Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jan 1.
Published in final edited form as: Nat Immunol. 2020 Nov 30;22(1):10–18. doi: 10.1038/s41590-020-00816-x

The Immunology of Rheumatoid Arthritis

Cornelia M Weyand 1,*, Jörg J Goronzy 1
PMCID: PMC8557973  NIHMSID: NIHMS1749342  PMID: 33257900

Abstract

The immunopathogenesis of rheumatoid arthritis (RA) spans multiple decades and begins with production of autoantibodies against posttranslationally modified proteins (Checkpoint 1). After years of asymptomatic autoimmunity and progressive immune system remodeling, tissue tolerance erodes, and joint inflammation ensures as tissue-invasive T effector cells emerge, and protective joint-resident macrophages fail (Checkpoint 2). The transition of synovial stromal cells into auto-aggressive effector cells converts acute synovitis into chronic-destructive synovitis, which is often irreversible (Checkpoint 3). Mechanisms of immune tolerance breakdown are now much better understood. The loss of T cell tolerance is linked to cell-intrinsic defects in the DNA repair machinery, resulting in abnormal cell cycle dynamics, telomere fragility and instability of mitochondrial DNA. Mitochondrial and lysosomal anomalies culminate in a T cell differentiation defect favoring differentiation into short-lived effector T cells with high proliferative activity and tissue invasive potential. This differentiation defect builds on a metabolic platform that shunts glucose away from energy generation towards the cell building and motility program. The next frontier in RA will utilize the prolonged period of asymptomatic autoimmunity to attempt the development of curative interventions by reprogramming T cells back to a tolerant state.

Introduction

Initial evidence for an autoimmune pathogenesis of RA came from the discovery of autoantibodies against the Fc portion of IgG, the so-called rheumatoid factor (RF) first described by the Norwegian investigator Dr Erik Waaler in 1940. Since then, concepts of autoimmunity have been extensively modified and accordingly, RA is now understood as a chronic immune-mediated disease in which multiple immune cells types and signaling networks misfunction to elicit a maladaptive tissue repair process that leads to organ damage predominantly in the joints, but also in the lungs and the vascular system.

Genetic studies have identified >100 genetic polymorphisms conferring disease risk, have confirmed the strong association of RA with MHC class II alleles, and have reinforced the central role of adaptive immunity, particularly effector T cells. Twin studies have emphasized the contribution of environmental exposures, which may function as triggers of innate immunity and may contribute to the breakdown of self-tolerance through modifying protein antigens.

Two developments have dovetailed to give rise to new concepts in the understanding of human autoimmune disease and both have spearheaded new perspectives in the conception of RA. Immunology has seen astonishing progress in the definition of cell types, cell-cell interactions, intracellular signaling pathways, and the genetic control of immune system components. Recognition that T cell and B cell responses to (auto)antigen are under contextual control has opened understanding of non-antigenic determinants in autoimmunity, ranging from microenvironmental regulation to metabolic control of cell fate. Genomics, proteomics, and single cell technology have added granularity to the analysis of immune responses. Equally impactful have been epidemiologic studies that have revised the understanding of when and where self-tolerance is lost and how autoimmunity can persist in individuals for decades before it causes clinically apparent disease.

It is now evident that RA is an almost life-long process in which genetically predisposed individuals lose self-tolerance and begin to produce autoantibodies. A phase of disease risk is followed by a phase of asymptomatic autoimmunity, documented by prototypic autoantibodies that are reactive against posttranslationally modified proteins, often citrullinated antigens. Individuals carrying such antibodies to modified protein antigens (AMPA) are asymptomatic for years to decades. Eventually, some enter a new phase and becomes symptomatic with synovitis. Often, acute joint inflammation transitions into chronic, destructive synovitis. At this stage of disease, the tissue responds with a maladaptive wound healing response, described as pannus formation, which by itself has destructive features and will lead to irreversible tissue injury targeting tendons, cartilage, and bone. The evolution of RA over lifetime includes distinct transition points, at which the disease process takes a dimensional step (Fig.1). At each checkpoint, the immune system fails to maintain tolerance, and protective immunity switches to pathogenic immunity in a disease-specific manner.

Figure 1. Evolution of Rheumatoid Arthritis over Lifetime.

Figure 1.

The disease process of RA begins with transition of an at-risk host to a host with overt autoimmunity. Recognition of modified protein antigens and emergence of autoantibodies mark the loss of self-tolerance (Checkpoint 1). Timing and space of this tolerance defect are not precisely identified, but autoantibodies appear years to decades prior to frank joint disease. The host with overt autoimmunity is clinically asymptomatic. A cell-intrinsic shift in metabolic networks and DNA instability drive T cell mal-differentiation towards tissue-invasive short-lived effector T cells. Failure of tissue tolerance manifests as early synovitis during the sixth decade of life (Checkpoint 2) and transitions relatively fast into chronic synovitis (Checkpoint 3). Auto-aggressive de-differentiated synoviocytes, loss of tissue-protective macrophage populations and T cell pyroptosis drive joint damage. The loss of tissue tolerance is irreversible in the majority of cases.

The Immunology of Pre-clinical RA

As a clinical entity, RA is a disease of the 6th decade of life1. As an immunological entity, RA begins years to decades earlier (Fig.1)2. Genetic polymorphisms contribute 30–60% of risk3,4. HLA-class II alleles confer the strongest risk; specifically, HLA-DRB1 alleles containing a sequence stretch from position 71–74 of the β-chain5,6. This sequence stretch (“shared epitope”), together with amino acid residue 11 and 13, forms an antigen-binding pocket, pocket 4, implicating the HLA-antigen-T-cell-receptor complex in disease risk. Amongst the >100 non-HLA genes predisposing to RA, many are involved in shaping the selection, maturation, and function of T cells7,8.

The conversion of immune health to autoimmunity and the transition to relentless tissue inflammation develops over decades, consistent with progressive immune system remodeling. Functionality of the T cell compartment is highly time-dependent, with profound restructuring as the host ages. Dominated by thymic generation during the prenatal period and during childhood, T cell production switches to alternate processes thereafter, when homeostatic proliferation takes over as the dominant replenishment mechanism and peripheral tolerance mechanisms trump thymic selection9. The repertoire of protective and pathogenic T cells in a 20-year-old is fundamentally different from the repertoire in a 55-year-old with clinically apparent RA10. Time dependence identifies immune system and tissue aging as critical in all stages of RA. The concept of a temporal separation of tolerance breakdown and immune-mediated tissue destruction and the recognition that autoantibodies precede clinical disease by years to decades is a generalized rule in human autoimmune disease. Prospective studies of children at-risk for type 1 diabetes mellitus (T1DM) revealed antibodies reactive against pancreatic β-cells antigens long before target tissue destruction. Autoantibodies against nuclear antigens are present years before the diagnosis of SLE and the spectrum and number of autoantibody specificities progressively increases up to the time of clinical disease11,12.

Checkpoint 1 – Systemic Breakdown of self-tolerance

Moving the initial tolerance breakdown outside of the joint has opened opportunities to define causally relevant upstream events. Epidemiologic studies in the early 90’s revealed the presence of RFs long before synovitis13. Subsequent reports of autoantibodies to citrullinated and carbamylated antigens in the presynovitic stage documented that tolerance breakdown precedes clinically apparent RA14. In contrast to T1DM, there is no clear temporal or functional relationship between the different antigenic systems in RA. Citrullination and carbamylation (resulting in homocitrulline formation) are post-translational protein modifications15. Similarly, RFs can recognize glycation-modified IgG Fc fragment16. Thus, RA autoimmunity is not directed against native peptide sequences but, as a rule, against post-translationally modified proteins, introducing a high degree of multi-reactivity. Since thymic selection does not purge T cells recognizing protein modifications, a peripheral tolerance defect is believed dominant in RA17.

Evidence suggests that the major genetic risk factors for RA promote autoantibody production. Shared epitope positive HLA-DRB1 alleles and the PTPN22 variant are associated with production of RFs and anti-citrullinated protein antibodies (ACPA). T cells can recognize citrullinated antigens in the context of HLA-DRB1*04 and the autoimmune B cell response encompasses a large spectrum of citrullinated proteins, consistent with primary T cell responses to citrullinated peptides presented by B cells1821.

Aside genetic risk factors, peripheral tolerance is maintained by the context in which post-translational modifications occur. Citrullination and carbamylation are quite distinct chemical reactions, both universal and physiological, with no clues for a common denominator to explain disease specificity21. Carbamylation is an enzyme-independent derivatization of lysine, requires the reactive metabolite cyanate and is essentially irreversible. In contrast, citrullination is an enzyme-mediated deimination of an arginine residue. Tissue inflammation and smoking upregulate peptidyl arginine deiminase (PAD) expression22. Although intuitive, self-antigen abundance appears not to be important in the process of quorum sensing that regulates tolerance. Mucosal tissue sites23 and neutrophil nets24,25 may provide relevant context signals through which citrullinated antigens gain immunogenicity, supporting the concept that early steps of tolerance loss may occur in the lung and in the gut. However, how tolerance to the modified peptides is broken remains unclear and a common denominator between citrullination, carbamylation and glycation as opposed to other post-translational modifications has not emerged.

Mechanisms that control the stability/progression of the autoimmune stage are dependent on T cells. Healthy HLA-DR4+ individuals and asymptomatic first-degree relatives of RA patients have typical telomere damage and abnormal T cell differentiation26,27, indicative of pre-disease immune system remodeling. Individuals who lose their autoantibodies and do not develop synovitis lack RA-associated HLA-DRB1 alleles. B cell depletion in subjects with arthralgias delayed the onset but did not prevent development of synovitis28. T cell-dependent antibody isotype switching occurs early in the autoimmune process since autoantibodies are generally present in different isotypes15. Also, somatic hypermutation appears important in disease progression as it generates uncommon N-glycosylation sites in the V region of ACPAs29. More than 90% of ACPAs carry N-linked glycans at their V region, exemplifying the dependence of autoimmunity on T cell help.

Checkpoint 2 – Transition from asymptomatic autoimmunity to tissue inflammation

After a prolonged period of asymptomatic autoimmunity, RA patients experience a second fundamental tolerance defect (Fig.1). The initial breakdown of self-tolerance triggering autoantibody production occurs outside of the joint. Eventually, the disease process shifts localization and innate and adaptive immune cells enter the synovial membrane. Environmental exposures, such as the host’s gut microbiota may function as decisive risk elements30. CD3+ T cells are present in most early synovitis cases31,32 and the histologic phenotype of synovial biopsy samples predicts disease persistence and severity. Decreased frequencies of naïve CD4+ T cells are the strongest predictor for progression from ACPA positivity to synovitis33. Unbiased multiparametric studies in early and untreated patients have confirmed that circulating p-p38+p-cJun+p-NFkB+CD4+ T cells serve as the best classification parameter to distinguish RA patients from healthy individuals34. Epigenetic studies at the earliest stages of joint inflammation and in drug-naïve patients have identified differential methylation pattern specifically in naïve CD4+ T cells35. Lymphopenia-induced T cell expansion functions as a strong inducer and amplifier of chronic synovitis36.

Checkpoint 2, marked clinically by the onset of synovial inflammation, is closely linked to cell-intrinsic defects in CD4+ T cells and is functionally defined by a misdifferentiation step as naïve resting CD4+ T cells convert into memory and effector T cells3739. Naive CD4+ T cells from RA patients transition into highly proliferative, tissue-invasive, and pro-inflammatory effector cells, instead of giving rise to relatively quiescent memory T cells. Equipped with tissue-invasive features, RA CD4+ T cells rapidly induce synovitis in a human-synovium mouse chimera model. This differentiation abnormality is mechanistically linked to defects in the DNA repair machinery and reprogramming of cellular bioenergetics, with both deficiencies connected through insufficient repair of mitochondrial DNA37,38,40 (Fig.2). Presence of these defects in the naïve CD4+ T cell population and in lymph node-residing T cells40 places the pathology outside of the joint. The major offspring of naïve RA CD4+ T cells, short-lived effector T cells, swiftly enter the synovial tissue environment, where they undergo pyroptotic cell death40, sparking intense inflammation. Therefore, they may function as inducers of leukocyte-rich as well as leukocyte-poor synovitis.

Figure 2. DNA Repair Deficits in Rheumatoid Arthritis.

Figure 2.

In patients with RA, molecular defects have been localized to the machinery that senses and repairs DNA double strand breaks. Insufficient DNA repair occurs in hematopoietic stem cells, neutrophils and naïve and memory CD4+ T cells. Age-inappropriate decline of the repair molecules ATM and MRE11A leads to accumulation of damaged DNA in the nucleus, at the telomeric ends and in the mitochondria. Downstream consequences include deviations in cell cycle passage, premature telomeric loss and cytoplasmic leakage of mitochondrial DNA triggering inflammasome activation. Ultimately, insufficient DNA repair gives rise to a T cell differentiation defect favoring the induction of short-lived effector T cells at the expense of long-lived memory precursor cells. Also, unrepaired DNA damage promotes T cell pyroptosis, immune exhaustion and premature immune aging. Breakage of mitochondrial DNA affects the electron transport chain, impairs production of ATP and metabolic intermediates, and reprograms the cell’s energy production and biosynthetic program.

Early studies revealed that naïve RA CD4 T cells have shortening of telomeric sequences41,42 and have shifted their cell surface phenotype33,43,44. Compared to patients with chronic hepatitis C infection or age-matched healthy individuals, RA patients contract the diversity of the CD4 T cell receptor repertoire41. These findings gave rise to the “premature immune aging” hypothesis45,46 proposing that a diagnosis of RA is associated with accelerated immune aging, accumulation of expanded clonotypes and depletion of the proliferative reserve inherent in a healthy naïve T cell population47,48.

Telomeric erosion could be a consequence of high proliferative pressure. Recent studies, however, have introduced a new concept, linking telomeric shortening to fragility of the terminal sequences, thus recognizing the loss of telomeric sequences as a defect in DNA repair49. Underlying molecular processes are beginning to be understood (Fig.2).

Mechanistic studies relevant for the breakdown of tissue tolerance (Checkpoint 2) need to capture the RA disease process before the establishment of synovitis. One approach is to examine naïve T and B cell populations, which reside outside of the synovial lesions, and study their transition from the naïve to the effector/memory state, a transition required for tissue entry. This approach has yielded evidence for several defects in the DNA repair machinery (Fig.2). RA T cells are low-expressers of the serine/threonine kinase ataxia telangiectasis mutated (ATM)50, which senses DNA double-strand breaks and activates the DNA damage checkpoint, controlling cell cycle progression and susceptibility to cell death51. Upon activation, ATM-deficient RA T cells accumulate unrepaired DNA50 and bypass the G2-M cell cycle checkpoint, promoting a hyperproliferative phenotype52. While healthy naïve T cells activate distinct signaling and transcription factor networks that guide their differentiation into short-lived effector (SLEC) or memory precursor cells (MPEC)53, ATM-deficient T cells are strongly biased to develop into SLECs, capable of infiltrating into the synovial membrane, where they function as effective drivers of tissue inflammation. Accumulation of DNA double strand breaks induces upregulation of the damage sensor DNA-PKcs and activation of the stress kinase pathway, culminating in cell death54. As part of what appears to be a coordinated program of dampened DNA repair, RA T cells are also low expressers of the DNA repair nuclease meiotic recombination 11 homolog 1 (MRE11A)49,55, a component of a multifunctional DNA damage repair machine that co-ordinates genomic stability programs at DNA replication forks and at double-strand break sites56. In naïve and memory RA T cells, MRE11A is depleted from two critical territories; the telomeric ends and the mitochondrial genome. Functional consequences include instability of chromosomal ends, leading to damage patterns that are highly infrequent in healthy CD4 T cells, but enriched in RA CD4 T cells49. MRE11A deficiency manifests with telomere fragility, accelerated aging exemplified by CD57 induction and high propensity to invade synovial tissue and promote synovitis. Failure of MRE11A to protect genome stability has equally devastating consequences for mitochondrial DNA (mtDNA). MRE11Alow RA T cells are compromised in operating the mitochondrial electron transport chain, expend low amounts of oxygen, and produce low concentrations of ATP55. The bioenergetic failure is coupled with accumulation and leakage of damaged mtDNA, leading to inflammasome assembly, caspase1 activation and immunogenic cell death55,57.

In essence, defective DNA repair responses commit RA T cells to abnormalities in cell differentiation. Functional outcomes include a bias towards SLEC instead of memory generation, a reduction in T cell longevity and accumulation of effector cells inclined to leave lymphoid storage sites and settle in peripheral tissue environments (Fig.3). The skewing towards SLECs away from protective memory T cells may also compromise host immunity that is so typical for RA.

Figure 3. Metabolic reprogramming and pro-inflammatory effector functions in RA T cells.

Figure 3.

Several molecular defects have been identified which shift the metabolic and bioenergetic conditions in RA T cells, ultimately deviating the cell’s differentiation program and shortening survival. Transcriptional repression of the glycolytic enzyme phosphofructokinase (PFKFB3) impairs pyruvate production and thus mitochondrial function, favoring induction of short-lived effector T cells (SLEC) over long-lived memory T cells. Increased activity of glucose-6-phosphate dehydrogenase (G6DH) shunts glucose into the pentose phosphate pathway, supporting biosynthetic activity and daughter cell generation. Oversupply of NADPH supports the lipogenic program and formation of tissue-invasive membrane structures. Damage of mitochondrial DNA weakens mitochondrial fitness and energy production and triggers inflammasome activation and ultimately immunogenic T cell death. Misrouting of the energy sensor AMPK away from the lysosomal surface, caused by deficiency of N-myristoyltransferase 1, enables unopposed mTORC1 activation and SLEC generation.

The disease concept gives rise to several predictions, many of which are fulfilled in RA patients. (1) T cell memory responses in RA patients would be predicted to be weakened. Supporting data come from studies of antibody responses against biopharmaceutical proteins, so-called anti-drug antibodies elicited by therapeutic biologics58. RA patients are significantly less efficient in producing anti-drug antibodies than patients with spondylarthritis59. (2) Longevity of RA T cells should be compromised43,60. In support, T cell populations in lymph nodes of RA patients are less dense and activated caspase155. The global T cell repertoire is contracted with expanded clonotypes filling the space41,61. (3) The preferential differentiation of naïve CD4+ T cells into SLECs at the expense of MPECs imposes proliferative pressure to compensate for T cell loss. Surviving memory T cell populations, including central and effector memory T cells, are under constant demand to repopulate. First described in RA synovium and peripheral blood61, CD4+CD28 T cells have features of premature aging6264, are clonally expanded and autoreactive. The expansion of CD4+CD28 T cells is a shared feature of patients with RA and patients with coronary artery disease(CAD)6567 providing a common pathomechanisms for these two chronic inflammatory and age-associated disease states. In support, CAD risk in RA patients is related to telomeric lengths as a marker of accelerated immune aging68. Typifying features of aged T cells are the high production of IFN-γ69 and the acquisition of natural killer cell receptors and cytolytic activities6971, classifying them as “innate-like” cells72,73. In most recent work, CD27CD28 T cells were found to mediate cytotoxicity through a sestrin-NKG2D-DAP12 complex, reminiscent of the killing by NK cells74. In contrast to young and less differentiated T cells, such aged T cells are less dependent on antigen and on classical co-stimulatory signaling networks, rendering them highly competent in promoting chronic tissue inflammation. The enrichment of highly differentiated CD27 cytotoxic effector memory T cells in synovial lesion of RA patients has been confirmed by recent single cell transcriptomic and mass cytometry approaches75,76.

The exact timing of when at-risk individuals develop expansion of end-differentiated and clonally expanded effector CD4+ T cells is undetermined, but studies of unaffected siblings of RA patients have described CD4+ T cell oligoclonality27, consistent with inheritance of the differentiation defect and excluding synovial inflammation as the causative driver. Similarly, telomeric erosion has been described in healthy individuals carrying the HLA-DRB1*04 disease risk haplotype26, indicating that the premature immune aging phenotype in the adaptive immune system is part of the risk profile of individuals predisposed to RA, but not a consequence of rheumatoid synovitis. Telomere fragility with age-inappropriate telomeric loss affects the neutrophil lineage of healthy HLA-DRB1*04+ individuals26 and the hematopoietic stem cell compartment in patients77, impairing the capacity to replenish innate as well as adaptive immune cells.

Immunometabolic signatures of pro-inflammatory effector T cells

Insufficient DNA repair not only deviates cell cycle passage, cell differentiation and cell fate, it also reprograms cellular bioenergetics of T cells. Rewiring of metabolic networks in RA T cells supports the cells’ divergence towards SLEC generation (Fig. 3), linking metabolic programs to breakdown of tissue tolerance78. Besides fostering the emergence of short-lived effectors, metabolic activity in RA T cells directly supports pro-inflammatory behaviors, e.g. the T cell motility program38,79 and immunogenic cell death55.

Four key metabolic abnormalities have been identified in the patient-derived naïve CD4 T cells undergoing activation (Fig.3)37:

  1. RA T cells transcriptionally repress the key glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3)80, resulting in reduced pyruvate and lactate production.

  2. RA T cells upregulate Glucose-6-phosphate dehydrogenase (G6PD), shunting glucose away from glycolysis to the pentose phosphate pathway (PPP). With increased concentration of NADPH52,81, cellular redox signaling is impaired, the redox sensor ATM is insufficiently dimerized, the G2/M cell cycle checkpoint fails and cells hyperproliferate. Surplus of NADPH facilitates the lipogenic program, promotes the formation of invasive membrane structures and accelerates tissue invasion79,82.

  3. RA T cells lose mitochondrial MRE11A, damaging mtDNA, impairing the electron transport chain and suppressing ATP production. mtDNA fragments leak into the cytoplasm, are recognized by the inflammasome, activate caspase1 and trigger lytic cell death (T cell pyroptosis). Caspase1 activation is a feature of RA lymph node T cells. Pyroptotic T cells are strongly pro-inflammatory, but remain invisible in the tissue, and escape detection by transcriptomic approaches55,83. Mitochondrial failure reduces production of reactive oxygen species (ROS), aggravating the failure of redox signaling.

    T cell mitochondrial failure has been identified as the mechanism driving autoimmunity in C1q deficiency84.

  4. Energy sensing is defective in RA T cells. Due to insufficiency of N-myristoyltransferase 1 (NMT-1), RA T cells fail to posttranslationally modify the energy sensor 5’ AMP-activated protein kinase (AMPK)85. Non-myristoylated AMPK cannot be recruited to the lysosomal surface, remains inactivated and fails to prevent mTORC1 activation. Persistent mTORC1 activation is a hallmark of SLECs.

The metabolic program of RA T cells is disease-specific and distinct from that in other autoimmune disease, specifically SLE. SLE T cells rely on high mitochondrial ROS production8689. SLE Th17 cells depend on metabolic signaling90,91 consistent with the concept that metabolic conditioning diverges T cell differentiation from protective to pathogenic immunity.

Checkpoint 3 – Transformation of acute synovitis into chronic relentless synovitis

Many, but not all patients, who present to an early synovitis clinic, progress to chronic synovitis and irreversible restructuring of the synovial environment92. Chronic synovitis, imposing further proliferative stress upon innate and adaptive immune cells, may per se function as a disease amplifier93. The synovitic component of RA can be successfully targeted by a multitude of highly sophisticated anti-rheumatic drugs and drug-induced remission is now accepted as the treatment goal. Nevertheless, withdrawal of anti-inflammatory medication mostly prompts recurrence of joint inflammation94, indicating that Checkpoint 3 appears to have no regulatory control over the immune system’s basic defects. Critical determinants in the transition of acute to chronic synovitis must derive from the local tissue environment and detailed studies have demonstrated the active pathogenic role of de-differentiated synoviocytes95. How such synovial fibroblasts are activated is insufficiently understood, but tissue-invasive T cells that undergo pyroptosis are potent inducers of inflammation and remodeling55. Another avenue through which pathogenic T cells drive chronicity relates to the formation of organized lymphoid structures96, that lend intensity and durability to inflammatory immunity.

Application of high-resolution techniques has provided unprecedented insight into the cellular composition of the synovial lesion75. In 51 synovial tissue samples (n=36 RA; n=15 osteoarthritis (OA)), four cell populations were separated (T cells, B cells, monocytes, stromal fibroblast) and subjected to bulk RNA seq analysis. Twenty-six samples were analyzed by cytometry and 21 by single cell RNA expression. To account for lesional variability, RA tissues were split into leukocyte-rich (n=19) and leukocyte-poor (n=17) samples. By integrating multiple data modalities, the study defined 18 cellular subpopulations (Table1), including 4 fibroblast subsets, 4 monocyte subsets, 6 T cell subsets and 4 B cell subsets. Mechanistic studies will need to explore how each of these 18 cell populations contributes to the lasting structural damage in the joint.

Table 1. Heterogeneity of cell populations in synovial tissue from patients with osteoarthritis (OA) and patients with rheumatoid arthritis (RA).

RA samples were subdivided according to the leukocyte content into leukocyte-poor and leukocyte-rich samples. Cell subsets were assigned based on clustering of transcriptomic single cell RNA sequences. Highly expressed genes for each subset are given. Topographic assignments were made based on immunostaining of tissue sections. TNF-α producer status and expression of tumor necrosis factor receptor 1 (TNFR1) are inferred from transcriptomic analysis. Modified from Zhang et al (ref. 75).

Cell type OA Leukocyte poor RA Leukocyte rich RA # Marker Highly expressed genes Feature TNF-α producer TNF R1
Fibroblasts ~60% ~50% ~25% SC-F1 CD90+ CD34+ C3, FOS, PTGFR sublining TNFRSF1A
SC-F2 CD90+ HLA-DRAh HLA-DRB1, HLA-DPA1, IL-6, IFI30 sublining
enriched in RA
IL-6 producer
TNFRSF1A
SC-F3 CD90+ DKK3+ CADM1, AKR1C2, CAPG, COL8A2 sublining TNFRSF1A
SC-F4 CD90 CD55+ ITGA6, HBEGF, CLIC5, HTRA4, DNASE1L3 lining
enriched in OA
TNFRSF1A
Macrophages ~20% ~20% ~20% SC-M1 IL-1B+ RGS2, NR4A2, PLAUR, HBEGF, IL1B, ATF3 pro-inflammatory
IL-1 producer
enriched in RA
TNFA TNFRSF1A
SC-M2 NUPR1+ VSIG4, GPNMB, MERTK, NUPR1, CTSK, HTRA1 IL-1 producer
lost in RA
TNFA TNFRSF1A
SC-M3 C1QA+ CD14, MARCO TNFA TNFRSF1A
SC-M4 SPP1+ LY6E, IFITM3, IFI6, SPP1 IFN-activated enriched in RA TNFA TNFRSF1A
T cells ~8% ~8% ~35% SC-T1 CD4+ CCR7+ SELL, NFKBIZ, LEF1, IL7R TNFA
SC-T2 CD4+ FOXP3+ CTLA4, TIGIT, DUSP4, FOXP3 regulatory T cells TNFA
SC-T3 CD4+ PDCD1+ CXCL13+ CD200 TPH and TFH
IFN-γ producer
enriched in RA
TNFA
SC-T4 CD8+ GZMK+ NKG7, GZMA IFN-γ producer TNFA
SC-T5 CD8+ GNLY+GZMB+ ZNF683, PRF1 cytotoxic lymphocytes (CTLs)
IFN-γ producer
TNFA
SC-T6 CD8+ GZMK+ GZMB+ HLA-DPA1+ HLA-DRB1+ IFN-γ, HLA-DPA1, HLA-DRB1 IFN-γ producer TNFA
B cells ~2% ~1% ~8% SC-B1 IGHD+CD27 IL6, IGHM, CD83, BACH2, CXCR4 naïve
IL-6 producer
TNFA
SC-B2 IGHG3+CD27+ HLA-DPB1, MS4A1, HLA - DRA memory TNFA
SC-B3 CD11c ITGAX+ TBX21+(t-bet) ACTB+ GBP1, ISG15, ITGAX, IFI44L, AICDA, ZEB2 autoimmune-associated B cell (ABC) TNFA
SC-B4 immunoglobulin genes and XBP1 SSR4, MZB1, XBP1, DERL3, CD27 plasmablasts

(1). Synovial fibroblasts

The healthy synovium is a thin mesenchymal membrane that upon inflammatory attack expands through fibroblast growth and extracellular matrix deposition. The inflamed synovial membrane is thickened, extensively vascularized and the subintima is occupied by inflammatory cells, (macrophages, T cells, B cells). Fibroblast growth, transformation and matrix production are a generalized wound response, exemplified by intimal hyperplasia in inflamed blood vessels9799. Fibrotic responses in response to chronic tissue inflammation are maladaptive; proper tissue repair fails, and stromal cells adopt tissue-destructive and pro-inflammatory features.

Elegant studies in RA synovial samples and in experimental murine arthritides have identified 4–5 synovial fibroblast (SF) types, all expressing the “lineage marker” podoplanin (PDPN)75,100102. One SF subset, characterized by the absence of the fibroblast marker CD90/Thy-1, has been located to the lining layer. Sublining SFs, all positive for CD90, have been subsetted into 3–4 subsets and have been functionally characterized as pro-inflammatory effector cells. In adoptive transfer experiments in a murine arthritis model, CD90/Thy-1+FAPa+ sublining fibroblasts function as amplifiers of inflammation, driving more severe and more persistent inflammation102. Transcriptomic analysis suggests that the SF subset with high HLA-DRA expression is a cellular source for IL-675. Notably, all SF subsets express TNF receptor 1, but none of the stromal subpopulations was identified as a TNF-α producer75.

Considering the ability of SFs to sustain and exacerbate synovial inflammation, therapeutic modalities have been developed to block SF growth and function. A phase 2 clinical trial testing an anti-cadherin11 antibody in RA has failed to demonstrate benefit103.

(2). Synovial macrophages

Synovitic tissues of RA patients are a macrophage-rich tissue niche104. Over the last 20 years, remarkable progress has been made in understanding mononuclear phagocytes. Especially, the recognition that tissues contain tissue-resident macrophages that are seeded during embryogenesis and fetal development have prompted studies to examine the cellular identity and the origin of synovial macrophages. Tissue resident macrophages adapt to the specialized tissue compartment and contribute to the formation of tissues niches in which cell-cell interactions are optimized and tailored to local requirements. Notable examples are the microglia of the brain, alveolar macrophages, and epidermal Langerhans cells. It appears that tissue-resident macrophages possess self-renewal capacities, possibly into adulthood105. During adulthood fetal-derived resident macrophages can be gradually replaced by bone-marrow derived macrophages that then adapt to niche-specific phenotypes106. In case of increased demand for tissue-residing macrophages, such as under inflammatory conditions, bone-marrow derived monocytes can compensate107. Thus, organ sites contain different macrophage subsets, with distinct origin and distinct longevity. Superimposed on this cellular heterogeneity of tissue-resident macrophages is their functional heterogeneity, ranging from the well-established pro-defense and pro-inflammatory functions to the reparative and inflammation-resolving capabilities associated with scavenger receptor expression, high phagocytic capacities and contributions to wound healing108,109. Upon inflammatory stimuli, bone-marrow derived monocytes rapidly travel to injured tissues sites and add to the resident macrophage population. Here, a clear distinction between the pro-inflammatory M1 and the pro-resolving M2 phenotypes may not always be possible, particularly not during chronic inflammation, when overlapping waves of pro-inflammatory and anti-inflammatory signals and cells are co-existing110.

CD45+CD14+ synovial cells fall into 4 subpopulations. In labeling studies, the pool of synovial macrophages was majorly dependent on the influx of CD14+ blood monocytes111. Studies applying fate mapping and reporter mice as well as light sheet microscopy have established a specific protective role of lining macrophages in the synovium. CX3CR1+ lining macrophages expressed VSIG4 and TREM2112, indicative of anti-inflammatory and phagocytic capacities. VSIG4 inhibits pro-inflammatory macrophages by driving metabolic adaptations25. A remarkable feature of such synovial lining macrophages is the expression of a gene program suggestive for tight junctions, desmosomes, and cell polarity, compatible with a barrier function, separating the tissue lining and the non-tissue space. In mice, CX3CR1+ lining macrophages are constantly replenished from MHCII+ interstitial macrophages seated in the synovial sublining. Interestingly, MHCII+ macrophages supply a third population of RELMA+ interstitial macrophages with features of anti-inflammatory CD206+ macrophages. Integrity of the shield formed by synovial lining macrophages proofed highly protective against inflammation and elegant studies in murine models demonstrated the disintegration of the macrophage barrier upon synovitis induction. Protective functions of this specialized macrophage subset may be related to the expression of a gene program implicated in the uptake of dying cells and lipid debris (MERTK, AXL, TREM2).

In contrast to the protective VISG4+MERTK+TREM2+ lining macrophages (SC-M2, Tab. 1), IL-1B+ HBEGF+ macrophages (SC-M1, Tab. 1) have all characteristics of pro-inflammatory, tissue-damaging effector cells. Enriched in RA synovium, HBEGF+ macrophages promote fibroblast invasiveness in an epidermal growth factor receptor-dependent manner113.

(3). Synovial T cells

Synovial T cells are dominated by CD4+ T cells, but CD8+ T cells are functionally relevant as well114. Consistent with a persistent immune response, tissue-residing T cells have transitioned from the naïve to the memory status. Single cell transcriptomic analysis combined with cytometry has distinguished three CD4 and three CD8+ subsets75. CD8 T cell clusters have distinct granzyme expression patterns. In leukocyte-rich RA samples, the CD4 T cell population includes CD4+PD-1+ICOS+ T cells able to produce the chemokine CXCL13. Unlike circulating Tfh cells, these CD4 T cells lack expression of CXCR5; they have been named T peripheral helper (Tph) cells, and have been implicated in stimulating B cell responses in both RA and SLE115117. Circulating, clonally expanded, immunosenescent CD4+CD28 end-differentiated effector T cells, equipped with cytotoxic functions and high IFN-γ release, home to the synovial tissue niche61,62.

An unexpected result was the finding that synovial T cells, together with synovial macrophages are the cellular origin of TNF-α75, with TNF receptor 1 expressed on SFs and macrophages. TNF-α’s role in RA synovitis is undebated, raising the possibility that the cytokines is a critical connector between abnormal adaptive immune responses and the adverse tissue remodeling process.

(4). Synovial B cells

B cells are distinctly infrequent in OA and leukocyte-poor RA synovial tissue and account for only 8% of synovial cells in leukocyte-rich RA samples. Nevertheless, they may be functionally important as suggested by B-cell depletion studies in engrafted synovial tissues96 and by the clinical benefit of B-cell depletion in both autoantibody-positive and autoantibody-negative RA patients. High-dimensional phenotyping revealed multiple activated B cell subsets as well as CD38+CD20IgMIgD plasmablasts, consistent with the notion that the inflamed synovium can serve as a site of tertiary lymphoid structures. The most interesting B cell subpopulation were CD11chiITGAX+TBX21+ACTB+ B cells reminiscent of the autoimmune B cells described in the blood of SLE patients118. Such autoimmune B cells were encountered in a distinctly small number of RA patients, commensurate with the cellular heterogeneity of the chronic synovial lesion.

Conclusions and future perspectives

RA is now understood as a decade-long, if not life-long process that falls into phases distinct in time, space, and pathogenesis. A pinnacle defect, or a causative antigen, giving rise to the series of pathogenic steps culminating in joint inflammation has not been identified. Genome-wide association studies have confirmed the MHC region as the strongest genetic risk factor and have identified >100 non-MHC RA risk loci. RA risk loci overlap with human primary immunodeficiency genes and genes mutated in hematological cancer, but the causal variants and the resulting pathogenic mechanisms remain largely unresolved. The systemic breakdown of peripheral self-tolerance as evidenced by autoantibodies occurs many years before detectable clinical manifestations and involves a set of antigens that are not synovium specific. Typically, it takes till the sixth decade of life before a second tolerance defect, the breakdown of tissue tolerance, allows the entrance of innate and adaptive immune cells into the synovial tissue space. This transition point is likely reached because of lifelong immune system remodeling combined with enabling structural deficiencies in the synovial environment. Elegant studies applying fate mapping and single cell sequencing have identified tissue-resident self-renewing synovial macrophages that build a joint-protective barrier. The loss of these endogenous synovial macrophages is a critical element in exposing the synovial membrane to pathogenic immunity. The failure in T cell tolerance has been attributed to cell-endogenous abnormalities that are already present in naïve T cells and together diverge the differentiation program to favor the generation of SLECs instead of long-lived memory T cells. These defects include impaired DNA repair compromising telomeric function and mitochondrial fitness and a distinctly different metabolic program, characterized by the slowdown of glycolytic breakdown, the preponderance of catabolic pathways to build biosynthetic precursors and the mistrafficking of AMPK away from the lysosome. This metabolic signature enables T cells to function as tissue-invasive, pro-inflammatory effector cells. A more recently described effector function of tissue-invading T cells, namely pyroptotic death triggered by the leakage of mitochondrial DNA, broadens the array of tissue-injurious pathways operational in synovitis.

What follows is the transition of acute tissue inflammation into chronic tissue inflammation and a maladaptive tissue remodeling process driven to a large extent by the stromal cells of the synovial tissue environment. Single cell transcriptomic analyses combined with cytometry have highlighted the heterogeneity among immune and stromal cells in the inflamed synovium (Table1). Functional studies have shown that select subsets of highly activated synovial fibroblasts adopt pro-inflammatory and tissue-invasive functionalities and join tissue-infiltrating macrophages and T cells as mediators of tissue damage.

Current treatment strategies target the end stage of disease and are broadly anti-inflammatory. The recognition that RA runs through relatively stable stages and the molecular characterization of the relevant transition points has the potential to identify upstream targets and re-engineer the immune system to halt the disease process prior to irreversible tissue damage. The emerging array of pathogenic pathways in RA inspires entirely new territories for combatting autoimmunity, e.g. by targeting genome stability, mitochondrial biology, organelle biogenesis and the cellular endomembrane system.

Acknowledgments.

This work was supported by the National Institutes of Health (R01 AR042527, R01 HL117913, R01 AI108906, R01 HL142068, and P01 HL129941 to CMW and R01 AI108891, R01 AG045779, U19 AI057266, R01 AI129191 to JJG and the Encrantz Family Discovery Fund.

References

  • 1.Myasoedova E, Davis J, Matteson EL & Crowson CS Is the epidemiology of rheumatoid arthritis changing? Results from a population-based incidence study, 1985–2014. Ann Rheum Dis 79, 440–444 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Mahler M, Martinez-Prat L, Sparks JA & Deane KD Precision medicine in the care of rheumatoid arthritis: Focus on prediction and prevention of future clinically-apparent disease. Autoimmun Rev 19, 102506 (2020). [DOI] [PubMed] [Google Scholar]
  • 3.Eyre S et al. High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis. Nat Genet 44, 1336–1340 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Alarcon GS Epidemiology of rheumatoid arthritis. Rheum Dis Clin North Am 21, 589–604 (1995). [PubMed] [Google Scholar]
  • 5.Viatte S et al. Association of HLA-DRB1 haplotypes with rheumatoid arthritis severity, mortality, and treatment response. JAMA 313, 1645–1656 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Okada Y et al. Risk for ACPA-positive rheumatoid arthritis is driven by shared HLA amino acid polymorphisms in Asian and European populations. Hum Mol Genet 23, 6916–6926 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Amariuta T, Luo Y, Knevel R, Okada Y & Raychaudhuri S Advances in genetics toward identifying pathogenic cell states of rheumatoid arthritis. Immunol Rev 294, 188–204 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hu X et al. Regulation of gene expression in autoimmune disease loci and the genetic basis of proliferation in CD4+ effector memory T cells. PLoS Genet 10, e1004404 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Goronzy JJ & Weyand CM Mechanisms underlying T cell ageing. Nat Rev Immunol 19, 573–583 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Goronzy JJ & Weyand CM Successful and Maladaptive T Cell Aging. Immunity 46, 364–378 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Arbuckle MR et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. N Engl J Med 349, 1526–1533 (2003). [DOI] [PubMed] [Google Scholar]
  • 12.McClain MT et al. The prevalence, onset, and clinical significance of antiphospholipid antibodies prior to diagnosis of systemic lupus erythematosus. Arthritis Rheum 50, 1226–1232 (2004). [DOI] [PubMed] [Google Scholar]
  • 13.Aho K, Heliovaara M, Maatela J, Tuomi T & Palosuo T Rheumatoid factors antedating clinical rheumatoid arthritis. J Rheumatol 18, 1282–1284 (1991). [PubMed] [Google Scholar]
  • 14.Deane KD, Norris JM & Holers VM Preclinical rheumatoid arthritis: identification, evaluation, and future directions for investigation. Rheum Dis Clin North Am 36, 213–241 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.van Delft MAM & Huizinga TWJ An overview of autoantibodies in rheumatoid arthritis. J Autoimmun 110, 102392 (2020). [DOI] [PubMed] [Google Scholar]
  • 16.Ligier S, Fortin PR & Newkirk MM A new antibody in rheumatoid arthritis targeting glycated IgG: IgM anti-IgG-AGE. Br J Rheumatol 37, 1307–1314 (1998). [DOI] [PubMed] [Google Scholar]
  • 17.Raposo B et al. T cells specific for post-translational modifications escape intrathymic tolerance induction. Nat Commun 9, 353 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Berglin E et al. A combination of autoantibodies to cyclic citrullinated peptide (CCP) and HLA-DRB1 locus antigens is strongly associated with future onset of rheumatoid arthritis. Arthritis Res Ther 6, R303–308 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Johansson M, Arlestig L, Hallmans G & Rantapaa-Dahlqvist S PTPN22 polymorphism and anti-cyclic citrullinated peptide antibodies in combination strongly predicts future onset of rheumatoid arthritis and has a specificity of 100% for the disease. Arthritis Res Ther 8, R19 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ge C & Holmdahl R The structure, specificity and function of anti-citrullinated protein antibodies. Nat Rev Rheumatol 15, 503–508 (2019). [DOI] [PubMed] [Google Scholar]
  • 21.Kissel T et al. Antibodies and B cells recognising citrullinated proteins display a broad cross-reactivity towards other post-translational modifications. Ann Rheum Dis 79, 472–480 (2020). [DOI] [PubMed] [Google Scholar]
  • 22.Makrygiannakis D et al. Smoking increases peptidylarginine deiminase 2 enzyme expression in human lungs and increases citrullination in BAL cells. Ann Rheum Dis 67, 1488–1492 (2008). [DOI] [PubMed] [Google Scholar]
  • 23.Holers VM et al. Rheumatoid arthritis and the mucosal origins hypothesis: protection turns to destruction. Nat Rev Rheumatol 14, 542–557 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Khandpur R et al. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci Transl Med 5, 178ra140 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Carmona-Rivera C et al. Synovial fibroblast-neutrophil interactions promote pathogenic adaptive immunity in rheumatoid arthritis. Sci Immunol 2 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Schonland SO et al. Premature telomeric loss in rheumatoid arthritis is genetically determined and involves both myeloid and lymphoid cell lineages. Proc Natl Acad Sci U S A 100, 13471–13476 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Waase I, Kayser C, Carlson PJ, Goronzy JJ & Weyand CM Oligoclonal T cell proliferation in patients with rheumatoid arthritis and their unaffected siblings. Arthritis Rheum 39, 904–913 (1996). [DOI] [PubMed] [Google Scholar]
  • 28.Gerlag DM et al. Effects of B-cell directed therapy on the preclinical stage of rheumatoid arthritis: the PRAIRI study. Ann Rheum Dis 78, 179–185 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kissel T et al. On the presence of HLA-SE alleles and ACPA-IgG variable domain glycosylation in the phase preceding the development of rheumatoid arthritis. Ann Rheum Dis 78, 1616–1620 (2019). [DOI] [PubMed] [Google Scholar]
  • 30.Scher JU et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife 2, e01202 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Humby F 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 78, 761–772 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lewis MJ et al. Molecular Portraits of Early Rheumatoid Arthritis Identify Clinical and Treatment Response Phenotypes. Cell Rep 28, 2455–2470 e2455 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ponchel F et al. T-cell subset abnormalities predict progression along the Inflammatory Arthritis disease continuum: implications for management. Sci Rep 10, 3669 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bader L et al. Candidate Markers for Stratification and Classification in Rheumatoid Arthritis. Front Immunol 10, 1488 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Pitaksalee R et al. Differential CpG DNA methylation in peripheral naive CD4(+) T-cells in early rheumatoid arthritis patients. Clin Epigenetics 12, 54 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tuncel J et al. Self-reactive T cells induce and perpetuate chronic relapsing arthritis. Arthritis Res Ther 22, 95 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Weyand CM & Goronzy JJ Immunometabolism in early and late stages of rheumatoid arthritis. Nat Rev Rheumatol 13, 291–301 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Weyand CM, Shen Y & Goronzy JJ Redox-sensitive signaling in inflammatory T cells and in autoimmune disease. Free Radic Biol Med 125, 36–43 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wu B, Goronzy JJ & Weyand CM Metabolic Fitness of T Cells in Autoimmune Disease. Immunometabolism 2 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Li Y, Goronzy JJ & Weyand CM DNA damage, metabolism and aging in pro-inflammatory T cells: Rheumatoid arthritis as a model system. Exp Gerontol 105, 118–127 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wagner UG, Koetz K, Weyand CM & Goronzy JJ Perturbation of the T cell repertoire in rheumatoid arthritis. Proc Natl Acad Sci U S A 95, 14447–14452 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Koetz K et al. T cell homeostasis in patients with rheumatoid arthritis. Proc Natl Acad Sci U S A 97, 9203–9208 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Burgoyne CH et al. Abnormal T cell differentiation persists in patients with rheumatoid arthritis in clinical remission and predicts relapse. Ann Rheum Dis 67, 750–757 (2008). [DOI] [PubMed] [Google Scholar]
  • 44.Ponchel F et al. Dysregulated lymphocyte proliferation and differentiation in patients with rheumatoid arthritis. Blood 100, 4550–4556 (2002). [DOI] [PubMed] [Google Scholar]
  • 45.Weyand CM, Fujii H, Shao L & Goronzy JJ Rejuvenating the immune system in rheumatoid arthritis. Nat Rev Rheumatol 5, 583–588 (2009). [DOI] [PubMed] [Google Scholar]
  • 46.Goronzy JJ, Shao L & Weyand CM Immune aging and rheumatoid arthritis. Rheum Dis Clin North Am 36, 297–310 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Weyand CM, Yang Z & Goronzy JJ T-cell aging in rheumatoid arthritis. Curr Opin Rheumatol 26, 93–100 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Goronzy JJ, Li G, Yang Z & Weyand CM The janus head of T cell aging - autoimmunity and immunodeficiency. Front Immunol 4, 131 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Li Y et al. Deficient Activity of the Nuclease MRE11A Induces T Cell Aging and Promotes Arthritogenic Effector Functions in Patients with Rheumatoid Arthritis. Immunity 45, 903–916 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Shao L et al. Deficiency of the DNA repair enzyme ATM in rheumatoid arthritis. J Exp Med 206, 1435–1449 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Burger K, Ketley RF & Gullerova M Beyond the Trinity of ATM, ATR, and DNA-PK: Multiple Kinases Shape the DNA Damage Response in Concert With RNA Metabolism. Front Mol Biosci 6, 61 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Yang Z et al. Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Sci Transl Med 8, 331ra338 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Amsen D, Backer RA & Helbig C Decisions on the road to memory. Adv Exp Med Biol 785, 107–120 (2013). [DOI] [PubMed] [Google Scholar]
  • 54.Shao L, Goronzy JJ & Weyand CM DNA-dependent protein kinase catalytic subunit mediates T-cell loss in rheumatoid arthritis. EMBO Mol Med 2, 415–427 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Li Y et al. The DNA Repair Nuclease MRE11A Functions as a Mitochondrial Protector and Prevents T Cell Pyroptosis and Tissue Inflammation. Cell Metab 30, 477–492 e476 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Syed A & Tainer JA The MRE11-RAD50-NBS1 Complex Conducts the Orchestration of Damage Signaling and Outcomes to Stress in DNA Replication and Repair. Annu Rev Biochem 87, 263–294 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mathur A, Hayward JA & Man SM Molecular mechanisms of inflammasome signaling. J Leukoc Biol 103, 233–257 (2018). [DOI] [PubMed] [Google Scholar]
  • 58.Mehta P & Manson JJ What Is the Clinical Relevance of TNF Inhibitor Immunogenicity in the Management of Patients With Rheumatoid Arthritis? Front Immunol 11, 589 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Balsa A et al. Drug immunogenicity in patients with inflammatory arthritis and secondary failure to tumour necrosis factor inhibitor therapies: the REASON study. Rheumatology (Oxford) 57, 688–693 (2018). [DOI] [PubMed] [Google Scholar]
  • 60.Ponchel F et al. Interleukin-7 deficiency in rheumatoid arthritis: consequences for therapy-induced lymphopenia. Arthritis Res Ther 7, R80–92 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Schmidt D, Goronzy JJ & Weyand CM CD4+ CD7- CD28- T cells are expanded in rheumatoid arthritis and are characterized by autoreactivity. J Clin Invest 97, 2027–2037 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Weyand CM, Brandes JC, Schmidt D, Fulbright JW & Goronzy JJ Functional properties of CD4+ CD28- T cells in the aging immune system. Mech Ageing Dev 102, 131–147 (1998). [DOI] [PubMed] [Google Scholar]
  • 63.Yamada H et al. Interleukin-15 selectively expands CD57+ CD28- CD4+ T cells, which are increased in active rheumatoid arthritis. Clin Immunol 124, 328–335 (2007). [DOI] [PubMed] [Google Scholar]
  • 64.Weyand CM, Fulbright JW & Goronzy JJ Immunosenescence, autoimmunity, and rheumatoid arthritis. Exp Gerontol 38, 833–841 (2003). [DOI] [PubMed] [Google Scholar]
  • 65.Liuzzo G et al. Perturbation of the T-cell repertoire in patients with unstable angina. Circulation 100, 2135–2139 (1999). [DOI] [PubMed] [Google Scholar]
  • 66.Liuzzo G et al. Monoclonal T-cell proliferation and plaque instability in acute coronary syndromes. Circulation 101, 2883–2888 (2000). [DOI] [PubMed] [Google Scholar]
  • 67.Gerli R et al. CD4+CD28- T lymphocytes contribute to early atherosclerotic damage in rheumatoid arthritis patients. Circulation 109, 2744–2748 (2004). [DOI] [PubMed] [Google Scholar]
  • 68.Ormseth MJ et al. Telomere Length and Coronary Atherosclerosis in Rheumatoid Arthritis. J Rheumatol 43, 1469–1474 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Liuzzo G et al. Molecular fingerprint of interferon-gamma signaling in unstable angina. Circulation 103, 1509–1514 (2001). [DOI] [PubMed] [Google Scholar]
  • 70.Fasth AE, Bjorkstrom NK, Anthoni M, Malmberg KJ & Malmstrom V Activating NK-cell receptors co-stimulate CD4(+)CD28(−) T cells in patients with rheumatoid arthritis. Eur J Immunol 40, 378–387 (2010). [DOI] [PubMed] [Google Scholar]
  • 71.Warrington KJ, Takemura S, Goronzy JJ & Weyand CM CD4+,CD28- T cells in rheumatoid arthritis patients combine features of the innate and adaptive immune systems. Arthritis Rheum 44, 13–20 (2001). [DOI] [PubMed] [Google Scholar]
  • 72.Snyder MR, Nakajima T, Leibson PJ, Weyand CM & Goronzy JJ Stimulatory killer Ig-like receptors modulate T cell activation through DAP12-dependent and DAP12-independent mechanisms. J Immunol 173, 3725–3731 (2004). [DOI] [PubMed] [Google Scholar]
  • 73.Snyder MR, Lucas M, Vivier E, Weyand CM & Goronzy JJ Selective activation of the c-Jun NH2-terminal protein kinase signaling pathway by stimulatory KIR in the absence of KARAP/DAP12 in CD4+ T cells. J Exp Med 197, 437–449 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Pereira BI et al. Sestrins induce natural killer function in senescent-like CD8(+) T cells. Nat Immunol (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhang F et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat Immunol 20, 928–942 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Fonseka CY et al. Mixed-effects association of single cells identifies an expanded effector CD4(+) T cell subset in rheumatoid arthritis. Sci Transl Med 10 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Colmegna I et al. Defective proliferative capacity and accelerated telomeric loss of hematopoietic progenitor cells in rheumatoid arthritis. Arthritis Rheum 58, 990–1000 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Makowski L, Chaib M & Rathmell JC Immunometabolism: From basic mechanisms to translation. Immunol Rev 295, 5–14 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Shen Y et al. Metabolic control of the scaffold protein TKS5 in tissue-invasive, proinflammatory T cells. Nat Immunol 18, 1025–1034 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Yang Z, Fujii H, Mohan SV, Goronzy JJ & Weyand CM Phosphofructokinase deficiency impairs ATP generation, autophagy, and redox balance in rheumatoid arthritis T cells. J Exp Med 210, 2119–2134 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Yang Z, Goronzy JJ & Weyand CM The glycolytic enzyme PFKFB3/phosphofructokinase regulates autophagy. Autophagy 10, 382–383 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Yang Z, Matteson EL, Goronzy JJ & Weyand CM T-cell metabolism in autoimmune disease. Arthritis Res Ther 17, 29 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Weyand CM, Wu B & Goronzy JJ The metabolic signature of T cells in rheumatoid arthritis. Curr Opin Rheumatol 32, 159–167 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ling GS et al. C1q restrains autoimmunity and viral infection by regulating CD8(+) T cell metabolism. Science 360, 558–563 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wen Z et al. N-myristoyltransferase deficiency impairs activation of kinase AMPK and promotes synovial tissue inflammation. Nat Immunol 20, 313–325 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Krishnan S et al. Alterations in lipid raft composition and dynamics contribute to abnormal T cell responses in systemic lupus erythematosus. J Immunol 172, 7821–7831 (2004). [DOI] [PubMed] [Google Scholar]
  • 87.Fernandez D & Perl A Metabolic control of T cell activation and death in SLE. Autoimmun Rev 8, 184–189 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Fernandez DR et al. Activation of mammalian target of rapamycin controls the loss of TCRzeta in lupus T cells through HRES-1/Rab4-regulated lysosomal degradation. J Immunol 182, 2063–2073 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Choi SC et al. Inhibition of glucose metabolism selectively targets autoreactive follicular helper T cells. Nat Commun 9, 4369 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kono M et al. Pyruvate dehydrogenase phosphatase catalytic subunit 2 limits Th17 differentiation. Proc Natl Acad Sci U S A 115, 9288–9293 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kono M et al. Pyruvate kinase M2 is requisite for Th1 and Th17 differentiation. JCI Insight 4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Taylor PC & Law ST When the first visit to the rheumatologist is established rheumatoid arthritis. Best Pract Res Clin Rheumatol, 101479 (2020). [DOI] [PubMed] [Google Scholar]
  • 93.Verburg RJ et al. Outcome of intensive immunosuppression and autologous stem cell transplantation in patients with severe rheumatoid arthritis is associated with the composition of synovial T cell infiltration. Ann Rheum Dis 64, 1397–1405 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Law ST & Taylor PC Role of biological agents in treatment of rheumatoid arthritis. Pharmacol Res 150, 104497 (2019). [DOI] [PubMed] [Google Scholar]
  • 95.Nygaard G & Firestein GS Restoring synovial homeostasis in rheumatoid arthritis by targeting fibroblast-like synoviocytes. Nat Rev Rheumatol 16, 316–333 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Takemura S et al. Lymphoid neogenesis in rheumatoid synovitis. J Immunol 167, 1072–1080 (2001). [DOI] [PubMed] [Google Scholar]
  • 97.Grumbach IM & Nguyen EK Metabolic Stress. Arterioscler Thromb Vasc Biol 39, 991–997 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wadey K, Lopes J, Bendeck M & George S Role of smooth muscle cells in coronary artery bypass grafting failure. Cardiovasc Res 114, 601–610 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Tinajero MG & Gotlieb AI Recent Developments in Vascular Adventitial Pathobiology: The Dynamic Adventitia as a Complex Regulator of Vascular Disease. Am J Pathol 190, 520–534 (2020). [DOI] [PubMed] [Google Scholar]
  • 100.Croft AP et al. Rheumatoid synovial fibroblasts differentiate into distinct subsets in the presence of cytokines and cartilage. Arthritis Res Ther 18, 270 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Mizoguchi F et al. Functionally distinct disease-associated fibroblast subsets in rheumatoid arthritis. Nat Commun 9, 789 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Croft AP et al. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature 570, 246–251 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Finch R et al. RESULTS OF A PHASE 2 STUDY OF RG6125, ANANTI-CADHERIN-11 MONOCLONAL ANTIBODY, INRHEUMATOID ARTHRITIS PATIENTS WITH ANINADEQUATE RESPONSE TO ANTI-TNFALPHATHERAPY. Ann Rheum Dis 78 (2019). [Google Scholar]
  • 104.Orr C et al. Synovial tissue research: a state-of-the-art review. Nat Rev Rheumatol 13, 463–475 (2017). [DOI] [PubMed] [Google Scholar]
  • 105.Davies LC, Jenkins SJ, Allen JE & Taylor PR Tissue-resident macrophages. Nat Immunol 14, 986–995 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Chakarov S et al. Two distinct interstitial macrophage populations coexist across tissues in specific subtissular niches. Science 363 (2019). [DOI] [PubMed] [Google Scholar]
  • 107.Molawi K & Sieweke MH Monocytes compensate Kupffer cell loss during bacterial infection. Immunity 42, 10–12 (2015). [DOI] [PubMed] [Google Scholar]
  • 108.Murray PJ Immune regulation by monocytes. Semin Immunol 35, 12–18 (2018). [DOI] [PubMed] [Google Scholar]
  • 109.Murray PJ Macrophage Polarization. Annu Rev Physiol 79, 541–566 (2017). [DOI] [PubMed] [Google Scholar]
  • 110.Udalova IA, Mantovani A & Feldmann M Macrophage heterogeneity in the context of rheumatoid arthritis. Nat Rev Rheumatol 12, 472–485 (2016). [DOI] [PubMed] [Google Scholar]
  • 111.Herenius MM et al. Monocyte migration to the synovium in rheumatoid arthritis patients treated with adalimumab. Ann Rheum Dis 70, 1160–1162 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Culemann S et al. Locally renewing resident synovial macrophages provide a protective barrier for the joint. Nature 572, 670–675 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kuo D et al. HBEGF(+) macrophages in rheumatoid arthritis induce fibroblast invasiveness. Sci Transl Med 11 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kang YM et al. CD8 T cells are required for the formation of ectopic germinal centers in rheumatoid synovitis. J Exp Med 195, 1325–1336 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Crotty S Follicular helper CD4 T cells (TFH). Annu Rev Immunol 29, 621–663 (2011). [DOI] [PubMed] [Google Scholar]
  • 116.Bocharnikov AV et al. PD-1hiCXCR5- T peripheral helper cells promote B cell responses in lupus via MAF and IL-21. JCI Insight 4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Rao DA et al. Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature 542, 110–114 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Wang S et al. IL-21 drives expansion and plasma cell differentiation of autoreactive CD11c(hi)T-bet(+) B cells in SLE. Nat Commun 9, 1758 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES