, X.; Westerlind, H.; Hansson, M.; Jakobsson, P.-J.; Lundberg, K.; Skriner, K.; Serre, G.; et al. Distinct HLA associations with rheumatoid arthritis subsets defined by serological subphenotype. Am. J. Hum. Genet. 2019, 105, 616–624. [Google Scholar] [CrossRef]
  20. Irigoyen, P.; Lee, A.T.; Wener, M.H.; Li, W.; Kern, M.; Batliwalla, F.; Lum, R.F.; Massarotti, E.; Weisman, M.; Bombardier, C.; et al. Regulation of anti-cyclic citrullinated peptide antibodies in rheumatoid arthritis: Contrasting effects of HLA-DR3 and the shared epitope alleles. Arthritis Rheum. 2005, 52, 3813–3818. [Google Scholar] [CrossRef]
  21. Verpoort, K.N.; van Gaalen, F.A.; van der Helm-van Mil, A.H.; Schreuder, G.M.T.; Breedveld, F.C.; Huizinga, T.W.J.; de Vries, R.R.P.; Toes, R.E.M. Association of HLA-DR3 with anti-cyclic citrullinated peptide antibody-negative rheumatoid arthritis. Arthritis Rheum. 2005, 52, 3058–3062. [Google Scholar] [CrossRef] [PubMed]
  22. Petrelli, A.; van Wijk, F. CD8(+) T cells in human autoimmune arthritis: The unusual suspects. Nat. Rev. Rheumatol. 2016, 12, 421–428. [Google Scholar] [CrossRef] [PubMed]
  23. Kelkka, T.; Savola, P.; Bhattacharya, D.; Huuhtanen, J.; Lönnberg, T.; Kankainen, M.; Paalanen, K.; Tyster, M.; Lepistö, M.; Ellonen, P.; et al. Adult-Onset Anti-Citrullinated Peptide Antibody-Negative Destructive Rheumatoid Arthritis Is Characterized by a Disease-Specific CD8+ T Lymphocyte Signature. Front. Immunol. 2020, 11, 578848. [Google Scholar] [CrossRef] [PubMed]
  24. Scally, S.W.; Petersen, J.; Law, S.C.; Dudek, N.L.; Nel, H.J.; Loh, K.L.; Wijeyewickrema, L.; Eckle, S.; Van Heemst, J.; Pike, R.; et al. A molecular basis for the association of the HLA-DRB1 locus, citrullination, and rheumatoid arthritis. J. Exp. Med. 2013, 210, 2569–2582. [Google Scholar] [CrossRef] [PubMed]
  25. Hinks, A.; Bowes, J.; Cobb, J.; Ainsworth, H.C.; Marion, M.C.; Comeau, M.; Sudman, M.; Han, B.; Becker, M.L.; Bohnsack, J.F.; et al. Fine-mapping the MHC locus in juvenile idiopathic arthritis (JIA) reveals genetic heterogeneity corresponding to distinct adult inflammatory arthritic diseases. Ann. Rheum. Dis. 2017, 76, 765–772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Viatte, S.; Plant, D.; Bowes, J.; Lunt, M.; Eyre, S.; Barton, A.; Worthington, J. Genetic markers of rheumatoid arthritis susceptibility in anti-citrullinated peptide antibody negative patients. Ann. Rheum. Dis. 2012, 71, 1984–1990. [Google Scholar] [CrossRef] [Green Version]
  27. Okada, Y.; Wu, D.; Trynka, G.; Raj, T.; Terao, C.; Ikari, K.; Kochi, Y.; Ohmura, K.; Suzuki, A.; Yoshida, S.; et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 2014, 506, 376–381. [Google Scholar] [CrossRef]
  28. Terao, C.; Ohmura, K.; Kochi, Y.; Ikari, K.; Okada, Y.; Shimizu, M.; Nishina, N.; Suzuki, A.; Myouzen, K.; Kawaguchi, T.; et al. Anti-citrullinated peptide/protein antibody (ACPA)-negative RA shares a large proportion of susceptibility loci with ACPA-positive RA: A meta-analysis of genome-wide association study in a Japanese population. Arthritis Res. Ther. 2015, 17, 104. [Google Scholar] [CrossRef] [Green Version]
  29. Viatte, S.; Massey, J.; Bowes, J.; Duffus, K.; Eyre, S.; Barton, A.; Worthington, J. Replication of Associations of Genetic Loci Outside the HLA Region with Susceptibility to Anti–Cyclic Citrullinated Peptide–Negative Rheumatoid Arthritis. Arthritis Rheumatol. 2016, 68, 1603–1613. [Google Scholar] [CrossRef] [PubMed]
  30. Ugidos, N.; Mena, J.; Baquero, S.; Alloza, I.; Azkargorta, M.; Elortza, F.; Vandenbroeck, K. Interactome of the Autoimmune Risk Protein ANKRD55. Front. Immunol. 2019, 10, 2067. [Google Scholar] [CrossRef]
  31. Wei, W.-H.; Viatte, S.; Merriman, T.R.; Barton, A.; Worthington, J. Genotypic variability based association identifies novel non-additive loci DHCR7 and IRF4 in sero-negative rheumatoid arthritis. Sci. Rep. 2017, 7, 5261. [Google Scholar] [CrossRef] [Green Version]
  32. Sugiyama, D.; Nishimura, K.; Tamaki, K.; Tsuji, G.; Nakazawa, T.; Morinobu, A.; Kumagai, S. Impact of smoking as a risk factor for developing rheumatoid arthritis: A meta-analysis of observational studies. Ann. Rheum. Dis. 2010, 69, 70–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Di Giuseppe, D.; Discacciati, A.; Orsini, N.; Wolk, A. Cigarette smoking and risk of rheumatoid arthritis: A dose-response meta-analysis. Arthritis Res. Ther. 2014, 16, R61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hedström, A.K.; Rönnelid, J.; Klareskog, L.; Alfredsson, L. Complex Relationships of Smoking, HLA-DRB1 Genes, and Serologic Profiles in Patients with Early Rheumatoid Arthritis: Update from a Swedish Population-Based Case-Control Study. Arthritis Rheumatol. 2019, 71, 1504–1511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Ishikawa, Y.; Ikari, K.; Hashimoto, M.; Ohmura, K.; Tanaka, M.; Ito, H.; Taniguchi, A.; Yamanaka, H.; Mimori, T.; Terao, C. Shared epitope defines distinct associations of cigarette smoking with levels of anticitrullinated protein antibody and rheumatoid factor. Ann. Rheum. Dis. 2019, 78, 1480–1487. [Google Scholar] [CrossRef] [PubMed]
  36. Regueiro, C.; Rodriguez-Rodriguez, L.; Lopez-Mejias, R.; Nuño, L.; Triguero-Martinez, A.; Perez-Pampin, E.; Corrales, A.; Villalba, A.; Lopez-Golan, Y.; Abasolo, L.; et al. A predominant involvement of the triple seropositive patients and others with rheumatoid factor in the association of smoking with rheumatoid arthritis. Sci. Rep. 2020, 10, 3355. [Google Scholar] [CrossRef] [Green Version]
  37. Bang, S.-Y.; Lee, K.-H.; Cho, S.-K.; Lee, H.-S.; Lee, K.W.; Bae, S.-C. Smoking increases rheumatoid arthritis susceptibility in individuals carrying the HLA-DRB1 shared epitope, regardless of rheumatoid factor or anti-cyclic citrullinated peptide antibody status. Arthritis Rheum. 2010, 62, 369–377. [Google Scholar] [CrossRef]
  38. Myasoedova, E.; Davis, J.; Matteson, E.L.; Crowson, C.S. Is the epidemiology of rheumatoid arthritis changing? Results from a population-based incidence study, 1985–2014. Ann. Rheum. Dis. 2020, 79, 440–444. [Google Scholar] [CrossRef]
  39. Hart, J.E.; Laden, F.; Puett, R.C.; Costenbader, K.H.; Karlson, E.W. Exposure to Traffic Pollution and Increased Risk of Rheumatoid Arthritis. Environ. Health Perspect. 2009, 117, 1065–1069. [Google Scholar] [CrossRef] [Green Version]
  40. De Roos, A.J.; Koehoorn, M.; Tamburic, L.; Davies, H.W.; Brauer, M. Proximity to traffic, ambient air pollution, and community noise in relation to incident rheumatoid arthritis. Environ. Health Perspect. 2014, 122, 1075–1080. [Google Scholar] [CrossRef] [Green Version]
  41. Ilar, A.; Gustavsson, P.; Wiebert, P.; Alfredsson, L. Occupational exposure to organic dusts and risk of developing rheumatoid arthritis: Findings from a Swedish population-based case–control study. RMD Open 2019, 5, e001049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Miller, F.W.; Alfredsson, L.; Costenbader, K.H.; Kamen, D.L.; Nelson, L.M.; Norris, J.M.; De Roos, A.J. Epidemiology of environmental exposures and human autoimmune diseases: Findings from a National Institute of Environmental Health Sciences Expert Panel Workshop. J. Autoimmun. 2012, 39, 259–271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ilar, A.; Klareskog, L.; Saevarsdottir, S.; Wiebert, P.; Askling, J.; Gustavsson, P.; Alfredsson, L. Occupational exposure to asbestos and silica and risk of developing rheumatoid arthritis: Findings from a Swedish population-based case-control study. RMD Open 2019, 5, e000978. [Google Scholar] [CrossRef] [PubMed]
  44. Zeng, P.; Chen, Z.; Klareskog, L.; Alfredsson, L.; Bengtsson, C.; Jiang, X. Amount of smoking, duration of smoking cessation and their interaction with silica exposure in the risk of rheumatoid arthritis among males: Results from the Swedish Epidemiological Investigation of Rheumatoid Arthritis (EIRA) study. Ann. Rheum. Dis. 2018, 77, 1238–1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Ishikawa, Y.; Terao, C. The Impact of Cigarette Smoking on Risk of Rheumatoid Arthritis: A Narrative Review. Cells 2020, 9, 475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kronzer, V.L.; Westerlind, H.; Alfredsson, L.; Crowson, C.S.; Nyberg, F.; Tornling, G.; Klareskog, L.; Holmqvist, M.; Askling, J. Respiratory Diseases as Risk Factors for Seropositive and Seronegative Rheumatoid Arthritis and in Relation to Smoking. Arthritis Rheumatol. 2021, 73, 61–68. [Google Scholar] [CrossRef] [PubMed]
  47. Honda, K.; Littman, D.R. The Microbiome in Infectious Disease and Inflammation. Annu. Rev. Immunol. 2012, 30, 759–795. [Google Scholar] [CrossRef] [Green Version]
  48. Wilson, T.M.; Trent, B.; Kuhn, K.A.; Demoruelle, M.K. Microbial Influences of Mucosal Immunity in Rheumatoid Arthritis. Curr. Rheumatol. Rep. 2020, 22, 83. [Google Scholar] [CrossRef]
  49. Gómez-Bañuelos, E.; Mukherjee, A.; Darrah, E.; Andrade, F. Rheumatoid Arthritis-Associated Mechanisms of Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans. J. Clin. Med. 2019, 8, 1309. [Google Scholar] [CrossRef] [Green Version]
  50. Cheng, Z.; Do, T.; Mankia, K.; Meade, J.; Hunt, L.; Clerehugh, V.; Speirs, A.; Tugnait, A.; Emery, P.; Devine, D. Dysbiosis in the oral microbiomes of anti-CCP positive individuals at risk of developing rheumatoid arthritis. Ann. Rheum. Dis. 2021, 80, 162–168. [Google Scholar] [CrossRef]
  51. Rooney, C.M.; Mankia, K.; Mitra, S.; Moura, I.B.; Emery, P.; Wilcox, M.H. Perturbations of the gut microbiome in anti-CCP positive individuals at risk of developing rheumatoid arthritis. Rheumatology 2021, 60, 3380–3387. [Google Scholar] [CrossRef]
  52. Zaiss, M.M.; Wu, H.-J.J.; Mauro, D.; Schett, G.; Ciccia, F. The gut–joint axis in rheumatoid arthritis. Nat. Rev. Rheumatol. 2021, 17, 224–237. [Google Scholar] [CrossRef]
  53. Rogier, R.; Ederveen, T.H.A.; Boekhorst, J.; Wopereis, H.; Scher, J.U.; Manasson, J.; Frambach, S.J.C.M.; Knol, J.; Garssen, J.; Van Der Kraan, P.M.; et al. Aberrant intestinal microbiota due to IL-1 receptor antagonist deficiency promotes IL-17- and TLR4-dependent arthritis. Microbiome 2017, 5, 63. [Google Scholar] [CrossRef]
  54. Hammad, D.B.M.; Hider, S.L.; Liyanapathirana, V.C.; Tonge, D.P. Molecular Characterization of Circulating Microbiome Signatures in Rheumatoid Arthritis. Front. Cell. Infect. Microbiol. 2020, 9, 440. [Google Scholar] [CrossRef]
  55. Pedersen, M.; Jacobsen, S.; Klarlund, M.; Pedersen, B.V.; Wiik, A.; Wohlfahrt, J.; Frisch, M. Environmental risk factors differ between rheumatoid arthritis with and without auto-antibodies against cyclic citrullinated peptides. Arthritis Res. Ther. 2006, 8, R133. [Google Scholar] [CrossRef] [Green Version]
  56. Lahiri, M.; Luben, R.; Morgan, C.; Bunn, D.K.; Marshall, T.; Lunt, M.; Verstappen, S.; Symmons, D.; Khaw, K.-T.; Wareham, N.; 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, 219–226. [Google Scholar] [CrossRef] [PubMed]
  57. Wesley, A.; Bengtsson, C.; Elkan, A.-C.; Klareskog, L.; Alfredsson, L.; Wedrén, S.; Epidemiological Investigation of Rheumatoid Arthritis Study Group. 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. 2013, 65, 107–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Feng, J.; Chen, Q.; Yu, F.; Wang, Z.; Chen, S.; Jin, Z.; Cai, Q.; Liu, Y.; He, J. Body Mass Index and Risk of Rheumatoid Arthritis: A Meta-Analysis of Observational Studies. Medicine 2016, 95, e2859. [Google Scholar] [CrossRef] [PubMed]
  59. Lu, B.; Hiraki, L.T.; Sparks, J.; Malspeis, S.; Chen, C.-Y.; Awosogba, J.A.; Arkema, E.; Costenbader, K.H.; Karlson, E.W. Being overweight or obese and risk of developing rheumatoid arthritis among women: A prospective cohort study. Ann. Rheum. Dis. 2014, 73, 1914–1922. [Google Scholar] [CrossRef] [PubMed]
  60. Turk, S.A.; Van Schaardenburg, D.; Boers, M.; De Boer, S.; Fokker, C.; Lems, W.F.; Nurmohamed, M.T. An unfavorable body composition is common in early arthritis patients: A case control study. PLoS ONE 2018, 13, e0193377. [Google Scholar] [CrossRef] [Green Version]
  61. Giles, J.T.; Ling, S.M.; Ferrucci, L.; Bartlett, S.; Andersen, R.E.; Towns, M.; Muller, D.; Fontaine, K.R.; Bathon, J.M. Abnormal body composition phenotypes in older rheumatoid arthritis patients: Association with disease characteristics and pharmacotherapies. Arthritis Rheum. 2008, 59, 807–815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Neumann, E.; Hasseli, R.; Ohl, S.; Lange, U.; Frommer, K.; Müller-Ladner, U. Adipokines and Autoimmunity in Inflammatory Arthritis. Cells 2021, 10, 216. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, Y.; Hazlewood, G.; Kaplan, G.G.; Eksteen, B.; Barnabe, C. Impact of Obesity on Remission and Disease Activity in Rheumatoid Arthritis: A Systematic Review and Meta-Analysis. Arthritis Care Res. 2017, 69, 157–165. [Google Scholar] [CrossRef] [PubMed]
  64. Klareskog, L.; Rönnelid, J.; Saevarsdottir, S.; Padyukov, L.; Alfredsson, L. The importance of differences; On environment and its interactions with genes and immunity in the causation of rheumatoid arthritis. J. Intern. Med. 2020, 287, 514–533. [Google Scholar] [CrossRef]
  65. Salliot, C.; Nguyen, Y.; Boutron-Ruault, M.-C.; Seror, R. Environment and Lifestyle: Their Influence on the Risk of RA. J. Clin. Med. 2020, 9, 3109. [Google Scholar] [CrossRef] [PubMed]
  66. Gan, R.W.; Demoruelle, M.K.; Deane, K.D.; Weisman, M.H.; Buckner, J.H.; Gregersen, P.K.; Mikuls, T.R.; O’Dell, J.R.; Keating, R.M.; Fingerlin, T.; et al. Omega-3 fatty acids are associated with a lower prevalence of autoantibodies in shared epitope-positive subjects at risk for rheumatoid arthritis. Ann. Rheum. Dis. 2017, 76, 147–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Gan, R.W.; Bemis, E.A.; Demoruelle, M.K.; Striebich, C.C.; Brake, S.; Feser, M.L.; Moss, L.; Clare-Salzler, M.; Holers, V.M.; Deane, K.D.; et al. The association between omega-3 fatty acid biomarkers and inflammatory arthritis in an anti-citrullinated protein antibody positive population. Rheumatology 2017, 56, 2229–2236. [Google Scholar] [CrossRef] [Green Version]
  68. Hu, Y.; Sparks, J.; Malspeis, S.; Costenbader, K.H.; Hu, F.B.; Karlson, E.W.; Lu, B. Long-term dietary quality and risk of developing rheumatoid arthritis in women. Ann. Rheum. Dis. 2017, 76, 1357–1364. [Google Scholar] [CrossRef]
  69. Källberg, H.; Jacobsen, S.; Bengtsson, C.; Pedersen, M.; Padyukov, L.; Garred, P.; Frisch, M.; Karlson, E.W.; Klareskog, L.; Alfredsson, L. Alcohol consumption is associated with decreased risk of rheumatoid arthritis: Results from two Scandinavian case–control studies. Ann. Rheum. Dis. 2009, 68, 222–227. [Google Scholar] [CrossRef]
  70. Silman, A.; Kay, A.; Brennan, P. Timing of Pregnancy in Relation to the Onset of Rheumatoid Arthritis. Arthritis Rheum. 1992, 35, 152–155. [Google Scholar] [CrossRef]
  71. Lansink, M.; De Boer, A.; Dijkmans, B.A.; Vandenbroucke, J.P.; Hazes, J.M. The onset of rheumatoid arthritis in relation to pregnancy and childbirth. Clin. Exp. Rheumatol. 1993, 11, 171–174. [Google Scholar]
  72. Förger, F.; Villiger, P.M. Immunological adaptations in pregnancy that modulate rheumatoid arthritis disease activity. Nat. Rev. Rheumatol. 2020, 16, 113–122. [Google Scholar] [CrossRef] [PubMed]
  73. Jethwa, H.; Lam, S.; Smith, C.; Giles, I. Does Rheumatoid Arthritis Really Improve During Pregnancy? A Systematic Review and Metaanalysis. J. Rheumatol. 2019, 46, 245–250. [Google Scholar] [CrossRef]
  74. de Man, Y.A.; Bakker-Jonges, L.E.; Goorbergh, C.M.; Tillemans, S.P.; Hooijkaas, H.; Hazes, J.M.; Dolhain, R.J.E.M. Women with rheumatoid arthritis negative for anti-cyclic citrullinated peptide and rheumatoid factor are more likely to improve during pregnancy, whereas in autoantibody-positive women autoantibody levels are not influenced by pregnancy. Ann. Rheum. Dis. 2010, 69, 420–423. [Google Scholar] [CrossRef] [PubMed]
  75. Wallenius, M.; Skomsvoll, J.F.; Irgens, L.M.; Salvesen, K.; Koldingsnes, W.; Mikkelsen, K.; Kaufmann, C.; Kvien, T.K. Postpartum onset of rheumatoid arthritis and other chronic arthritides: Results from a patient register linked to a medical birth registry. Ann. Rheum. Dis. 2010, 69, 332–336. [Google Scholar] [CrossRef]
  76. Orellana, C.; Wedrén, S.; Källberg, H.; Holmqvist, M.; Karlsson, E.W.; Alfredsson, L.; Bengtsson, C.; EIRA Study Group. Parity and the risk of developing rheumatoid arthritis: Results from the Swedish Epidemiological Investigation of Rheumatoid Arthritis study. Ann. Rheum. Dis. 2014, 73, 752–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Ren, L.; Guo, P.; Sun, Q.-M.; Liu, H.; Chen, Y.; Huang, Y.; Cai, X.-J. Number of parity and the risk of rheumatoid arthritis in women: A dose-response meta-analysis of observational studies. J. Obstet. Gynaecol. Res. 2017, 43, 1428–1440. [Google Scholar] [CrossRef] [Green Version]
  78. Chen, W.M.; Subesinghe, S.; Muller, S.; Hider, S.L.; Mallen, C.; Scott, I.C. The association between gravidity, parity and the risk of developing rheumatoid arthritis: A systematic review and meta-analysis. Semin. Arthritis Rheum. 2020, 50, 252–260. [Google Scholar] [CrossRef] [PubMed]
  79. Guthrie, K.A.; Gammill, H.S.; Madeleine, M.M.; Dugowson, C.E.; Nelson, J.L. Parity and HLA alleles in risk of rheumatoid arthritis. Chimerism 2011, 2, 11–15. [Google Scholar] [CrossRef]
  80. Bengtsson, C.; Malspeis, S.; Orellana, C.; Sparks, J.A.; Costenbader, K.H.; Karlson, E.W. Association Between Menopausal Factors and the Risk of Seronegative and Seropositive Rheumatoid Arthritis: Results from the Nurses’ Health Studies. Arthritis Care Res. 2017, 69, 1676–1684. [Google Scholar] [CrossRef] [Green Version]
  81. Pikwer, M.; Bergström, U.; Nilsson, J.; Jacobsson, L.; Turesson, C. Early menopause is an independent predictor of rheumatoid arthritis. Ann. Rheum. Dis. 2012, 71, 378–381. [Google Scholar] [CrossRef] [PubMed]
  82. Wong, L.E.; Huang, W.-T.; Pope, J.; Haraoui, B.; Boire, G.; Thorne, J.C.; Hitchon, C.A.; Tin, D.; Keystone, E.C.; Bykerk, V.P.; et al. Effect of Age at Menopause on Disease Presentation in Early Rheumatoid Arthritis: Results from the Canadian Early Arthritis Cohort. Arthritis Care Res. 2015, 67, 616–623. [Google Scholar] [CrossRef] [PubMed]
  83. Alpízar-Rodríguez, D.; Mueller, R.B.; Möller, B.; Dudler, J.; Ciurea, A.; Zufferey, P.; Kyburz, D.; Walker, U.A.; Von Mühlenen, I.; Roux-Lombard, P.; et al. Female hormonal factors and the development of anti-citrullinated protein antibodies in women at risk of rheumatoid arthritis. Rheumatology 2017, 56, 1579–1585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Engdahl, C.; Bondt, A.; Harre, U.; Raufer, J.; Pfeifle, R.; Camponeschi, A.; Wuhrer, M.; Seeling, M.; Mårtensson, I.-L.; Nimmerjahn, F.; et al. Estrogen induces St6gal1 expression and increases IgG sialylation in mice and patients with rheumatoid arthritis: A potential explanation for the increased risk of rheumatoid arthritis in postmenopausal women. Arthritis Res. Ther. 2018, 20, 84. [Google Scholar] [CrossRef] [Green Version]
  85. Dominguez, P.; Han, J.; Li, T.; Ascherio, A.; Qureshi, A. Depression and the risk of psoriasis in US women. J. Eur. Acad. Dermatol. Venereol. 2013, 27, 1163–1167. [Google Scholar] [CrossRef] [Green Version]
  86. Lewinson, R.T.; Vallerand, I.A.; Lowerison, M.W.; Parsons, L.M.; Frolkis, A.D.; Kaplan, G.G.; Bulloch, A.G.; Swain, M.G.; Patten, S.; Barnabe, C. Depression Is Associated with an Increased Risk of Psoriatic Arthritis among Patients with Psoriasis: A Population-Based Study. J. Investig. Dermatol. 2017, 137, 828–835. [Google Scholar] [CrossRef] [Green Version]
  87. Ananthakrishnan, A.N.; Khalili, H.; Pan, A.; Higuchi, L.M.; de Silva, P.; Richter, J.M.; Fuchs, C.S.; Chan, A.T. Association between depressive symptoms and incidence of Crohn’s disease and ulcerative colitis: Results from the Nurses’ Health Study. Clin. Gastroenterol. Hepatol. 2013, 11, 57–62. [Google Scholar] [CrossRef] [Green Version]
  88. Lu, M.-C.; Guo, H.-R.; Lin, M.-C.; Livneh, H.; Lai, N.-S.; Tsai, T.-Y. Bidirectional associations between rheumatoid arthritis and depression: A nationwide longitudinal study. Sci. Rep. 2016, 6, 20647. [Google Scholar] [CrossRef] [Green Version]
  89. Vallerand, I.; Lewinson, R.T.; Frolkis, A.D.; Lowerison, M.W.; Kaplan, G.G.; Swain, M.G.; Bulloch, A.G.M.; Patten, S.B.; Barnabe, C. Depression as a risk factor for the development of rheumatoid arthritis: A population-based cohort study. RMD Open 2018, 4, e000670. [Google Scholar] [CrossRef] [Green Version]
  90. Sparks, J.A.; Malspeis, S.; Hahn, J.; Wang, J.; Roberts, A.L.; Kubzansky, L.D.; Costenbader, K.H. Depression and Subsequent Risk for Incident Rheumatoid Arthritis Among Women. Arthritis Care Res. 2021, 73, 78–89. [Google Scholar] [CrossRef]
  91. Lee, Y.C.; Agnew-Blais, J.; Malspeis, S.; Keyes, K.M.; Costenbader, K.H.; Kubzansky, L.D.; Roberts, A.L.; Koenen, K.C.; Karlson, E.W. Post-Traumatic Stress Disorder and Risk for Incident Rheumatoid Arthritis. Arthritis Care Res. 2016, 68, 292–298. [Google Scholar] [CrossRef] [Green Version]
  92. Bookwalter, D.B.; Roenfeldt, K.A.; LeardMann, C.A.; Kong, S.Y.; Riddle, M.S.; Rull, R. Posttraumatic stress disorder and risk of selected autoimmune diseases among US military personnel. BMC Psychiatry 2020, 20, 23. [Google Scholar] [CrossRef]
  93. Miller-Archie, S.A.; Izmirly, P.M.; Berman, J.R.; Brite, J.; Walker, D.J.; Dasilva, R.C.; Petrsoric, L.J.; Cone, J.E. Systemic autoimmune disease among adults exposed to the September 11, 2001, terrorist attack. Arthritis Rheumatol. 2020, 72, 849–859. [Google Scholar] [CrossRef] [Green Version]
  94. Nerurkar, L.; Siebert, S.; McInnes, I.B.; Cavanagh, J. Rheumatoid arthritis and depression: An inflammatory perspective. Lancet Psychiatry 2019, 6, 164–173. [Google Scholar] [CrossRef]
  95. Michot, J.; Bigenwald, C.; Champiat, S.; Collins, M.; Carbonnel, F.; Postel-Vinay, S.; Berdelou, A.; Varga, A.; Bahleda, R.; Hollebecque, A.; et al. Immune-related adverse events with immune checkpoint blockade: A comprehensive review. Eur. J. Cancer 2016, 54, 139–148. [Google Scholar] [CrossRef] [PubMed]
  96. Calabrese, L.H.; Calabrese, C.; Cappelli, L.C. Rheumatic immune-related adverse events from cancer immunotherapy. Nat. Rev. Rheumatol. 2018, 14, 569–579. [Google Scholar] [CrossRef] [PubMed]
  97. Liu, Y.; Jaquith, J.M.; McCarthy-Fruin, K.; Zhu, X.; Zhou, X.; Li, Y.; Crowson, C.; Davis, J.M.; Thanarajasingam, U.; Zeng, H. Immune checkpoint inhibitor-induced inflammatory arthritis: A novel clinical entity with striking similarities to seronegative rheumatoid arthritis. Clin. Rheumatol. 2020, 39, 3631–3637. [Google Scholar] [CrossRef]
  98. Richter, M.D.; Crowson, C.; Kottschade, L.A.; Finnes, H.D.; Markovic, S.N.; Thanarajasingam, U. Rheumatic Syndromes Associated with Immune Checkpoint Inhibitors: A Single-Center Cohort of Sixty-One Patients. Arthritis Rheumatol. 2019, 71, 468–475. [Google Scholar] [CrossRef] [Green Version]
  99. Braaten, T.J.; Brahmer, J.R.; Forde, P.M.; Le, D.; Lipson, E.J.; Naidoo, J.; Schollenberger, M.; Zheng, L.; Bingham, C.O.; Shah, A.A.; et al. Immune checkpoint inhibitor-induced inflammatory arthritis persists after immunotherapy cessation. Ann. Rheum. Dis. 2020, 79, 332–338. [Google Scholar] [CrossRef]
  100. Cappelli, L.C.; Brahmer, J.R.; Forde, P.M.; Le, D.T.; Lipson, E.J.; Naidoo, J.; Zheng, L.; Bingham, C.O., 3rd; Shah, A.A. Clinical presentation of immune checkpoint inhibitor-induced inflammatory arthritis differs by immunotherapy regimen. Semin. Arthritis Rheum. 2018, 48, 553–557. [Google Scholar] [CrossRef]
  101. Rönnelid, J.; Hansson, M.; Mathsson-Alm, L.; Cornillet, M.; Reed, E.; Jakobsson, P.-J.; Alfredsson, L.; Holmdahl, R.; Skriner, K.; Serre, G.; et al. Anticitrullinated protein/peptide antibody multiplexing defines an extended group of ACPA-positive rheumatoid arthritis patients with distinct genetic and environmental determinants. Ann. Rheum. Dis. 2017, 77, 203–211. [Google Scholar] [CrossRef] [PubMed]
  102. Trouw, L.; Rispens, T.; Toes, R. Beyond citrullination: Other post-translational protein modifications in rheumatoid arthritis. Nat. Rev. Rheumatol. 2017, 13, 331–339. [Google Scholar] [CrossRef] [PubMed]
  103. Carubbi, F.; Alunno, A.; Gerli, R.; Giacomelli, R. Post-Translational Modifications of Proteins: Novel Insights in the Autoimmune Response in Rheumatoid Arthritis. Cells 2019, 8, 657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Bugatti, S.; Manzo, A.; Montecucco, C.; Caporali, R.F. The Clinical Value of Autoantibodies in Rheumatoid Arthritis. Front. Med. 2018, 5, 339. [Google Scholar] [CrossRef] [PubMed]
  105. Bugatti, S.; Bogliolo, L.; Montecucco, C.; Manzo, A. B cell autoimmunity and bone damage in rheumatoid arthritis. Reumatismo 2016, 68, 117–125. [Google Scholar] [CrossRef] [Green Version]
  106. Ajeganova, S.; van Steenbergen, H.W.; Verheul, M.K.; Forslind, K.; Hafström, I.; Toes, R.E.; Huizinga, T.W.J.; Svensson, B.; Trouw, L.A.; van der Helm-van Mil, A.H.M. The association between anti-carbamylated protein (anti-CarP) antibodies and radiographic progression in early rheumatoid arthritis: A study exploring replication and the added value to ACPA and rheumatoid factor. Ann. Rheum. Dis. 2017, 76, 112–118. [Google Scholar] [CrossRef]
  107. Truchetet, M.E.; Dublanc, S.; Barnetche, T.; Vittecoq, O.; Mariette, X.; Richez, C.; Blanco, P.; Mahler, M.; Contin-Bordes, C.; Schaeverbeke, 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 Rheumatol. 2017, 69, 2292–2302. [Google Scholar] [CrossRef] [Green Version]
  108. Lamacchia, C.; Courvoisier, D.S.; Jarlborg, M.; Bas, S.; Roux-Lombard, P.; Möller, B.; Ciurea, A.; Finckh, A.; Bentow, C.; Martinez-Prat, L.; et al. Predictive value of anti-CarP and anti-PAD3 antibodies alone or in combination with RF and ACPA for the severity of rheumatoid arthritis. Rheumatology 2021, 60, 4598–4608. [Google Scholar] [CrossRef]
  109. Regueiro, C.; Casares-Marfil, D.; Lundberg, K.; Knevel, R.; Acosta-Herrera, M.; Rodriguez-Rodriguez, L.; Lopez-Mejias, R.; Perez-Pampin, E.; Triguero-Martinez, A.; Nuño, L.; et al. HLA–B*08 Identified as the Most Prominently Associated Major Histocompatibility Complex Locus for Anti–Carbamylated Protein Antibody–Positive/Anti–Cyclic Citrullinated Peptide–Negative Rheumatoid Arthritis. Arthritis Rheumatol. 2020, 73, 963–969. [Google Scholar] [CrossRef]
  110. Curran, A.M.; Naik, P.; Giles, J.T.; Darrah, E. PAD enzymes in rheumatoid arthritis: Pathogenic effectors and autoimmune targets. Nat. Rev. Rheumatol. 2020, 16, 301–315. [Google Scholar] [CrossRef]
  111. Li, K.; Mo, W.; Wu, L.; Wu, X.; Luo, C.; Xiao, X.; Jia, X.; Yang, H.; Fei, Y.; Chen, H.; et al. Novel autoantibodies identified in ACPA-negative rheumatoid arthritis. Ann. Rheum. Dis. 2021, 80, 739–747. [Google Scholar] [CrossRef] [PubMed]
  112. Daigo, K.; Inforzato, A.; Barajon, I.; Garlanda, C.; Bottazzi, B.; Meri, S.; Mantovani, A. Pentraxins in the activation and regulation of innate immunity. Immunol. Rev. 2016, 274, 202–217. [Google Scholar] [CrossRef] [Green Version]
  113. McInnes, I.B.; Buckley, C.D.; Isaacs, J. Cytokines in rheumatoid arthritis—Shaping the immunological landscape. Nat. Rev. Rheumatol. 2015, 12, 63–68. [Google Scholar] [CrossRef] [PubMed]
  114. Schett, G.; McInnes, I.B.; Neurath, M.F. Reframing Immune-Mediated Inflammatory Diseases through Signature Cytokine Hubs. N. Engl. J. Med. 2021, 385, 628–639. [Google Scholar] [CrossRef] [PubMed]
  115. Lv, Q.; Yin, Y.; Li, X.; Shan, G.; Wu, X.; Liang, D.; Li, Y.; Zhang, X. The status of rheumatoid factor and anti-cyclic citrullinated peptide antibody are not associated with the effect of anti-TNFalpha agent treatment in patients with rheumatoid arthritis: A meta-analysis. PLoS ONE 2014, 9, e89442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Greenwood, M.; Shipa, M.; Yeoh, S.-A.; Roussou, E.; Mukerjee, D.; Ehrenstein, M.R. Methotrexate reduces withdrawal rates of TNF inhibitors due to ineffectiveness in rheumatoid arthritis but only in patients who are seropositive. Ann. Rheum. Dis. 2020, 79, 1516–1517. [Google Scholar] [CrossRef]
  117. Witte, T. Methotrexate as combination partner of TNF inhibitors and tocilizumab. What is reasonable from an immunological viewpoint? Clin. Rheumatol. 2015, 34, 629–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Kruglov, A.; Drutskaya, M.; Schlienz, D.; Gorshkova, E.; Kurz, K.; Morawietz, L.; Nedospasov, S. Contrasting contributions of TNF from distinct cellular sources in arthritis. Ann. Rheum. Dis. 2020, 79, 1453–1459. [Google Scholar] [CrossRef]
  119. Dennis, G., Jr.; Holweg, C.T.; Kummerfeld, S.K.; Choy, D.F.; Setiadi, A.F.; Hackney, J.A.; Haverty, P.M.; Gilbert, H.; Lin, W.Y.; Diehl, L.; et al. Synovial phenotypes in rheumatoid arthritis correlate with response to biologic therapeutics. Arthritis Res. Ther. 2014, 16, R90. [Google Scholar] [CrossRef] [Green Version]
  120. Humby, F.; Durez, P.; Buch, M.H.; Lewis, M.J.; Rizvi, H.; Rivellese, F.; Nerviani, A.; Giorli, G.; Mahto, A.; Montecucco, C.; et al. Rituximab versus tocilizumab in anti-TNF inadequate responder patients with rheumatoid arthritis (R4RA): 16-week outcomes of a stratified, biopsy-driven, multicentre, open-label, phase 4 randomised controlled trial. Lancet 2021, 397, 305–317. [Google Scholar] [CrossRef]
  121. Szekanecz, Z.; McInnes, I.B.; Schett, G.; Szamosi, S.; Benkő, S.; Szűcs, G. Autoinflammation and autoimmunity across rheumatic and musculoskeletal diseases. Nat. Rev. Rheumatol. 2021, 17, 585–595. [Google Scholar] [CrossRef]
  122. Cohen, S.; Hurd, E.; Cush, J.; Schiff, M.; Weinblatt, M.E.; Moreland, L.; Kremer, J.; Bear, M.B.; Rich, W.J.; McCabe, D. Treatment of rheumatoid arthritis with anakinra, a recombinant human interleukin-1 receptor antagonist, in combination with methotrexate: Results of a twenty-four-week, multicenter, randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2002, 46, 614–624. [Google Scholar] [CrossRef]
  123. Patel, D.D.; Lee, D.M.; Kolbinger, F.; Antoni, C. Effect of IL-17A blockade with secukinumab in autoimmune diseases. Ann. Rheum. Dis. 2013, 72 (Suppl. 2), iii116–iii123. [Google Scholar] [CrossRef]
  124. Orr, C.; Vieira-Sousa, E.; Boyle, D.L.; Buch, M.H.; Buckley, C.D.; Cañete, J.D.; Catrina, A.I.; Choy, E.H.S.; Emery, P.; Fearon, U.; et al. Synovial tissue research: A state-of-the-art review. Nat. Rev. Rheumatol. 2017, 13, 463–475. [Google Scholar] [CrossRef]
  125. Manzo, A.; Bugatti, S.; Caporali, R.F.; Montecucco, C. Histopathology of the synovial tissue: Perspectives for biomarker development in chronic inflammatory arthritides. Reumatismo 2018, 70, 121–132. [Google Scholar] [CrossRef] [Green Version]
  126. Manzo, A.; Bugatti, S.; Rossi, S. Clinical Applications of Synovial Biopsy. Front. Med. 2019, 6, 102. [Google Scholar] [CrossRef] [PubMed]
  127. Bugatti, S.; Bozzalla Cassione, E.; De Stefano, L.; Manzo, A. Established rheumatoid arthritis. The pathogenic aspects. Best Pract. Res. Clin. Rheumatol. 2019, 33, 101478. [Google Scholar] [CrossRef]
  128. Bugatti, S.; Manzo, A.; Bombardieri, M.; Vitolo, B.; Humby, F.; Kelly, S.; Montecucco, C.; Pitzalis, C. Synovial Tissue Heterogeneity and Peripheral Blood Biomarkers. Curr. Rheumatol. Rep. 2011, 13, 440–448. [Google Scholar] [CrossRef] [PubMed]
  129. Van de Sande, M.G.; Baeten, D.L. Immunopathology of synovitis: From histology to molecular pathways. Rheumatology 2016, 55, 599–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Humby, F.; Lewis, M.; Ramamoorthi, N.; Hackney, J.; Barnes, M.R.; Bombardieri, M.; Setiadi, A.F.; Kelly, S.; Bene, F.; DiCicco, M.; et al. Synovial cellular and molecular signatures stratify clinical response to csDMARD therapy and predict radiographic progression in early rheumatoid arthritis patients. Ann. Rheum. Dis. 2019, 78, 761–772. [Google Scholar] [CrossRef] [PubMed]
  131. Bugatti, S.; Manzo, A.; Vitolo, B.; Benaglio, F.; Binda, E.; Scarabelli, M.; Humby, F.; Caporali, R.; Pitzalis, C.; Montecucco, C. High expression levels of the B cell chemoattractant CXCL13 in rheumatoid synovium are a marker of severe disease. Rheumatology 2014, 53, 1886–1895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Bugatti, S.; Manzo, A.; Benaglio, F.; Klersy, C.; Vitolo, B.; Todoerti, M.; Sakellariou, G.; Montecucco, C.; Caporali, R. Serum levels of CXCL13 are associated with ultrasonographic synovitis and predict power Doppler persistence in early rheumatoid arthritis treated with non-biological disease-modifying anti-rheumatic drugs. Arthritis Res. Ther. 2012, 14, R34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Orr, C.; Najm, A.; Biniecka, M.; McGarry, T.; Ng, C.T.; Young, F.; Fearon, U.; Veale, D.J. Synovial Immunophenotype and Anti-Citrullinated Peptide Antibodies in Rheumatoid Arthritis Patients: Relationship to Treatment Response and Radiologic Prognosis. Arthritis Rheumatol. 2017, 69, 2114–2123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Lliso-Ribera, G.; Humby, F.; Lewis, M.; Nerviani, A.; Mauro, D.; Rivellese, F.; Kelly, S.; Hands, R.; Bene, F.; Ramamoorthi, N.; et al. Synovial tissue signatures enhance clinical classification and prognostic/treatment response algorithms in early inflammatory arthritis and predict requirement for subsequent biological therapy: Results from the pathobiology of early arthritis cohort (PEAC). Ann. Rheum. Dis. 2019, 78, 1642–1652. [Google Scholar] [CrossRef] [PubMed]
  135. Rivellese, F.; Humby, F.; Bugatti, S.; Fossati-Jimack, L.; Rizvi, H.; Lucchesi, D.; Lliso-Ribera, G.; Nerviani, A.; Hands, R.E.; Giorli, G.; et al. B Cell Synovitis and Clinical Phenotypes in Rheumatoid Arthritis: Relationship to Disease Stages and Drug Exposure. Arthritis Rheumatol. 2020, 72, 714–725. [Google Scholar] [CrossRef] [Green Version]
  136. Bugatti, S.; Manzo, A.; Caporali, R.F.; Montecucco, C. Assessment of synovitis to predict bone erosions in rheumatoid arthritis. Ther. Adv. Musculoskelet. Dis. 2012, 4, 235–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Van Oosterhout, M.; Bajema, I.; Levarht, E.W.; Toes, R.E.; Huizinga, T.W.; van Laar, J.M. Differences in synovial tissue infiltrates between anti-cyclic citrullinated peptide-positive rheumatoid arthritis and anti-cyclic citrullinated peptide-negative rheumatoid arthritis. Arthritis Rheum. 2008, 58, 53–60. [Google Scholar] [CrossRef]
  138. Floudas, A.; Canavan, M.; McGarry, T.; Mullan, R.; Nagpal, S.; Veale, D.; Fearon, U. ACPA Status Correlates with Differential Immune Profile in Patients with Rheumatoid Arthritis. Cells 2021, 10, 647. [Google Scholar] [CrossRef]
  139. Wu, X.; Liu, Y.; Jin, S.; Wang, M.; Jiao, Y.; Yang, B.; Lu, X.; Ji, X.; Fei, Y.; Yang, H.; et al. Single-cell sequencing of immune cells from anticitrullinated peptide antibody positive and negative rheumatoid arthritis. Nat. Commun. 2021, 12, 4977. [Google Scholar] [CrossRef]
  140. Cañete, J.D.; Santiago, B.; Cantaert, T.; Sanmartí, R.; Palacin, A.; Celis, R.; Graell, E.; Gil-Torregrosa, B.; Baeten, D.; Pablos, J.L. Ectopic lymphoid neogenesis in psoriatic arthritis. Ann. Rheum. Dis. 2007, 66, 720–726. [Google Scholar] [CrossRef] [Green Version]
  141. Alivernini, S.; Bruno, D.; Tolusso, B.; Bui, L.; Petricca, L.; Gigante, M.R.; Birra, D.; Fedele, A.L.; Peluso, G.; Federico, F.; et al. Differential synovial tissue biomarkers among psoriatic arthritis and rheumatoid factor/anti-citrulline antibody-negative rheumatoid arthritis. Arthritis Res. Ther. 2019, 21, 116. [Google Scholar] [CrossRef] [Green Version]
  142. Taylor, P.; Atzeni, F.; Balsa, A.; Gossec, L.; Müller-Ladner, U.; Pope, J. The Key Comorbidities in Patients with Rheumatoid Arthritis: A Narrative Review. J. Clin. Med. 2021, 10, 509. [Google Scholar] [CrossRef] [PubMed]
  143. Pope, J.E.; Choy, E.H. C-reactive protein and implications in rheumatoid arthritis and associated comorbidities. Semin. Arthritis Rheum. 2021, 51, 219–229. [Google Scholar] [CrossRef] [PubMed]
  144. Goodson, N.J.; Wiles, N.J.; Lunt, M.; Barrett, E.M.; Silman, A.J.; Symmons, D.P.M. Mortality in early inflammatory polyarthritis: Cardiovascular mortality is increased in seropositive patients. Arthritis Rheum. 2002, 46, 2010–2019. [Google Scholar] [CrossRef] [PubMed]
  145. Mohning, M.P.; Amigues, I.; Demoruelle, M.K.; Pérez, E.R.F.; Huie, T.J.; Keith, R.K.; Olson, A.L.; Yunt, Z.X.; Chung, J.H.; Hobbs, S.; et al. Duration of rheumatoid arthritis and the risk of developing interstitial lung disease. ERJ Open Res. 2021, 7, 00633–2020. [Google Scholar] [CrossRef] [PubMed]
  146. Bugatti, S.; Bogliolo, L.; Vitolo, B.; Manzo, A.; Montecucco, C.; Caporali, R. Anti-citrullinated protein antibodies and high levels of rheumatoid factor are associated with systemic bone loss in patients with early untreated rheumatoid arthritis. Arthritis Res. Ther. 2016, 18, 226. [Google Scholar] [CrossRef] [Green Version]
  147. Dong, H.; Julien, P.J.; Demoruelle, M.K.; Deane, K.D.; Weisman, M.H. Interstitial lung abnormalities in patients with early rheumatoid arthritis: A pilot study evaluating prevalence and progression. Eur. J. Rheumatol. 2018, 6, 193–198. [Google Scholar] [CrossRef]
  148. Bugatti, S.; Bogliolo, L.; Manzo, A.; De Stefano, L.; Delvino, P.; Motta, F.; Montecucco, C. Impact of Anti-Citrullinated Protein Antibodies on Progressive Systemic Bone Mineral Density Loss in Patients with Early Rheumatoid Arthritis After Two Years of Treat-to-Target. Front. Immunol. 2021, 12, 701922. [Google Scholar] [CrossRef]
  149. Barra, L.J.; Pope, J.; Hitchon, C.; Boire, G.; Schieir, O.; Lin, D.; Thorne, C.J.; Tin, D.; Keystone, E.C.; Haraoui, B.; et al. The effect of rheumatoid arthritis–associated autoantibodies on the incidence of cardiovascular events in a large inception cohort of early inflammatory arthritis. Rheumatology 2017, 56, 768–776. [Google Scholar] [CrossRef] [Green Version]
  150. Kastbom, A.; Ärlestig, L.; Rantapää-Dahlqvist, S. Genetic Variants of the NLRP3 Inflammasome Are Associated with Stroke in Patients with Rheumatoid Arthritis. J. Rheumatol. 2015, 42, 1740–1745. [Google Scholar] [CrossRef]
  151. Lwin, M.N.; Serhal, L.; Holroyd, C.; Edwards, C.J. Rheumatoid Arthritis: The Impact of Mental Health on Disease: A Narrative Review. Rheumatol. Ther. 2020, 7, 457–471. [Google Scholar] [CrossRef] [PubMed]
  152. Süß, P.; Rothe, T.; Hoffmann, A.; Schlachetzki, J.C.M.; Winkler, J. The Joint-Brain Axis: Insights from Rheumatoid Arthritis on the Crosstalk Between Chronic Peripheral Inflammation and the Brain. Front. Immunol. 2020, 11, 612104. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Proposed model of disease pathogenesis in autoantibody-negative rheumatoid arthritis.
Figure 1. Proposed model of disease pathogenesis in autoantibody-negative rheumatoid arthritis.
Ijms 22 12386 g001
Table 1. Genetic, environmental, and immunopathologic differences between autoantibody-positive and -negative rheumatoid arthritis.
Table 1. Genetic, environmental, and immunopathologic differences between autoantibody-positive and -negative rheumatoid arthritis.
Autoantibody-PositiveAutoantibody-Negative
Genetic associationsshared epitope-containing HLA-DRB1 allelesHLA-B*08 with aspartate at position 9 and DRB1*03 with serine at position 11
non-HLA genes
Environmental and endogenous factorsSmoking
silica
air pollution
organic dusts
asbestos
pulmonary inflammation
dysbiosis
early menopause
air pollution
organic dusts
asbestos
pulmonary inflammation
dysbiosis
excess body weight
postpartum
early menopause
depression
post-traumatic stress
immune checkpoint inhibitors
Autoantibodies
Anti-CarP
Anti-acetylated
Anti-PAD
15–65%
40–60%
2–18%
10–15%
10–25%
3–19%
CytokinesTNF-α (lymphoid)
IL-6
TNF-α (myeloid)
IL-6
IL-1β
Synovial pathologymostly lympho-myeloid pattern
higher levels of CD19+ B cells, CD3+ T cells, lymphoid aggregates, germinal centers
lympho-myeloid, diffuse-myeloid and
pauci-immune pattern
lower antigen processing and presentation activity in B cells
lower cytotoxic and exhausted gene expression in T cells
higher proinflammatory cytokine expression in macrophages
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

De Stefano, L.; D’Onofrio, B.; Manzo, A.; Montecucco, C.; Bugatti, S. The Genetic, Environmental, and Immunopathological Complexity of Autoantibody-Negative Rheumatoid Arthritis. Int. J. Mol. Sci. 2021, 22, 12386. https://doi.org/10.3390/ijms222212386

AMA Style

De Stefano L, D’Onofrio B, Manzo A, Montecucco C, Bugatti S. The Genetic, Environmental, and Immunopathological Complexity of Autoantibody-Negative Rheumatoid Arthritis. International Journal of Molecular Sciences. 2021; 22(22):12386. https://doi.org/10.3390/ijms222212386

Chicago/Turabian Style

De Stefano, Ludovico, Bernardo D’Onofrio, Antonio Manzo, Carlomaurizio Montecucco, and Serena Bugatti. 2021. "The Genetic, Environmental, and Immunopathological Complexity of Autoantibody-Negative Rheumatoid Arthritis" International Journal of Molecular Sciences 22, no. 22: 12386. https://doi.org/10.3390/ijms222212386

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop