Next Article in Journal
REDD1 Deletion Suppresses NF-κB Signaling in Cardiomyocytes and Prevents Deficits in Cardiac Function in Diabetic Mice
Previous Article in Journal
Altered Morpho-Functional Features of Neurogenesis in Zebrafish Embryos Exposed to Non-Combustion-Derived Magnetite
Previous Article in Special Issue
Autoimmune Hemolytic Anemias: Classifications, Pathophysiology, Diagnoses and Management
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Lung in Rheumatoid Arthritis—Friend or Enemy?

by
Maria-Luciana Anton
1,2,
Anca Cardoneanu
1,2,*,
Alexandra Maria Burlui
1,2,
Ioana Ruxandra Mihai
1,2,
Patricia Richter
1,2,
Ioana Bratoiu
1,2,
Luana Andreea Macovei
1,2 and
Elena Rezus
1,2
1
Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
2
Clinical Rehabilitation Hospital, 700661 Iasi, Romania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(12), 6460; https://doi.org/10.3390/ijms25126460
Submission received: 9 April 2024 / Revised: 9 June 2024 / Accepted: 9 June 2024 / Published: 12 June 2024
(This article belongs to the Special Issue Autoimmune Diseases: A Swing Dance of Immune Cells, 2nd Edition)

Abstract

:
Rheumatoid arthritis (RA) is a chronic autoimmune condition frequently found in rheumatological patients that sometimes raises diagnosis and management problems. The pathogenesis of the disease is complex and involves the activation of many cells and intracellular signaling pathways, ultimately leading to the activation of the innate and acquired immune system and producing extensive tissue damage. Along with joint involvement, RA can have numerous extra-articular manifestations (EAMs), among which lung damage, especially interstitial lung disease (ILD), negatively influences the evolution and survival of these patients. Although there are more and more RA-ILD cases, the pathogenesis is incompletely understood. In terms of genetic predisposition, external environmental factors act and subsequently determine the activation of immune system cells such as macrophages, neutrophils, B and T lymphocytes, fibroblasts, and dendritic cells. These, in turn, show the ability to secrete molecules with a proinflammatory role (cytokines, chemokines, growth factors) that will produce important visceral injuries, including pulmonary changes. Currently, there is new evidence that supports the initiation of the systemic immune response at the level of pulmonary mucosa where the citrullination process occurs, whereby the autoantibodies subsequently migrate from the lung to the synovial membrane. The aim of this paper is to provide current data regarding the pathogenesis of RA-associated ILD, starting from environmental triggers and reaching the cellular, humoral, and molecular changes involved in the onset of the disease.

1. Introduction

Rheumatoid arthritis (RA) is considered a “state-of-the-art” autoimmune disorder, being defined by a long-standing inflammation that mainly involves the synovial membrane, leading to symmetrical polyarthritis and having complex extra-articular manifestations (EAMs) [1,2,3,4].
After the cardiovascular system, the lungs represent a frequent site for EAMs (the involvement being extensive) from the pleura to blood vessels. Interstitial lung disease (ILD), a multifaceted disease that was first described by Ellman and Ball in 1948, is known as the most important EAM of RA. The rate of survival from the moment of diagnosis is around 2.6 to 3 years [5,6]. The prevalence of RA-ILD is very large, ranging from 6% to 67% and depends on various aspects such as preexisting risk factors, the setup of RA, disease activity, imagistic patterns, and clinical and paraclinical findings [7,8,9,10]. Even if the pulmonary involvement frequently appears after joint destruction, the lungs may become affected many years in advance [3]. The lifetime risk of progressing to RA-ILD is highlighted to be between 6% and 15% [11,12].
An early diagnosis of RA-ILD is very difficult to make due to the asymptomatic stages of the disease. The evolution of ILD may start with some incidental findings of mild diffuse inflammation, swiftly leading up to life-threatening fibrosis. Depending on the radiographic method used, the prevalence of ILD in RA using a lung X-ray can be below 5% and can reach up to 20–30% when high resolution computed tomography (HRCT) is performed [13,14].
Although there are more and more RA-ILD cases, the pathogenesis is incompletely understood [15,16]. Age, male gender, active smoker status, high titers of anticyclic citrullinated peptide antibodies (ACPAs) or rheumatoid factor (RF), genetic predisposition, and high disease activity are risk factors for RA-associated ILD [17,18,19]. As one of the most significant causes of mortality and morbidity, ILD influences the therapeutic decision and interferes with the “treat-to-target” strategy [20].
The exact pathogenetic mechanisms of RA-ILD are still poorly understood. Paulin et al. highlighted the following two possible pathways: (1) the first site of the immune response is located in the synovial tissue, spreading inflammation into the lungs through the Th1, Th2, and Th17 specific cytokine cascade; fibroblasts undergo differentiation into myofibroblasts, which, in turn, generate an important inflammatory status and produce fibrosis in the pulmonary parenchyma (leading to the characteristic appearance of non-specific interstitial pneumonia (NSIP)); (2) the second theory shows a progressive fibrotic lung pattern due to the preexisting immunological and genetic background, generating an aberrant response in the activation of myofibroblasts and alveolar epithelial cells; after the process of citrullination, ACPAs migrate from the lungs to the synovium (usual interstitial pneumonia (UIP)) [21,22]. With regard to the imaging appearance, the UIP pattern is usually highlighted by honeycombing and a subpleural reticular appearance distributed more at the base of the lung. Moreover, the UIP pattern is defined by unusual damage recovery and unusual epithelial cell activity, being an important risk factor for RA-ILD evolution. In addition, the NSIP pattern is also defined by ground-glass opacities with subpleural distribution, occurring in one-third of RA-ILD patients. This pattern is specific for a longer duration of articular symptoms, a decreased risk of disease evolution, and a superior response to treatment compared with the UIP pattern [23]. Thus, new evidence sustains that the response of the immune system is initiated at the level of pulmonary mucosa [6,23,24].
The first piece of evidence in the pathogenesis of RA was published by Erik Waaler in the 1940s; RF playing the most important role at that time. Nowadays, ACPAs are more specific, although their long presence is not always correlated with RA clinical manifestations. RA is an autoimmune disorder in which many types of cells and signaling paths lead to multiorgan damage [25]. Numerous cells, including dendritic cells (DC), subtypes of T and B cells, macrophages, neutrophils, fibroblasts, and osteoclasts, have been involved in the development and maintenance of joint inflammation [26,27]. RA-associated ILD disease is characterized by a complex interplay between environmental triggers and a distinctive genetic background, leading to a specific loss of immune tolerance [28]. The production of RFs and ACPAs is correlated with human leukocyte antigen (HLA)-DRB1 alleles and the PTPN22 variant. Some HLA haplotypes that encode the β-chain of major histocompatibility complex class two (MHC II) antigens, well known as “shared-epitopes”, raise the risk of developing RA [29,30].
The aim of this paper is to provide a comprehensive review regarding the pathogenesis of RA-ILD. The literature research included computerized databases such as MEDLINE (PubMed), Web of Science (Clarivate Analytics), and Cochrane Library (Cochrane). Search strategies were assembled using database-specific subject headings and keywords such as rheumatoid arthritis, interstitial lung disease, RA-ILD, cytokines, ACPA, and HLA. The research focused on studies published in the last 10 years (2014–2024). We only selected articles regarding human subject studies. This article is based on data from previously published studies.

2. Genetic Predisposition

Of all the genes involved in RA development, HLA-DRB1 alleles (DRB1*01 and DRB1*04; DQ8) represent about 50% [31,32,33]. Different HLA-DRB alleles have a certain risk factor in the appearance of RA-ILD, HLA-DRB1*16 and HLA-DRB1*15 being related to a higher risk [34,35]. Mori et al. [36] noticed that RA patients positive for HLA-DRB1*1501 and *1502 alleles had a higher risk of developing ILD. Charles et al. [37] found a correlation between HLA-B40 and lung involvement in RA. They noticed an increased risk of about 40.54-fold for lung involvement compared with other EAMs. Other studies highlighted a higher frequency of HLA-DR4 (60%) and HLA-B54 (63.2%) polymorphisms in RA-ILD patients versus controls [38,39]. Juge et al. [40] highlighted the correlations between RA-ILD and mutations in familial pulmonary fibrosis-linked genes (TERT, RTEL1, PARN, SFTPC). The MUC5B promoter variant could be involved in the occurrence of RA-associated ILD, but it seems that it plays no role in RA pathogenesis. In addition, the MUC5B variant even has a beneficial role in patients with interstitial pneumonia who have RA-ILD or another autoimmune predisposition [6,41,42,43,44,45]. A variant of the MUC5B promoter (rs35705950) is the strongest genetic risk factor for fibrosis and a strong risk factor for RA-ILD, particularly in patients who develop an UIP pattern [46,47,48]. In a recent study, Palomaki et al. showed that MUC5B carriers had a three-fold risk of developing ILD, with men being the most susceptible [49]. In a recent meta-analysis, the variant of RPA3-UMAD1 was included as a risk factor for RA-ILD in the Japanese population [50]. Another study on Japanese subjects presented rs2609255G as an allele associated with a higher risk for developing the UIP pattern in RA-ILD male patients [51].

3. Risk Factors for RA-ILD

3.1. Age

Advanced age is considered a risk factor in the occurrence of RA-ILD [52,53]. This is due to some changes in the lung parenchyma such as immunosenescence and a decreased regenerative function caused by inflammation and fibrosis [54,55]. RA-ILD occurs frequently in the fifth–sixth decade, while age is considered an independent risk factor for ILD [56,57].

3.2. Patient Gender

Regarding the gender of RA-ILD patients, there are various controversial hypotheses. Although male smokers represent a well-defined risk category, there have been studies that reported a higher incidence in women [58]. Male gender has been evidenced as an independent risk factor for RA-ILD, nodules, or bronchiectasis [59,60], and it has been associated with an unfavorable prognostic [61].

3.3. Race

In an observational study over a period of 13 years, it has been emphasized that the patient’s gender plays an important role with regard to mortality and morbidity rates. A percentage of 26% of the Hispanic population has been diagnosed with RA-ILD, followed by the White population, and it has been concluded that the Black population has the lowest incidence [62]. Native American patients are more predisposed to developing RA [63]. Indians and Japanese diagnosed with RA also have a higher risk of developing ILD, while severe exacerbations have often been observed in Japanese individuals [64,65].

3.4. Smoking

Smoking may be considered the most important exogenous risk factor for RA-ILD, being first described 30 years ago. The risk is correlated with the number of cigarettes smoked and is maintained several years after stopping smoking [66,67]. Various cigarette components have been associated with RA pathogenesis, particularly polycyclic aromatic hydrocarbons (PAHs), which have been shown to trigger a specific transcription factor in synovial DCs, which leads to a higher production of interleukin-6 (IL-6). The ligand-activating transcription factor for PAHs is also present in the synovial tissue of RA patients [68]. Particularly, in RA smokers, joint inflammation is initiated using a subset of synovial DCs, suggesting a possible correlation between DCs, cigarette smoke, and inflammation. Individuals who smoke have been shown to have a higher number of DCs at the level of the airways and interstitial lung tissue [69]. Nicotine can trigger autoimmunity related to RA-ILD pathogenesis due to the secretion of neutrophil extracellular traps (NETs), which are a source of ACPAs and support the activation and secretion of cytokines by synoviocytes [70,71]. Moreover, nicotine can mediate airway and parenchymal lung injury through damage of the endothelial and epithelial cell barriers, which stimulates the recruitment of proinflammatory cells, the secretion of transforming growth factor beta (TGFβ), and the epithelial-to-mesenchymal transition (EMT) [72,73]. Miyake et al. [74] noticed that individuals who smoke have a 2.21 higher risk of developing ILD. Saag et al. [75] reported that smokers with a smoking history of ≥25 pack-years have a 3.8 higher risk for ILD. In another case-control study, Baumgartner et al. [76] demonstrated that people with a history of smoking had a 1.6 higher risk of developing ILD. Restrepo et al. found that patients who had HLA-DRB1 shared epitopes and an active smoker status had an increased susceptibility to develop RA-ILD. This particular HLA genotype increases the risk of developing RA and ACPAs, especially in smokers [17].

3.5. External Pollutants

Workplace exposure to silica, ammonium, mineral dust, and black carbon particles increases the risk of developing pulmonary manifestations in RA [77,78,79,80]. Following contact with external pollutants, an elevated number of inflammatory cells and cytokines are secreted and this causes the inflammatory reaction. Regarding the disruption of the immune system, any change participates in the activation of the inflammatory process, both local and systemic. Liu et al. evidenced that environmental pollutants such as PM2.5, PM10, sulfur dioxide, and nitrogen dioxide may be considered causative factors for frequent hospitalizations in RA-ILD patients [81]. The exact mechanism of the involvement of air pollutants in the development of RA-ILD is not yet fully understood.

3.6. Disease Activity of RA

Low disease activity or complete remission have been associated with better RA prognosis [82,83]. Using the disease activity score 28 (DAS28) and the clinical disease activity index (CDAI), the activity of RA is the most important risk factor for RA-ILD [84,85]. In a recent study, Zhuo et al. showed that RA may present increased disease activity during ILD progression [86]. RA-ILD female patients in the fifth decade are most likely to have a moderate or severe disease [87]. Sparks et al. highlighted that patients with mild-to-severe disease activity had a double risk for RA-ILD development. By stopping the inflammation due to RA, pulmonary complications can be avoided [84]. Patients who had high activity scores (DAS28, CDAI) also had an increased mortality and morbidity risk due to RA-ILD [85,88,89].

3.7. Lung Microbiota

Starting in the 19th century, correlations between RA and lung microbiota have been taken into consideration as the “mucosal origin” theory. It was demonstrated that infections with Porphyromonas gingivalis (P. gingivalis) can induce an immunological response through citrullinated peptides [11,90]. The process is activated by PADs, with arginine components of self-proteins being transformed into neutral citrulline residues. Following these changes, the susceptibility of self-citrullinated proteins to degradation is higher, and many neoepitopes are generated [31]. Moreover, Epstein–Barr virus and bacterial infection with Proteus mirabilis and Escherichia coli were considered triggers for RA through a molecular mimicry mechanism [26,27,91]. Known as “not sterile” sites, the tracheobronchial structure and the lung parenchyma have complex and diverse microbiomes, which can be easily influenced by triggers such as smoking, antibiotics, exogenous substances, or other endogenous factors.
In RA patients, microbiota dysbiosis begins at the level of oral cavity and later extends to the pulmonary level [92,93,94]. It was highlighted that early-onset RA was associated with periodontal diseases. Infections with Prevotella species or P. gingivalis are trigger factors for RA, usually in ACPA-positive patients [95,96,97]. The reactions between bacteria and lung parenchyma influence the innate and adaptive immune system and induce autoimmunity via molecular mimicry, bacteria-induced autoantigen production, and immune regulatory reactions [98,99,100]. Moreover, some studies revealed that ACPAs overreact with citrullinated molecules, which are derived from bacterial species [66,101]. Figure 1 summarizes the main risk factors involved in the development of RA-ILD disease.

4. Immunological Profile in RA-ILD

4.1. ACPA and RF

Antibodies related to RA, RF, and ACPA (also including carbamylated proteins) appear several years before the clinical manifestations and participate in the development of the disease [45]. The agglutinating IgM RFs confer RA seropositivity [102]. Citrullination represents the post-translational change of peptides, catalyzed by PAD, leading to the transformation of arginine into citrulline. This structural adjustment modifies the intercommunication of the immune system with the proteins. Following this modification, the immune system does not recognize the protein anymore, and therefore auto anticitrullinated antibodies are formed [15]. These autoantibodies can bind to residues of citrullinated protein and “self” proteins like fibrinogen, type II collagen, vimentin, α-enolase, fibronectin, and histones. ACPAs can be found in 60–80% of RA patients, with a specificity up to 96–99%. RF and ACPA-positive patients have a 40% risk of disease onset [103]. Regarding lung damage, ACPAs were found in higher titers in RA-ILD patient serum than in the serum of patients with RA without ILD [104].
The pathogenic role of ACPAs in RA-ILD has not been completely understood, even though affected patients showing higher ACPA serum titers and increased ACPA levels have been correlated with ILD and airway disease. ACPAs can contribute to cell injury through the formation of immune complexes and by increasing the secretion of proinflammatory cytokines like tumor necrosis factor (TNF), IL-6, and IL-8; moreover, ACPAs can increase the secretion of NETs. Along with RF, ACPAs create immune complexes and activate macrophages, increasing the secretion of inflammatory cytokines such as IL-6 and TNF-α [3]. It is interesting that, in pre-RA, ACPAs can be formed up to 10 years before the onset of the first symptoms of the disease. There are studies that have shown an association between ACPAs and the risk of developing bone erosions. In this case, ACPAs may increase bone resorption via the following two ways: (i) direct recognition of citrullinated proteins from the surface of osteoclast precursor cells, which leads to the formation of osteoclasts, or (ii) activation of macrophages mediated by immune complexes, which may release proinflammatory cytokines, which in turn activate osteoclasts. Furthermore, activation and differentiation of IL-8-dependent osteoclasts by ACPAs has been associated with joint pain [105,106]. In addition, ACPA may regulate the release of NETs and neutrophil cell death, thus maintaining inflammation and autoimmunity [34].
Some studies revealed a strong correlation between high titers of ACPAs and ILD regarding the development of RA-ILD [107,108,109]. Dai et al. and Correia et al. showed the role of elevated levels of ACPAs in the higher prevalence of ILD [110,111]. Other studies showed that serum RF and ACPA levels were notably high in patients with RA-ILD [13,39]. Giles et al. indicated that, regardless of the type of ACPAs, the patients showed radiographic lung changes of ILD [112]. In a meta-analysis, Zhu et al. also revealed that the positivity of ACPAs was strongly correlated with a risk of developing RA-ILD [113].
Moreover, antibodies such as IgA and IgG RF can be present in the sputum of patients who have a higher risk of RA (like a first-degree relative with RA), even if RF or ACPAs are not highlighted in the serum. Due to the damaged integrity of the mucosal barrier, these antibodies can later leak into the systemic circulation [114]. Upon lung tissue examination, almost half of RA-ILD patients showed evidence of citrullinated proteins [115].
The percentage of citrullination in idiopathic pulmonary fibrosis is also important and almost identical to that in RA-ILD, which reinforces the essential pathogenic role of citrullination in the occurrence of lung damage [115,116]. Recent studies showed that higher titers of RF in association with smoking can lead to the emergence of RA-ILD [60,61,117]. Moreover, high levels of IgA-RF may be considered an important predictor for RA-ILD evolution [61]. A recent study found that there were other antibodies correlated with the development of ILD in RA patients, such as antibodies against citrullinated alpha-enolasepeptide 1, especially in Chinese and Italian populations [90].

4.2. Peptidyl Arginine Deiminases Enzymes

PADs have a decisive role in the process of citrullination. Anti-PAD2 antibodies are correlated with moderate disease and few EAMs, while anti-PAD4 antibodies are associated with a severe and rapidly progressive disease. Anti-PAD3/4XR are reactive antibodies that may signal the development of RA-ILD, especially in the case of non-smoking patients [42,118,119,120]. In a recent cross-sectional study, Nava-Quiroz et al. highlighted that there were some nucleotide variants of PADI2 and PADI4 that were highly correlated with RA-ILD. The rs1005753-GG of PADI2 and rs11203366-AA and rs11203367-GG of PADI4 were strongly connected to genetic predisposition in RA-ILD patients. PAD4 serum titers were elevated in RA-ILD subjects, while increased levels of PAD2 were observed in patients without lung damage [121].

4.3. Anticarbamylated Protein Antibodies

Anticarbamylated protein (Anti-CarPs) antibodies (highlighted for the first time in 2011) were correlated with poor prognosis and increased morbidity and were associated with RA-ILD [90,122]. Moreover, Anti-CarP antibodies have been evidenced in patients with other pulmonary pathologies, without RA [42,123,124]. Few Anti-CarP antibodies were in higher serum titers and strongly related to RA-ILD development, e.g., IgG anti-fetal calf serum (FCS), antichimeric fibrin/filaggrin homocitrullinated peptide (Anti-CFFHP), antifibrinogen (Anti-Fib), and IgA anti-FCS [90]. In a cross-sectional study, Castellanos-Moreira et al. showed increased levels of Anti-Fib and Anti-CFFHP and an independent association between ILD and Anti-CFFHP, Anti-FCS, and IgA Anti-FCS. Furthermore, increased levels of RFs and ACPAs were found among patients with Anti-CFFHP, Anti-FCS, IgA Anti-FCS, and Anti-Fib antibodies [122].

4.4. Antibodies against Malondialdehyde

Antibodies against malondialdehyde (anti-MMA) are strongly expressed in the lung parenchyma of RA-ILD patients, especially IgA and IgM isotypes. A moderate to severe disease activity was associated with the positivity of anti-MMA antibodies [125,126,127,128].

5. Cell Profiles in RA-ILD

5.1. Macrophages

Macrophages coordinate the inflammation process and mediate fibrosis formation. These cells are very versatile and show high plasticity and various immunity functions. Macrophages initiate the inflammatory response in the case of an injury and contribute to the repair and resolution of the injury. In the case of the lung and airway microenvironment, macrophages control the development and establishment of ILD. Macrophages and their products are involved in each main step of this process. The appearance of fibrosis occurs through three mechanisms. First, is the reduction of PAD2 in fibroblasts and the reduction of collagen deposits in the lungs; secondly, macrophages secrete high levels of IL-6, which determine the proliferation of T cells; the third stage involves high levels of TNF-α produced by macrophages that lead to inflammation and fibrosis in the lungs [129]. Two types of macrophages are defined regarding inflammation and immune modulation: M1 (classical or proinflammatory macrophage) and M2 (alternatively activated) [68]. M1 macrophages induce myofibroblast degradation and deteriorate the extracellular matrix, being present at the beginning of the injury. On the other side, M2 macrophages turn on fibroblasts over TGF-β1 and platelet-derived growth factor (PDGF-β) and suppress extracellular matrix deterioration, being involved in reducing the inflammatory process [52]. In the pathogenesis of RA-ILD, through the phagocytic function acquired during the inflammatory process, these two categories of macrophage contribute to the development of fibrosis in the lung parenchyma [130]. This macrophage activation plays an important role by amplifying the cytokine storm, inducing secretion of cytokines, activating the Th1/Th17 pathway, and suppressing regulatory cells [130,131]. M1 macrophages are involved in the process of fibrosis through the secretion of proinflammatory cytokines such as IL-6, IL-12, IL-23, IL-1β, and TNF-α. In addition, M2 macrophages contribute to fibrosis particularly by secreting profibrotic cytokines such as IL-4, IL-10, IL-33, and TGF-β [131]. Macrophage autoimmune–complement–interferon cascade genes are modified in RA-ILD and can have a profibrotic inflammatory response in the lungs. TNF-α leads to an inflammatory phase dominated by cell infiltration, followed by the fibrotic phase characterized by the irreversible deposition of collagen fibers in the pulmonary parenchyma [129,132].
Similar to fibroblasts, subpopulations of macrophages like alveolar macrophages, macrophages derived from bone marrow, and those derived from monocytes are involved in lung fibrosis; depending on the stages of fibrotic diseases, alveolar macrophages can also have antifibrotic effects. In a preclinical study, alveolar cell injury can activate inflammation and macrophages derived from monocytes, leading to fibrosis [133]. Lung samples from patients diagnosed with fibrosis showed various proportions of macrophage populations when compared with normal lungs. The high inflammatory capacity of alveolar macrophages in ILD is suggested by the increased release of reactive oxygen species (RoS), plasminogen activators, mitogens from mesenchymal cells, cytokines, chemokines, leukotrienes, and growth factors [134]. These macrophages have common alveolar and interstitial macrophage characteristics and profibrotic immune cell functions. In an inflamed synovium, the activated macrophages secrete an increased number of cytokines like IL-1β, TNF-α, and IL-6. These cytokines can recruit and activate other natural immune cells such as neutrophils. In turn, the activated neutrophils release high levels of cytokines and oxidants, like phospholipases, myeloperoxidases, proteases, defensins, and TNF-α, favoring joint destruction [135]. On the other hand, proinflammatory cells play a role in fibrosis via interleukin IL-4 and IL-13, inducing the production of TGF-β [136].

5.2. Neutrophils

Neutrophils produce tissue inhibitors of metalloproteinases and neutrophil elastase. They activate TGF-β and determine other inflammatory cells to migrate into the lungs, promoting pulmonary fibrosis [137,138].
Neutrophils have the highest cytotoxic potential of all the cells involved in the RA pathogenesis. This is achieved due to their ability to release reactive oxygen species and enzymes with a degrading role. They actively participate in the chemokine and cytokine cascades. Moreover, neutrophils regulate immune responses through cell–cell interactions. There is evidence that suggests that neutrophils play an established role in inflammation due to the release of NETs, which contain chromatin associated with granule enzymes. The neutrophils of RA patients have a higher predisposition to release NETs. They eliminate extracellular microorganisms and represent a source of autoantigens. This role was unrecognized before. Citrullinated proteins, which may act as neoepitopes in the situation of immune tolerance loss, are produced by PADs [39]. In RA patients, antibodies against citrullinated proteins can be found before any clinical symptoms, predicting an erosive disease. Neutrophils play an immunoregulatory and cytotoxic role in RA and may also represent an important source of autoantigens that drive the autoimmune processes [39].
The receptors of neutrophils can make interactions with vascular endothelial cells, which regulate the attachment and later migration of neutrophils from the circulation into tissues during inflammation. These circulating neutrophils are different and have specific properties. Inflammatory neutrophils have functions similar to macrophages, leading to the increased release of inflammatory molecules such as chemokines, cytokines, and MHC class II antigens. Most of these functions are the result of the selective changes in gene expression that take place upon neutrophil activation during inflammation [39].
In general, circulating neutrophils do not have a wide lifespan and experience constitutive apoptosis after 24 h. While migrating from the circulation into inflamed sites, the function and longevity of neutrophils is altered. Apoptotic neutrophils are inert in terms of function due to programmed shutdown of their receptor signal transduction pathways. Neutrophils express receptors on cell surfaces, allowing them to be recognized, and undergo phagocytosis by macrophages of other phagocytic cells. At inflammatory sites, this removal of apoptotic neutrophils can stop tissue damage. In the case of inflammatory diseases, impaired neutrophil apoptosis determines a longer survival, which leads to an increased release of cytotoxic products, immunoregulatory chemokines, and cytokines, resulting in prolonged inflammation. Hypoxia and antiapoptotic cytokines like TNF and IL-8 can extend the survival of neutrophils up to several days [6,39].

5.3. Fibroblasts

Fibroblasts are also very important because they offer the initial scaffold for the generation of tissue in wound healing [139]. Fibroblasts can differentiate into myofibroblasts, which support the fibrotic process, after they gain a profibrotic phenotype resistant to apoptosis [35]. This process is considered to have a crucial role in fibrosis pathogenesis, being defined as fibroblast-to-myofibroblast transition (FMT) [40,140]. The senescence of lung parenchyma cellularity plays a key role in pulmonary damage by activating FMT [141,142]. In this process, both adaptive and innate immune cells are involved. Fibrocytes produce chemokines, growth factors, matrix metalloproteinases (MMPs), and cross-linking enzymes, and promote pulmonary fibrosis [137,139]. There is another process called EMT, in which epithelial cells gain mesenchymal characteristics instead of epithelial characteristics. This transformation leads to deposits of myofibroblasts, which are correlated with ILD progression. Both endothelial-to-mesenchymal transformation and EMT represent other sources of myofibroblasts in the pathogenesis of RA-ILD, taking place in the presence of TGF-β. These two transitions involve the following four significant steps: decrease in epithelial adhesion properties, structural reorganization, elevated permeability, and higher capacity of migration [143,144]. Both transitions to mesenchymal transformation contribute to the pathogenesis of RA-ILD, favoring the fibrotic process by maintaining inflammation [130]. Figure 2 illustrates the role of macrophages, neutrophils, and fibroblasts in the pathogenesis of RA-ILD.

5.4. Dendritic Cells

DCs play a crucial role in cellular immune reactions by absorbing, processing, and presenting antigens to T and B cells. The DC phenotype, which expresses surface molecules and produces chemokines and cytokines, controls the equilibrium between the activation of the immune system or the induction and preservation of immune tolerance. There is increasing evidence that a changed function and distribution of DCs in RA-ILD favors autoimmune inflammation [35]. Regarding this, a reduced frequency of plasmacytoid DCs, and the presence of conventional DCs in the plasma of patients with RA, was reported to induce an increased DC migration into inflamed joints [145]. At the level of the joints, DCs produce an increased secretion of IL-23 and IL-12, which support Th17 responses, leading to an imbalance between Th2, Th1, and Th17 responses. Inflammatory DCs from the synovial fluid play an important role in the pathogenesis of RA-ILD by activating Th17 cells through the production of IL-1β, IL-23, IL-6, and TGF-β. In addition, activated plasmacytoid dendritic cells (pDCs) contribute to systemic inflammation through the secretion of interferon (IFN)-β, IFN-α, IL-23, and IL-18. Patients with RA-ILD and ACPAs have an increased level of pDCs in the synovial membrane compared with ACPA-negative patients [4,145].

5.5. T and B Cells

Both T and B cells are very important in the inflammatory process. T cells participate in the activation and formation of myofibroblasts through IL-17 and IL-13. The fibrotic modifications in the lung parenchyma are related to a disturbed Th1/Th2 balance. Th1 is considered the suppressor of fibrosis, while Th2 favors this process. B cells are correlated with ACPA formation, thus inducing the production of IL-6, IL-8, and TNF [35]. T-lymphocytes support the differentiation of B cells after exposure to specific antigens [90]. Studies highlighted that CD4+ and CD8+ T cells were more involved in the development of RA-ILD, particularly in smoking patients [11,44,146,147].

6. Cytokine Profile in RA-ILD

Inflammation of the alveolar epithelium can be induced by many environmental factors like oxidative stress, pollutants, infection, or smoking [34,148,149]. In general, inflammation triggers the repair pathway that includes normal fibroblasts and myofibroblasts, which leads to a normal homeostatic response that heals and restores the lung anatomy [150]. Proinflammatory cytokines influence important biological processes such as cell growth, differentiation, proliferation, regulation of the immune response, and tissue repair [151,152]. This pathway mediated by cytokines is crucial for RA pathogenesis [153,154].

6.1. TNF-α

TNF-α is one of the main regulators. TNF-α is a proinflammatory cytokine associated with ILD evolution. It stimulates the proliferation of fibroblasts and activates the expression of cytokines, PDGF-β, chemokines, and TGF-β [34,40,139]. TNF-α inhibitors are frequently used in the treatment of RA. However, more and more studies have suggested that TNF-α inhibitors are correlated with ILD development and can cause pulmonary toxicity [155,156]. Cytokines secreted by T cells are IFN-γ, interleukin (IL)-17A, receptor activator of nuclear factor KB ligand (RANK-L), and TNF-α [27]. TNF-α is also produced by B cells, NK-cells, and synovial macrophages. TNF-α has been proven to induce bone resorption and cartilage degradation; it also increases RANK-L secretion by osteocytes [157]. There are studies that have highlighted that TNF-α can directly stimulate the differentiation of the monocyte/macrophage lineage cells into osteoclasts through a mechanism independent of RANK-L. The second key role of TNF-α in inflammation is the capacity to stimulate the production of other inflammatory cytokines like IL-6 and IL-1β. These cytokines attract leukocytes and promote the formation of an inflammatory milieu in the synovial membrane [4].
Regarding other cytokines, IL-4 and IL-18 are also elevated in the serum of patients with ILD [42,158]. In addition, IL-13 is high, being associated with lung fibrosis in HRCT [159,160,161,162,163]. IL-11 and IL-33 are correlated with RA-ILD [162,163]. Zhang et al. highlighted that lung fibroblast expressed numerous IL-17A receptors, while IL-23 was associated with the EMT process in RA-ILD [164,165]. Table 1 synthesizes the main interleukins involved in the development of RA-ILD.

6.2. IL-17

IL-17 participates in the activation of osteoclastogenesis and of fibroblast-like synoviocytes and in the recruitment and activation of B cells, macrophages, and neutrophils [166]. Of the six members of the IL-17 cytokine family (IL-17A to IL-17F), IL-17A was the first one described [151]. IL-17A produced by Th17 cells favors neutrophil recruitment and the formation of other proinflammatory cytokines (IL-8 and IL-6 by fibroblastic, epithelial, and endothelial cells). Through all these processes, IL-17A contributes to neoangiogenesis, cartilage destruction, and bone erosion in patients with RA [33,167]. IL-17A has also been shown to increase the production of MMP-1 by synoviocytes, leading to cartilage destruction [4].
IL-17 has two main roles: firstly, to promote and initiate chemotaxis, and secondly to recruit and activate neutrophils into inflamed tissues [168]. IL-17 interacts with the TGF-β signaling pathway to induce pulmonary fibrosis [169].
Figure 3 illustrates the main role of IL-17 and TNF-α in RA-ILD.
Other molecules related to RA-ILD pathogenesis are MMPs. Resulting from epithelial degradation, these proteases play an important role in EMT and are involved in extracellular matrix degradation [90,170]. Moreover, MMPs sustain the connections between inflammatory and fibrotic processes by activating T and B cells, macrophages, and neutrophils [90]. The EMT fibrotic process is maintained by MMP-3, and these proteases are involved in RA-ILD pathogenesis [171,172]. MMP-7 can be considered a biomarker for RA-ILD, while high levels of MMP-3 and MMP-7 are associated with RA activity [173,174,175,176]. Chen et al. highlighted increased titers of MMP-7 in RA-ILD patients [137]. Doyle et al. tried to diagnose ILD in patients with RA using biomarkers such as MMP-7, activation/regulation chemokines, and surfactant protein D [175]. One study found another maker strongly associated with the development of RA-ILD, namely LOXL2 [177,178].
Growth factors, other than cytokines and chemokines, take part in the process of fibroblast transformation, forming a bridge between inflammation and fibrosis [90]. PDGF-β is a molecule that has both a profibrotic and a proinflammatory role, being secreted by macrophages, fibroblasts, and epithelial and endothelial cells. In the pathogenesis of ILD, PDGF-β is as important as TNF-α and TGF-β. The development of lung fibrosis depends on the transformation of fibroblasts into myofibroblasts. Regarding chemokines, their contribution is not well established in the lung inflammatory process [90].
The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) is a significant process generated by growth factors. This favors the transformation of macrophages into an inflammatory phenotype, leading to the secretion of IL-6 and TNF-α and promoting fibrosis. Cytokines that play important roles in the development of ILD such as IL-4, IL-13, IL-11, and IL-3 also stimulate the JAK/STAT pathway [179,180,181].
Krebs von den Lungen 6/MUC1 (KL-6) is a mucin-like glycoprotein that is involved in the fibrotic process, maintaining the inhibition of fibroblast apoptosis in the lungs. KL-6 is activated by type II pneumocytes and by epithelial cells from lung parenchyma. After the initiation of lung damage, this protein is considered a key sign for epithelial damage [182,183]. Being evidenced in increased titers in the serum of RA-ILD patients, KL-6 may contribute to the early detection of ILD [42,182]. It is also correlated with higher mortality and morbidity [184]. Lee et al. evidenced correlations between a certain lung pattern in HRCT, disease activity, and high titers of KL-6 in patient serum with RA-ILD [182].
Table 1. Interleukins associated with RA-ILD.
Table 1. Interleukins associated with RA-ILD.
InterleukinsReferencesPathogenic or Clinical Correlations
IL-4[161]↑ RA-ILD
IL-18[163]↑ RA-ILD
IL-13[159,160,161,162,163]↑ RA-ILD; lung fibrosis on HRCT
IL-11 [160,161]Correlated with RA-ILD activity
IL-33[160,161]Correlated with RA-ILD activity
IL-23[164,165]Associated with EMT
MMP-3[173,174,175,176]Correlated with RA-ILD activity
MMP-7[173,174,175,176]Correlated with RA-ILD activity
PDGF-β[90]Profibrotic/inflammatory role
JAK/STAT[179,180,181]Associated with lung fibrosis
KL-6[182,183,184]↑ RA-ILD; lung fibrosis on HRCT
IL—interleukin, HRCT—high resolution computed tomography, EMT—epithelial-to-mesenchymal transition, ↑—increased.

7. Conclusions

Nowadays, RA is nominated as a “decades-long” disease, and it stands out for its complex and distinguished immunological and genetic background, heterogeneity, and the ability to develop complications in time. RA-ILD is a severe and uncertain global problem, whose pathogenesis is not yet fully understood; the mystery remains whether the onset belongs to the inflammatory process located in the joints or whether it is a consequence of pulmonary parenchyma damage. ILD is known as a serious EAM, the course of RA-ILD being associated with a triple risk for morbidity and mortality.
Many new horizons are being revealed regarding the pathogenic pathways in RA-ILD. It is very important to take into consideration each potential trigger that may influence the development and progression of clinical manifestations. Following the causal relationship between RA and ILD, there is a certain mutual influence. Regarding a serious double-meaning problem, RA affects the lung parenchyma through the autoimmune disorder. In addition, new theories regarding the citrullination process in the lung target this aspect as an extremely important trigger for RA.
The activation of the innate and acquired immune system, the loss of self-tolerance, the activation of numerous cells, and the production of an inflammatory environment that causes tissue destruction remain the key points of these diseases.

Author Contributions

Conceptualization, M.-L.A., A.C. and E.R.; methodology, M.-L.A., A.C. and E.R.; software, A.M.B., P.R. and I.R.M.; validation, I.B., P.R. and L.A.M.; formal analysis, M.-L.A., L.A.M. and P.R.; investigation, A.C., M.-L.A., A.M.B. and I.R.M.; resources, A.C., M.-L.A., I.B. and L.A.M.; data curation, E.R.; writing—original draft preparation, M.-L.A., A.C. and E.R.; writing—review and editing, M.-L.A., A.M.B. and E.R.; visualization, A.C. and E.R.; supervision, E.R.; project administration, E.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Shaw, M.; Collins, B.F.; Ho, L.A.; Raghu, G. Rheumatoid arthritis-associated lung disease. Eur. Respir. Rev. 2015, 24, 1–16. [Google Scholar] [CrossRef] [PubMed]
  2. Jang, S.; Kwon, E.J.; Lee, J.J. Rheumatoid Arthritis: Pathogenic Roles of Diverse Immune Cells. Int. J. Mol. Sci. 2022, 23, 905. [Google Scholar] [CrossRef]
  3. Mitrović, J.; Hrkač, S.; Tečer, J.; Golob, M.; Ljilja Posavec, A.; Kolar Mitrović, H.; Grgurević, L. Pathogenesis of Extraarticular Manifestations in Rheumatoid Arthritis—A Comprehensive Review. Biomedicines 2023, 11, 1262. [Google Scholar] [CrossRef] [PubMed]
  4. Lin, Y.-J.; Anzaghe, M.; Schülke, S. Update on the Pathomechanism, Diagnosis, and Treatment Options for Rheumatoid Arthritis. Cells 2020, 9, 880. [Google Scholar] [CrossRef] [PubMed]
  5. Smolen, J.S.; Aletaha, D.; Barton, A.; Burmester, G.R.; Emery, P.; Firestein, G.S.; Kavanaugh, A.; McInnes, I.B.; Solomon, D.H.; Strand, V.; et al. Rheumatoid Arthritis. Nat. Rev. Dis. Prim. 2018, 4, 18001. [Google Scholar] [CrossRef] [PubMed]
  6. Cerro, C.G.; Parimon, T. Understanding Interstitial Lung Diseases Associated with Connective Tissue Disease (CTD-ILD): Genetics, Cellular Pathophysiology, and Biologic Drivers. Int. J. Mol. Sci. 2023, 24, 2405. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, M.; Yin, J.; Zhang, X. Factors associated with interstitial lung disease in patients with rheumatoid arthritis: A systematic review and meta-analysis. PLoS ONE 2023, 18, e0286191. [Google Scholar] [CrossRef] [PubMed]
  8. Matson, S.M.; Demoruelle, M.K.; Castro, M. Airway Disease in Rheumatoid Arthritis. Ann. Am. Thorac. Soc. 2022, 19, 343–352. [Google Scholar] [CrossRef] [PubMed]
  9. Spagnolo, P.; Lee, J.S.; Sverzellati, N.; Rossi, G.; Cottin, V. The Lung in Rheumatoid Arthritis: Focus on Interstitial Lung Disease. Arthritis Rheumatol. 2018, 70, 1544–1554. [Google Scholar] [CrossRef]
  10. Yunt, Z.X.; Solomon, J.J. Lung Disease in Rheumatoid Arthritis. Rheum. Dis. Clin. N. Am. 2015, 41, 225–236. [Google Scholar] [CrossRef]
  11. McDermott, G.C.; Doyle, T.J.; Sparks, J.A. Interstitial lung disease throughout the rheumatoid arthritis disease course. Curr. Opin. Rheumatol. 2021, 33, 284–291. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, D.; Zhang, J.; Lau, J.; Wang, S.; Taneja, V.; Matteson, E.L.; Vassallo, R. Mechanisms of lung disease development in rheumatoid arthritis. Nat. Rev. Rheumatol. 2019, 15, 581–596. [Google Scholar] [CrossRef] [PubMed]
  13. Olivas-Flores, E.M.; Bonilla-Lara, D.; Gamez-Nava, J.I.; Rocha-Muñoz, A.D.; Gonzalez-Lopez, L. Interstitial lung disease in rheumatoid arthritis: Current concepts in pathogenesis, diagnosis and therapeutics. World J. Rheumatol. 2015, 12, 1–22. [Google Scholar] [CrossRef]
  14. Gonzalez-Lopez, L.; Rocha-Muñoz, A.D.; Ponce-Guarneros, M.; Flores-Chavez, A.; Salazar-Paramo, M.; Nava, A.; Cardona-Muñoz, E.G.; Fajardo-Robledo, N.S.; Zavaleta-Muñiz, S.A.; Garcia-Cobian, T.; et al. Anti-cyclic citrullinated peptide (anti-CCP) and anti-mutated citrullinated vimentin (anti-MCV) relation with extra-articular manifestations in rheumatoid arthritis. J. Immunol. Res. 2014, 2014, 536050. [Google Scholar] [CrossRef] [PubMed]
  15. Matson, S.; Lee, J.; Eickelberg, O. Two sides of the same coin? A review of the similarities and differences between idiopathic pulmonary fibrosis and rheumatoid arthritis-associated interstitial lung disease. Eur. Respir. J. 2021, 57, 2002533. [Google Scholar] [CrossRef]
  16. De Zorzi, E.; Spagnolo, P.; Cocconcelli, E.; Balestro, E.; Iaccarino, L.; Gatto, M.; Benvenuti, F.; Bernardinello, N.; Doria, A.; Maher, T.M.; et al. Thoracic involvement in systemic autoimmune rheumatic diseases: Pathogenesis and management. Clin. Rev. Allergy Immunol. 2022, 63, 472–489. [Google Scholar] [CrossRef] [PubMed]
  17. Cavagna, L.; Monti, S.; Grosso, V.; Boffini, N.; Scorletti, E.; Crepaldi, G. The multifaceted aspects of interstitial lung disease in rheumatoid arthritis. BioMed Res. Int. 2013, 2013, 759760. [Google Scholar] [CrossRef] [PubMed]
  18. Doyle, T.J.; Patel, A.S.; Hatabu, H.; Nishino, M.; Wu, G.; Osorio, J.C.; Golzarri, M.F.; Traslosheros, A.; Chu, S.G.; Frits, M.L.; et al. Detection of rheumatoid arthritis-interstitial lung disease is enhanced by serum biomarkers. Am. J. Respir. Crit. Care Med. 2015, 191, 1403–1412. [Google Scholar] [CrossRef] [PubMed]
  19. Svendsen, A.J.; Junker, P.; Houen, G.; Kyvik, K.O.; Nielsen, C.; Skytthe, A.; Holst, R. Incidence of Chronic Persistent Rheumatoid Arthritis and the Impact of Smoking: A Historical Twin Cohort Study. Arthritis Care Res. 2017, 69, 616–624. [Google Scholar] [CrossRef]
  20. Kuwana, M.; Gil-Vila, A.; Selva-O’Callaghan, A. Role of autoantibodies in the diagnosis and prognosis of interstitial lung disease in autoimmune rheumatic disorders. Ther. Adv. Musculoskelet. Dis. 2021, 13, 1759720X211032457. [Google Scholar] [CrossRef]
  21. Paulin, F.; Babini, A.; Mamani, M.; Mercado, J.; Caro, F. Practical Approach to the Evaluation and Management of Rheumatoid Arthritis-Interstitial Lung Disease Based on its Proven and Hypothetical Mechanisms. Rev. Investig. Clin. 2017, 69, 235–242. [Google Scholar] [CrossRef] [PubMed]
  22. Paulin, F.; Doyle, T.J.; Fletcher, E.A.; Ascherman, D.P.; Rosas, I.O. Rheumatoid Arthritis-Associated Interstitial Lung Disease and Idiopathic Pulmonary Fibrosis: Shared Mechanistic and Phenotypic Traits Suggest Overlapping Disease Mechanisms. Rev. Investig. Clin. 2015, 67, 280–286. [Google Scholar]
  23. Quirke, A.M.; Lugli, E.B.; Wegner, N.; Hamilton, B.C.; Charles, P.; Chowdhury, M.; Ytterberg, A.J.; Zubarev, R.A.; Potempa, J.; Culshaw, S.; et al. Heightened immune response to autocitrullinated Porphyromonas gingivalis peptidylarginine deiminase: A potential mechanism for breaching immunologic tolerance in rheumatoid arthritis. Ann. Rheum. Dis. 2014, 73, 263–269. [Google Scholar] [CrossRef]
  24. Holers, V.M.; Demoruelle, M.K.; Kuhn, K.A.; Buckner, J.H.; Robinson, W.H.; Okamoto, Y.; Norris, J.M.; Deane, K.D. Rheumatoid Arthritis and the Mucosal Origins Hypothesis: Protection Turns to Destruction. Nat. Rev. Rheumatol. 2018, 14, 542–557. [Google Scholar] [CrossRef]
  25. Weyand, C.M.; Goronzy, J.J. The immunology of rheumatoid arthritis. Nat. Immunol. 2021, 22, 10–18. [Google Scholar] [CrossRef]
  26. Zhang, F.; Wei, K.; Slowikowski, K.; Fonseka, C.Y.; Rao, D.A.; Kelly, S.; Goodman, S.M.; Tabechian, D.; Hughes, L.B.; Salomon-Escoto, K.; et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat. Immunol. 2019, 20, 928–942. [Google Scholar] [CrossRef]
  27. Smolen, J.S.; Aletaha, D.; McInnes, I.B. Rheumatoid arthritis. Lancet Lond. Engl. 2016, 388, 2023–2038. [Google Scholar] [CrossRef]
  28. Manfredi, A.; Cassone, G.; Luppi, F.; Atienza-Mateo, B.; Cavazza, A.; Sverzellati, N.; González-Gay, M.A.; Salvarani, C.; Sebastiani, M. Rheumatoid arthritis related interstitial lung disease. Expert Rev. Clin. Immunol. 2021, 17, 485–497. [Google Scholar] [CrossRef] [PubMed]
  29. Viatte, S.; Plant, D.; Raychaudhuri, S. Genetics and epigenetics of rheumatoid arthritis. Nat. Rev. Rheumatol. 2013, 9, 141–153. [Google Scholar] [CrossRef]
  30. Al-Baldawi, S.; Zúñiga Salazar, G.; Zúñiga, D.; Balasubramanian, S.; Mehmood, K.T. Interstitial Lung Disease in Rheumatoid Arthritis: A Review. Cureus 2024, 16, e53632. [Google Scholar] [CrossRef]
  31. Littlejohn, E.A.; Monrad, S. Early Diagnosis and Treatment of Rheumatoid Arthritis. Prim. Care Clin. Off. Pract. 2018, 45, 237–255. [Google Scholar] [CrossRef] [PubMed]
  32. Arleevskaya, M.I.; Kravtsova, O.A.; Lemerle, J.; Renaudineau, Y.; Tsibulkin, A.P. How Rheumatoid Arthritis Can Result from Provocation of the Immune System by Microorganisms and Viruses. Front. Microbiol. 2016, 7, 1296. [Google Scholar] [CrossRef] [PubMed]
  33. Kadura, S.; Raghu, G. Rheumatoid arthritis-interstitial lung disease: Manifestations and current concepts in pathogenesis and management. Eur. Respir. Rev. 2021, 30, 210011. [Google Scholar] [CrossRef] [PubMed]
  34. Newton, C.A.; Oldham, J.M.; Ley, B.; Anand, V.; Adegunsoye, A.; Liu, G.; Batra, K.; Torrealba, J.; Kozlitina, J.; Glazer, C.; et al. Telomere length and genetic variant associations with interstitial lung disease progression and survival. Eur. Respir. J. 2019, 53, 1801641. [Google Scholar] [CrossRef] [PubMed]
  35. Lee, H.; Lee, S.I.; Kim, H.O. Recent Advances in Basic and Clinical Aspects of Rheumatoid Arthritis-associated Interstitial Lung Diseases. J. Rheum. Dis. 2022, 29, 61–70. [Google Scholar] [CrossRef] [PubMed]
  36. Mori, S.; Koga, Y.; Sugimoto, M. Different risk factors between interstitial lung disease and airway disease in rheumatoid arthri- tis. Respir. Med. 2012, 106, 1591–1599. [Google Scholar] [CrossRef] [PubMed]
  37. Charles, P.J.; Sweatman, M.C.; Markwick, J.R.; Maini, R.N. HLA-B40: A marker for susceptibility to lung disease in rheumatoid arthritis. Dis. Mark. 1991, 9, 97–101. [Google Scholar]
  38. Sugiyama, Y.; Ohno, S.; Kano, S.; Maeda, H.; Kitamura, S. Diffuse pan-bronchiolitis and rheumatoid arthritis: A possible correlation with HLA-B54. Intern Med. 1994, 33, 612–614. [Google Scholar] [CrossRef] [PubMed]
  39. McInnes, I.B.; Schett, G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet 2017, 389, 2328–2337. [Google Scholar] [CrossRef]
  40. Juge, P.-A.; Borie, R.; Kannengiesser, C.; Gazal, S.; Revy, P.; Wemeau-Stervinou, L.; Debray, M.-P.; Ottaviani, S.; Marchand-Adam, S.; Nathan, N. Shared genetic predisposition in rheumatoid arthritis-interstitial lung disease and familial pulmonary fibrosis. Eur. Respir. J. 2017, 49, 1602314. [Google Scholar] [CrossRef]
  41. Kim, Y.; Yang, H.-I.; Kim, K.-S. Etiology and Pathogenesis of Rheumatoid Arthritis-Interstitial Lung Disease. Int. J. Mol. Sci. 2023, 24, 14509. [Google Scholar] [CrossRef] [PubMed]
  42. Stainer, A.; Tonutti, A.; De Santis, M.; Amati, F.; Ceribelli, A.; Bongiovanni, G.; Torrisi, C.; Iacopino, A.; Mangiameli, G.; Aliberti, S.; et al. Unmet needs and perspectives in rheumatoid arthritis-associated interstitial lung disease: A critical review. Front. Med. 2023, 10, 1129939. [Google Scholar] [CrossRef]
  43. Jönsson, E.; Ljung, L.; Norrman, E.; Freyhult, E.; Ärlestig, L.; Dahlqvist, J.; Rantapää-Dahlqvist, S. Pulmonary fibrosis in relation to genetic loci in an inception cohort of patients with early rheumatoid arthritis from northern Sweden. Rheumatology 2022, 61, 943–952. [Google Scholar] [CrossRef]
  44. Akiyama, M.; Kaneko, Y. Pathogenesis, clinical features, and treatment strategy for rheumatoid arthritis-associated interstitial lung disease. Autoimmun. Rev. 2022, 21, 103056. [Google Scholar] [CrossRef]
  45. Khan, T.; Jose, R.J.; Renzoni, E.A.; Mouyis, M. A Closer Look at the Role of Anti-CCP Antibodies in the Pathogenesis of Rheumatoid Arthritis-Associated Interstitial Lung Disease and Bronchiectasis. Rheumatol. Ther. 2021, 8, 1463–1475. [Google Scholar] [CrossRef]
  46. Juge, P.A.; Lee, J.S.; Ebstein, E.; Furukawa, H.; Dobrinskikh, E.; Gazal, S.; Kannengiesser, C.; Ottaviani, S.; Oka, S.; Tohma, S.; et al. MUC5B promoter variant and rheumatoid arthritis with interstitial lung disease. N. Engl. J. Med. 2018, 379, 2209–2219. [Google Scholar] [CrossRef]
  47. Zhang, Q.; Wang, Y.; Qu, D.; Yu, J.; Yang, J. The Possible Pathogenesis of Idiopathic Pulmonary Fibrosis considering MUC5B. BioMed Res. Int. 2019, 2019, 9712464. [Google Scholar]
  48. Vermant, M.; Goos, T.; Gogaert, S.; De Cock, D.; Verschueren, P.; Wuyts, W.A. Are genes the missing link to detect and prognosticate RA-ILD? Rheumatol. Adv. Pract. 2023, 7, rkad023. [Google Scholar] [CrossRef] [PubMed]
  49. Palomäki, A.; Palotie, A.; Koskela, J.; Eklund, K.K.; Pirinen, M.; Ripatti, S.; Laitinen, T.; Mars, N.; Group, F.R.C.E. Lifetime risk of rheumatoid arthritis-associated interstitial lung disease in MUC5B mutation carriers. Ann. Rheum. Dis. 2021, 80, 1530–1536. [Google Scholar] [CrossRef]
  50. Shirai, Y.; Honda, S.; Ikari, K.; Kanai, M.; Takeda, Y.; Kamatani, Y.; Morisaki, T.; Tanaka, E.; Kumanogoh, A.; Harigai, M. Association of the RPA3-UMAD1 locus with interstitial lung diseases complicated with rheumatoid arthritis in Japanese. Ann. Rheum. Dis. 2020, 79, 1305–1309. [Google Scholar] [CrossRef]
  51. Higuchi, T.; Oka, S.; Furukawa, H.; Shimada, K.; Tsunoda, S.; Ito, S.; Okamoto, A.; Katayama, M.; Saisho, K.; Shinohara, S. Association of a FAM13A variant with interstitial lung disease in Japanese rheumatoid arthritis. RMD Open 2023, 9, e002828. [Google Scholar] [CrossRef] [PubMed]
  52. Innala, L.; Berglin, E.; Möller, B.; Ljung, L.; Smedby, T.; Södergren, A.; Magnusson, S.; Rantapää-Dahlqvist, S.; Wållberg-Jonsson, S. Age at onset determines severity and choice of treatment in early rheumatoid arthritis: A prospective study. Arthrit. Res. Ther. 2014, 16, R94. [Google Scholar] [CrossRef] [PubMed]
  53. Koduri, G.; Norton, S.; Young, A.; Cox, N.; Davies, P.; Devlin, J.; Dixey, J.; Gough, A.; Prouse, P.; Winfield, J. Interstitial lung disease has a poor prognosis in rheumatoid arthritis: Results from an inception cohort. Rheumatology 2010, 49, 1483–1489. [Google Scholar] [CrossRef]
  54. Aw, D.; Silva, A.B.; Palmer, D.B. Immunosenescence: Emerging challenges for an ageing population. Immunology 2007, 120, 435–446. [Google Scholar] [CrossRef]
  55. Murtha, L.A.; Morten, M.; Schuliga, M.J.; Mabotuwana, N.S.; Hardy, S.A.; Waters, D.W.; Burgess, J.K.; Ngo, D.T.; Sverdlov, A.L.; Knight, D.A. The role of pathological aging in cardiac and pulmonary fibrosis. Aging Dis. 2019, 10, 419–428. [Google Scholar] [CrossRef]
  56. Assayag, D.; Lubin, M.; Lee, J.S.; King, T.E.; Collard, H.R.; Ryerson, C.J. Predictors of mortality in rheumatoid arthritis-related interstitial lung disease. Respirology 2014, 19, 493–500. [Google Scholar] [CrossRef]
  57. Saag, K.G.; Cerhan, J.R.; Kolluri, S.; Ohashi, K.; Hunninghake, G.W.; Schwartz, D.A. Cigarette smoking and rheumatoid arthritis severity. Ann. Rheum. Dis. 1997, 56, 463–469. [Google Scholar] [CrossRef] [PubMed]
  58. Kelly, C.A.; Saravanan, V.; Nisar, M.; Arthanari, S.; Woodhead, F.A.; Price-Forbes, A.N.; Dawson, J.; Sathi, N.; Ahmad, Y.; Koduri, G. Rheumatoid arthritis-related interstitial lung disease: Associations, prognostic factors and physiological and radiological characteristics—A large multicentre UK study. Rheumatology 2014, 53, 1676–1682. [Google Scholar] [CrossRef] [PubMed]
  59. Restrepo, J.F.; Del Rincón, I.; Battafarano, D.F.; Haas, R.W.; Doria, M.; Escalante, A. Clinical and laboratory factors associated with interstitial lung disease in rheumatoid arthritis. Clin. Rheumatol. 2015, 34, 1529–1536. [Google Scholar] [CrossRef]
  60. Klester, E.; Klester, K.; Shoykhet, Y.; Elykomov, V.; Yarkova, V.; Berdyugina, A.; Mukhtarova, E. Risk factors of interstitial lung diseases in patients with rheumatoid arthritis. Eur. Respir. Soc. 2019, 54, PA1365. [Google Scholar]
  61. Chen, N.; Diao, C.-Y.; Gao, J.; Zhao, D.-B. Risk factors for the progression of rheumatoid arthritis-related interstitial lung disease: Clinical features, biomarkers, and treatment options. Semin. Arthritis Rheum. 2022, 55, 152004. [Google Scholar] [CrossRef] [PubMed]
  62. Jeganathan, N.; Nguyen, E.; Sathananthan, M. Rheumatoid arthritis and associated interstitial lung disease: Mortality rates and trends. Ann. Am. Thorac. Soc. 2021, 18, 1970–1977. [Google Scholar] [CrossRef] [PubMed]
  63. Peschken, C.A.; Hitchon, C.A.; Robinson, D.B.; Smolik, I.; Barnabe, C.R.; Prematilake, S.; El-Gabalawy, H.S. Rheumatoid arthritis in a north american native population: Longitudinal followup and comparison with a white population. J. Rheumatol. 2010, 37, 1589–1595. [Google Scholar] [CrossRef] [PubMed]
  64. Jo, H.E.; Corte, T.J. Nintedanib for idiopathic pulmonary fibrosis in the Japanese population. Respirology 2017, 22, 630–631. [Google Scholar] [CrossRef] [PubMed]
  65. Tekaya, A.B.; Mokaddem, S.; Athimini, S.; Kamoun, H.; Mahmoud, I.; Abdelmoula, L. Risk factors for rheumatoid arthritis-associated interstitial lung disease: A retrospective study. Multidiscip. Respir. Med. 2022, 17, 877. [Google Scholar] [PubMed]
  66. 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] [PubMed]
  67. Hedstroem, A.K.; Stawiarz, L.; Klareskog, L.; Alfredsson, L. Smoking and susceptibility to rheumatoid arthritis in a Swedish population-based case control study. Eur. J. Epidemiol. 2018, 33, 415–423. [Google Scholar] [CrossRef] [PubMed]
  68. Nguyen, N.T.; Hanieh, H.; Nakahama, T.; Kishimoto, T. The roles of aryl hydrocarbon receptor in immune responses. Int. Immunol. 2013, 25, 335–343. [Google Scholar] [CrossRef] [PubMed]
  69. Segura, E.; Touzot, M.; Bohineust, A.; Cappuccio, A.; Chiocchia, G.; Hosmalin, A.; Dalod, M.; Soumelis, V.; Amigorena, S. Human Inflammatory Dendritic Cells Induce Th17 Cell Differentiation. Immunity 2013, 38, 336–348. [Google Scholar] [CrossRef]
  70. Lee, J.; Luria, A.; Rhodes, C.; Raghu, H.; Lingampalli, N.; Sharpe, O.; Rada, B.; Sohn, D.H.; Robinson, W.H.; Sokolove, J. Nicotine Drives Neutrophil Extracellular Traps Formation and Accelerates Collagen-Induced Arthritis. Rheumatology 2017, 56, 644–653. [Google Scholar] [CrossRef]
  71. Lee, K.H.; Kronbichler, A.; Park, D.D.Y.; Park, Y.; Moon, H.; Kim, H.; Choi, J.H.; Choi, Y.; Shim, S.; Lyu, I.S.; et al. Neutrophil Extracellular Traps (NETs) in Autoimmune Diseases: A Comprehensive Review. Autoimmun. Rev. 2017, 16, 1160–1173. [Google Scholar] [CrossRef] [PubMed]
  72. Hedstroem, A.K.; Roennelid, 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]
  73. Ahmad, S.; Zafar, I.; Mariappan, N.; Husain, M.; Wei, C.-C.; Vetal, N.; Eltoum, I.A.; Ahmad, A. Acute pulmonary effects of aerosolized nicotine. Am. J. Physiol. Lung Cell Mol. Physiol. 2019, 316, L94–L104. [Google Scholar] [CrossRef] [PubMed]
  74. Miyake, Y.; Sasaki, S.; Yokoyama, T.; Chida, K.; Azuma, A.; Suda, T.; Kudoh, S.; Sakamoto, N.; Okamoto, K.; Kobashi, G.; et al. Occupational and environmental factors and idiopathic pulmonary fibrosis in Japan. Ann. Occup. Hyg. 2005, 49, 259–265. [Google Scholar] [PubMed]
  75. Saag, K.G.; Kolluri, S.; Koehnke, R.K.; Georgou, T.A.; Rachow, J.W.; Hunninghake, G.W.; Schwartz, D.A. Rheumatoid arthritis lung disease. Determinants of radiographic and physiologic abnormalities. Arthritis Rheum. 1996, 39, 1711–1719. [Google Scholar] [CrossRef] [PubMed]
  76. Baumgartner, K.B.; Samet, J.M.; Stidley, C.A.; Colby, T.V.; Waldron, J.A. Cigarette smoking: A risk factor for idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 1997, 155, 242–248. [Google Scholar] [CrossRef] [PubMed]
  77. Yahya, A.; Bengtsson, C.; Larsson, P.; Too, C.L.; Mustafa, A.N.; Abdullah, N.A.; Muhamad, N.A.; Klareskog, L.; Murad, S.; Alfredsson, L. Silica exposure is associated with an increased risk of developing ACPA-positive rheumatoid arthritis in an Asian population: Evidence from the Malaysian MyEIRA case–control study. Mod. Rheumatol. 2014, 24, 271–274. [Google Scholar] [CrossRef] [PubMed]
  78. Stolt, P.; Yahya, A.; Bengtsson, C.; Källberg, H.; Rönnelid, J.; Lundberg, I.; Klareskog, L.; Alfredsson, L.; Group, E.S. Silica exposure among male current smokers is associated with a high risk of developing ACPA-positive rheumatoid arthritis. Ann. Rheum. Dis. 2010, 69, 1072–1076. [Google Scholar] [CrossRef] [PubMed]
  79. Stolt, P.; Källberg, H.; Lundberg, I.; Sjögren, B.; Klareskog, L.; Alfredsson, L. Silica exposure is associated with increased risk of developing rheumatoid arthritis: Results from the Swedish EIRA study. Ann. Rheum. Dis. 2005, 64, 582–586. [Google Scholar] [CrossRef]
  80. Zhao, N.; Al-Aly, Z.; Zheng, B.; van Donkelaar, A.; Martin, R.V.; Pineau, C.A.; Bernatsky, S. Fine particulate matter components and interstitial lung disease in rheumatoid arthritis. Eur. Respir. J. 2022, 60, 2102149. [Google Scholar] [CrossRef]
  81. Liu, B.; Sun, G.; Liu, Y.; Hou, Y. Observational studies: Ambient air pollution and hospitalization for RA-ILD in a heavily polluted city in China. Medicine 2022, 101, e29309. [Google Scholar] [CrossRef] [PubMed]
  82. Aletaha, D.; Smolen, J.S. Diagnosis and management of rheumatoid arthritis: A review. JAMA 2018, 320, 1360–1372. [Google Scholar] [CrossRef] [PubMed]
  83. Solomon, D.; Reed, G.; Kremer, J.; Curtis, J.; Farkouh, M.; Harrold, L.; Hochberg, M.; Tsao, P.; Greenberg, J. Disease activity in rheumatoid arthritis and the risk of cardiovascular events. Arthritis Rheumatol. 2015, 67, 1449–1455. [Google Scholar] [CrossRef] [PubMed]
  84. Sparks, J.A.; He, X.; Huang, J.; Fletcher, E.A.; Zaccardelli, A.; Friedlander, H.M.; Gill, R.R.; Hatabu, H.; Nishino, M.; Murphy, D.J. Rheumatoid arthritis disease activity predicting incident clinically apparent rheumatoid arthritis–associated interstitial lung disease: A prospective cohort study. Arthritis Rheumatol. 2019, 71, 1472–1482. [Google Scholar] [CrossRef] [PubMed]
  85. Paulin, F.; Doyle, T.J.; Mercado, J.F.; Fassola, L.; Fernández, M.; Caro, F.; Alberti, M.L.; Espíndola, M.E.C.; Buschiazzo, E. Development of a risk indicator score for the identification of interstitial lung disease in patients with rheumatoid arthritis. Reumatol. Clin. 2021, 17, 207–211. [Google Scholar] [CrossRef] [PubMed]
  86. Zhuo, J.; Lama, S.; Knapp, K.; Gutierrez, C.; Lovett, K.; Thai, S.; Craig, G.L. Epidemiology and clinical characteristics of interstitial lung disease in patients with rheumatoid arthritis from the JointMan database. Sci. Rep. 2023, 13, 11678. [Google Scholar] [CrossRef] [PubMed]
  87. Zhou, W.; Zheng, J.; Yuan, M.; Yuan, L.; Jia, X.; Liu, H. Differentially expressed lncRNAs in peripheral blood mononuclear cells from middle-aged female patients with rheumatoid arthritis–associated interstitial lung disease. Clin. Rheumatol. 2020, 39, 2281–2289. [Google Scholar] [CrossRef] [PubMed]
  88. Severo, C.R.; Chomiski, C.; Valle, M.B.d.; Escuissato, D.L.; Paiva, E.d.S.; Storrer, K.M. Assessment of risk factors in patients with rheumatoid arthritis-associated interstitial lung disease. J. Bras. Pneumol. 2022, 48, e20220145. [Google Scholar] [PubMed]
  89. Rojas-Serrano, J.; Mejía, M.; Rivera-Matias, P.A.; Herrera-Bringas, D.; Pérez-Román, D.I.; Pérez-Dorame, R.; Mateos-Toledo, H. Rheumatoid arthritis-related interstitial lung disease (RA-ILD): A possible association between disease activity and prognosis. Clin. Rheumatol. 2022, 41, 1741–1747. [Google Scholar] [CrossRef]
  90. Florescu, A.; Gherghina, F.L.; Mușetescu, A.E.; Pădureanu, V.; Roșu, A.; Florescu, M.M.; Criveanu, C.; Florescu, L.-M.; Bobircă, A. Novel Biomarkers, Diagnostic and Therapeutic Approach in Rheumatoid Arthritis Interstitial Lung Disease—A Narrative Review. Biomedicines 2022, 10, 1367. [Google Scholar] [CrossRef]
  91. Demoruelle, M.K.; Solomon, J.J.; Fischer, A.; Deane, K.D. The lung may play a role in the pathogenesis of rheumatoid arthritis. Int. J. Clin. Rheumatol. 2014, 9, 295. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, J.; Wright, K.; Davis, J.M.; Jeraldo, P.; Marietta, E.V.; Murray, J.; Nelson, H.; Matteson, E.L.; Taneja, V. An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med. 2016, 8, 43. [Google Scholar] [CrossRef] [PubMed]
  93. Scher, J.U.; Joshua, V.; Artacho, A.; Abdollahi-Roodsaz, S.; Öckinger, J.; Kullberg, S.; Sköld, M.; Eklund, A.; Grunewald, J.; Clemente, J.C.; et al. The lung microbiota in early rheumatoid arthritis and autoimmunity. Microbiome 2016, 4, 60. [Google Scholar] [CrossRef] [PubMed]
  94. Mikuls, T.R.; Payne, J.B.; Deane, K.D.; Thiele, G.M. Autoimmunity of the lung and oral mucosa in a multisystem inflammatory disease: The spark that lights the fire in rheumatoid arthritis? J. Allergy Clin. Immunol. 2016, 137, 28–34. [Google Scholar] [CrossRef] [PubMed]
  95. Drakopanagiotakis, F.; Stavropoulou, E.; Tsigalou, C.; Nena, E.; Steiropoulos, P. The Role of the Microbiome in Connective-Tissue-Associated Interstitial Lung Disease and PulmonaryVasculitis. Biomedicines 2022, 10, 3195. [Google Scholar] [CrossRef] [PubMed]
  96. Kronzer, V.L.; Davis, J.M. Etiologies of rheumatoid arthritis: Update on mucosal, genetic, and cellular pathogenesis. Curr. Rheumatol. 2021, 23, 21. [Google Scholar] [CrossRef] [PubMed]
  97. Demoruelle, M.K.; Weisman, M.H.; Simonian, P.L.; Lynch, D.A.; Sachs, P.B.; Pedraza, I.F.; Harrington, A.R.; Kolfenbach, J.R.; Striebich, C.C.; Pham, Q.N.; et al. Brief report: Airways abnormalities and rheumatoid arthritis-related autoantibodies in subjects without arthritis: Early injury or initiating site of autoimmunity? Arthritis Rheum. 2012, 64, 1756–1761. [Google Scholar] [CrossRef] [PubMed]
  98. Fischer, A.; Solomon, J.J.; Du Bois, R.M.; Deane, K.D.; Olson, A.L.; Fernandez-Perez, E.R.; Huie, T.J.; Stevens, A.D.; Gill, M.B.; Rabinovitch, A.M.; et al. Lung disease with anti-CCP antibodies but not rheumatoid arthritis or connective tissue disease. Respir. Med. 2012, 106, 1040–1047. [Google Scholar] [CrossRef] [PubMed]
  99. Proal, A.D.; Albert, P.J.; Marshall, T.G. The human microbiome and autoimmunity. Curr. Opin. Rhumatol. 2013, 25, 234–240. [Google Scholar] [CrossRef]
  100. Yeoh, N.; Burton, J.P.; Suppiah, P.; Reid, G.; Stebbings, S. The role of the microbiome in rheumatic diseases. Curr. Rheumatol. Rep. 2013, 15, 314. [Google Scholar] [CrossRef]
  101. Kharlamova, N.; Jiang, X.; Sherina, N.; Potempa, B.; Israelsson, L.; Quirke, A.M.; Eriksson, K.; Yucel-Lindberg, T.; Venables, P.J.; Potempa, J.; et al. Antibodies to porphyromonas gingivalis indicate interaction between oral infection, smoking, and risk genes in rheumatoid arthritis etiology. Arthritis Rheumatol. 2016, 68, 604–613. [Google Scholar] [CrossRef] [PubMed]
  102. Sieghart, D.; Platzer, A.; Studenic, P.; Alasti, F.; Grundhuber, M.; Swiniarski, S.; Horn, T.; Haslacher, H.; Blüml, S.; Smolen, J.; et al. Determination of autoantibody isotypes increases the sensitivity of serodiag- nostics in rheumatoid arthritis. Front. Immunol. 2018, 9, 876. [Google Scholar] [CrossRef] [PubMed]
  103. Van Delft, M.A.M.; Huizinga, T.W.J. An Overview of Autoantibodies in Rheumatoid Arthritis. J. Autoimmun. 2020, 110, 102392. [Google Scholar] [CrossRef] [PubMed]
  104. Kissel, T.; Reijm, S.; Slot, L.M.; Cavallari, M.; Wortel, C.M.; Vergroesen, R.D.; Stoeken-Rijsbergen, G.; Kwekkeboom, J.C.; Kampstra, A.S.B.; Levarht, E.W.N.; et al. Antibodies and B cells recognising citrullinated proteins display a broad cross-reactivity towards other post-translational modifications. Ann. Rheum. Dis. 2020, 79, 472–480. [Google Scholar] [CrossRef] [PubMed]
  105. Coutant, F. Pathogenic effects of anti-citrullinated protein antibodies in rheumatoid arthritis—Role for glycosylation. Jt. Bone Spine 2019, 86, 562–567. [Google Scholar] [CrossRef] [PubMed]
  106. Krishnamurthy, A.; Joshua, V.; Hensvold, A.H.; Jin, T.; Sun, M.; Vivar, N.; Ytterberg, A.; Engström, M.; Fernandes-Cerqueira, C.; Amara, K.; et al. Identification of a novel chemokine-dependent molecular mechanism underlying rheumatoid arthritis-associated autoantibody-mediated bone loss. Ann. Rheum. Dis. 2015, 75, 721–729. [Google Scholar] [CrossRef] [PubMed]
  107. Van Kalsbeek, D.; Brooks, R.; Shaver, D.; Ebel, A.; Hershberger, D.; Schmidt, C.; Poole, J.A.; Ascherman, D.P.; Thiele, G.M.; Mikuls, T.R.; et al. Peripheral Blood Biomarkers for Rheumatoid Arthritis–Associated Interstitial Lung Disease: A Systematic Review. ACR Open Rheumatol. 2023, 5, 201–226. [Google Scholar] [CrossRef] [PubMed]
  108. Ge, C.; Holmdahl, R. The structure, specificity and function of anti-citrullinated protein antibodies. Nat. Rev. Rheumatol. 2019, 15, 503–508. [Google Scholar] [CrossRef] [PubMed]
  109. Rocha-Muñoz, A.D.; Ponce-Guarneros, M.; Gamez-Nava, J.I.; Olivas-Flores, E.M.; Mejía, M.; Juárez-Contreras, P.; Martínez-García, E.A.; Corona-Sánchez, E.G.; Rodríguez-Hernández, T.M.; Vázquez-del Mercado, M.; et al. Anti-cyclic citrullinated peptide antibodies and severity of interstitial lung disease in women with rheumatoid arthritis. J. Immunol. Res. 2015, 2015, 151626. [Google Scholar] [CrossRef] [PubMed]
  110. Dai, Y.; Wang, W.; Yu, Y.; Hu, S. Rheumatoid arthritis–associated interstitial lung disease: An overview of epidemiology, pathogenesis and management. Clin. Rheumatol. 2021, 40, 1211–1220. [Google Scholar] [CrossRef]
  111. Correia, C.S.; Briones, M.R.; Guo, R.; Ostrowski, R.A. Elevated anti-cyclic citrullinated peptide antibody titer is associated with increased risk for interstitial lung disease. Clin. Rheumatol. 2019, 38, 1201–1206. [Google Scholar] [CrossRef]
  112. Giles, J.T.; Danoff, S.K.; Sokolove, J.; Wagner, C.A.; Winchester, R.; Pappas, D.A.; Siegelman, S.; Connors, G.; Robinson, W.H.; Bathon, J.M. Association of fine specificity and repertoire expansion of anticitrullinated peptide antibodies with rheumatoid arthritis associated interstitial lung disease. Ann. Rheum. Dis. 2014, 73, 1487–1494. [Google Scholar] [CrossRef] [PubMed]
  113. Zhu, J.; Zhou, Y.; Chen, X.; Li, J. A metaanalysis of the increased risk of rheumatoid arthritis-related pulmonary disease as a result of serum anticitrullinated protein antibody positivity. J. Rheumatol. 2014, 41, 1282–1289. [Google Scholar] [CrossRef]
  114. Aubart, F.; Crestani, B.; Nicaise-Roland, P.; Tubach, F.; Bollet, C.; Dawidowicz, K.; Quintin, E.; Hayem, G.; Palazzo, E.; Meyer, O. High levels of anti-cyclic citrullinated peptide autoantibodies are associated with co-occurrence of pulmonary diseases with rheumatoid arthritis. J. Rheumatol. 2011, 38, 979–982. [Google Scholar] [CrossRef]
  115. Kronzer, V.L.; Huang, W.; Dellaripa, P.F.; Huang, S.; Feathers, V.; Lu, B.; Iannaccone, C.K.; Gill, R.R.; Hatabu, H.; Nishino, M. Lifestyle and clinical risk factors for incident rheumatoid arthritis-associated interstitial lung disease. J. Rheumatol. 2021, 48, 656–663. [Google Scholar] [CrossRef]
  116. Wijsenbeek, M.; Suzuki, A.; Maher, T.M. Interstitial lung diseases. Lancet 2022, 400, 769–786. [Google Scholar] [CrossRef] [PubMed]
  117. Kelmenson, L.B.; Demoruelle, M.K.; Deane, K.D. The complex role of the lung in the pathogenesis and clinical outcomes of rheumatoid arthritis. Curr. Rheumatol. Rep. 2016, 18, 69. [Google Scholar] [CrossRef]
  118. Darrah, E.; Giles, J.T.; Davis, R.L.; Naik, P.; Wang, H.; Konig, M.F.; Cappelli, L.C.; Bingham, C.O., 3rd; Danoff, S.K.; Andrade, F. Autoantibodies to peptidylarginine Deiminase 2 are associated with less severe disease in rheumatoid arthritis. Front. Immunol. 2018, 9, 2696. [Google Scholar] [CrossRef] [PubMed]
  119. Giles, J.T.; Darrah, E.; Danoff, S.; Johnson, C.; Andrade, F.; Rosen, A.; Bathon, J.M. Association of cross-reactive antibodies targeting peptidyl-arginine deiminase 3 and 4 with rheumatoid arthritis-associated interstitial lung disease. PLoS ONE 2014, 9, e98794. [Google Scholar] [CrossRef]
  120. Kolarz, B.; Ciesla, M.; Rosenthal, A.K.; Dryglewska, M.; Majdan, M. The value of anti-car P and anti-PAD4 as markers of rheumatoid arthritis in ACPA/RF negative rheumatoid arthritis patients. Ther. Adv. Musculoskelet. Dis. 2021, 13, 1759720X2198986. [Google Scholar] [CrossRef]
  121. Nava-Quiroz, K.J.; Rojas-Serrano, J.; Pérez-Rubio, G.; Buendia-Roldan, I.; Mejía, M.; Fernández-López, J.C.; Rodríguez-Henríquez, P.; Ayala-Alcantar, N.; Ramos-Martínez, E.; López-Flore, L.A.; et al. Molecular factors in PAD2 (PADI2) and PAD4 (PADI4) are associated with interstitial lung disease susceptibility in rheumatoid arthritis patients. Cells 2023, 12, 2235. [Google Scholar] [CrossRef] [PubMed]
  122. Castellanos-Moreira, R.; Rodríguez-García, S.C.; Gomara, M.J.; Ruiz-Esquide, V.; Cuervo, A.; Casafont-Solé, I.; Ramírez, J.; Holgado, S.; Gómez-Puerta, J.A.; Cañete, J.D.; et al. Anti-carbamylated proteins antibody repertoire in rheumatoid arthritis: Evidence of a new autoantibody linked to interstitial lung disease. Ann. Rheum. Dis. 2020, 79, 587–594. [Google Scholar] [CrossRef] [PubMed]
  123. Janssen, K.M.; de Smit, M.J.; Brouwer, E.; de Kok, F.A.; Kraan, J.; Altenburg, J.; Verheul, M.K.; Trouw, L.A.; van Winkelhoff, A.J.; Vissink, A.; et al. Rheumatoid arthritis–associated autoantibodies in non–rheumatoid arthritis patients with mucosal inflammation: A case–control study. Arthritis Res. Ther. 2015, 17, 174. [Google Scholar] [CrossRef] [PubMed]
  124. Verheul, M.K.; van Erp, S.J.H.; van der Woude, D.; Levarht, E.W.; Mallat, M.J.; Verspaget, H.W.; Stolk, J.; Toes, R.E.; van der Meulen-de Jong, A.E.; Hiemstra, P.S.; et al. Anti-carbamylated protein antibodies: A specific hallmark for rheumatoid arthritis. Comparison to conditions known for enhanced carbamylation; renal failure, smoking and chronic inflammation. Ann. Rheum. Dis. 2016, 75, 1575–1576. [Google Scholar] [CrossRef] [PubMed]
  125. England, B.R.; Duryee, M.J.; Roul, P.; Mahajan, T.D.; Singh, N.; Poole, J.A.; Ascherman, D.P.; Caplan, L.; Demoruelle, M.K.; Deane, K.D.; et al. Malondialdehyde-acetaldehyde adducts and antibody responses in rheumatoid arthritis-associated interstitial lung disease. Arthritis Rheumatol. 2019, 71, 1483–1493. [Google Scholar] [CrossRef] [PubMed]
  126. Mohamed, A.; Enein, A.; Abdelsalam, N.; Balata, M.; Abdellatif, S.; Rizk, E.; Fathy, A. Utility of Anti-Carbamylated Protein Antibodies in the Diagnosis of Early Rheumatoid Arthritis. Indian J. Rheumatol. 2019, 14, 37–41. [Google Scholar] [CrossRef]
  127. Nakabo, S. Clinical and Etiological Meaning of Anti-Carbamylated Protein Antibodies in Rheumatoid Arthritis. Immunol. Med. 2018, 41, 147–153. [Google Scholar] [CrossRef] [PubMed]
  128. Wu, C.Y.; Yang, H.Y.; Luo, S.F.; Lai, J.H. From Rheumatoid Factor to Anti-Citrullinated Protein Antibodies and Anti-Carbamylated Protein Antibodies for Diagnosis and Prognosis Prediction in Patients with Rheumatoid Arthritis. Int. J. Mol. Sci. 2021, 22, 686. [Google Scholar] [CrossRef] [PubMed]
  129. Tseng, C.C.; Sung, Y.W.; Chen, K.Y.; Wang, P.Y.; Yen, C.Y.; Sung, W.Y.; Wu, C.C.; Ou, T.T.; Tsai, W.C.; Liao, W.T.; et al. The Role of Macrophages in Connective Tissue Disease-Associated Interstitial Lung Disease: Focusing on Molecular Mechanisms and Potential Treatment Strategies. Int. J. Mol. Sci. 2023, 24, 11995. [Google Scholar] [CrossRef]
  130. Kulshrestha, R.; Dhanda, H.; Pandey, A.; Singh, A.; Kumar, R. Immunopathogenesis and therapeutic potential of macrophage influx in diffuse parenchymal lung diseases. Expert Rev. Respir. Med. 2020, 14, 917–928. [Google Scholar] [CrossRef]
  131. Tanguy, J.; Pommerolle, L.; Garrido, C.; Kolb, M.; Bonniaud, P.; Goirand, F.; Bellaye, P.S. Extracellular heat shock proteins as therapeutic targets and biomarkers in fibrosing interstitial lung diseases. Int. J. Mol. Sci. 2021, 22, 9316. [Google Scholar] [CrossRef] [PubMed]
  132. Matsuda, S.; Kotani, T.; Kuwabara, H.; Suzuka, T.; Kiboshi, T.; Wada, Y.; Ishida, T.; Fujiki, Y.; Shiba, H.; Hata, K.; et al. Association of M2 macrophages, Th2, and B cells with pathomechanism in microscopic polyangiitis complicated by interstitial lung disease. J. Rheumatol. 2022, 49, 913–921. [Google Scholar] [CrossRef] [PubMed]
  133. McCubbrey, A.L.; Barthel, L.; Mohning, M.P.; Redente, E.F.; Mould, K.J.; Thomas, S.M.; Leach, S.M.; Danhorn, T.; Gibbings, S.L.; Jakubzick, C.V.; et al. Deletion of c-FLIP from CD11b(hi) Macrophages Prevents Development of Bleomycin-induced Lung Fibrosis. Am. J. Respir. Cell Mol. Biol. 2018, 58, 66–78. [Google Scholar] [CrossRef] [PubMed]
  134. Byrne, A.J.; Maher, T.M.; Lloyd, C.M. Pulmonary Macrophages: A New Therapeutic Pathway in Fibrosing Lung Disease? Trends Mol. Med. 2016, 22, 303–316. [Google Scholar] [CrossRef] [PubMed]
  135. Chen, Z.; Bozec, A.; Ramming, A.; Schett, G. Anti-inflammatory and immune-regulatory cytokines in rheumatoid arthritis. Nat. Rev. Rheumatol. 2018, 15, 9–17. [Google Scholar] [CrossRef] [PubMed]
  136. Adams, T.S.; Schupp, J.C.; Poli, S.; Ayaub, E.A.; Neumark, N.; Ahangari, F.; Chu, S.G.; Raby, B.A.; DeIuliis, G.; Januszyk, M.; et al. Single-cell RNA-seq reveals ectopic and aberrant lung-resident cell populations in idiopathic pulmonary fibrosis. Sci. Adv. 2020, 6, eaba1983. [Google Scholar] [CrossRef]
  137. Yokoyama, Y.; Iwasaki, T.; Kitano, S.; Satake, A.; Nomura, S.; Furukawa, T.; Matsui, K.; Sano, H. IL-2-Anti-IL-2 Mono-clonal antibody immune complexes inhibit collagen induced arthritis by augmenting regulatory T cell functions. J. Immunol. 2018, 201, 1899–1906. [Google Scholar] [CrossRef] [PubMed]
  138. Kalliolias, G.D.; Ivashkiv, L.B. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat. Rev. Rheumatol. 2016, 12, 49–62. [Google Scholar] [CrossRef] [PubMed]
  139. Tu, J.; Hong, W.; Guo, Y.; Zhang, P.; Fang, Y.; Wang, X.; Chen, X.; Lu, S.; Wei, W. Ontogeny of synovial macrophages and the roles of synovial macrophages from different origins in arthritis. Front. Immunol. 2019, 10, 1146. [Google Scholar] [CrossRef]
  140. Gabbiani, G.; Ryan, G.; Majno, G. Presence of modified fibroblasts in granulation tissue and their possible role in wound contraction. Experientia 1971, 27, 549–550. [Google Scholar] [CrossRef]
  141. Schafer, M.J.; White, T.A.; Iijima, K.; Haak, A.J.; Ligresti, G.; Atkinson, E.J.; Oberg, A.L.; Birch, J.; Salmonowicz, H.; Zhu, Y. Cellular senescence mediates fibrotic pulmonary disease. Nat. Commun. 2017, 8, 14532. [Google Scholar] [CrossRef] [PubMed]
  142. Hansel, C.; Jendrossek, V.; Klein, D. Cellular senescence in the lung: The central role of senescent epithelial cells. Int. J. Mol. Sci. 2020, 21, 3279. [Google Scholar] [CrossRef] [PubMed]
  143. Yang, J.; Antin, P.; Berx, G.; Blanpain, C.; Brabletz, T.; Bronner, M.; Campbell, K.; Cano, A.; Casanova, J.; Christofori, G. Guidelines and definitions for research on epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2020, 21, 341–352. [Google Scholar] [CrossRef] [PubMed]
  144. Marmai, C.; Sutherland, R.E.; Kim, K.K.; Dolganov, G.M.; Fang, X.; Kim, S.S.; Jiang, S.; Golden, J.A.; Hoopes, C.W.; Matthay, M.A. Alveolar epithelial cells express mesenchymal proteins in patients with idiopathic pulmonary fibrosis. Am. J. Physiol. Lung Cell Mol. Physiol. 2011, 301, L71–L78. [Google Scholar] [CrossRef] [PubMed]
  145. Coutant, F.; Miossec, P. Altered dendritic cell functions in autoimmune diseases: Distinct and overlapping profiles. Nat. Rev. Rheumatol. 2016, 12, 703–715. [Google Scholar] [CrossRef]
  146. Johnson, C. Recent Advances in the Pathogenesis, Prediction, and Management of Rheumatoid Arthritis-Associated Interstitial Lung Disease. Curr. Opin. Rheumatol. 2017, 29, 254–259. [Google Scholar] [CrossRef] [PubMed]
  147. Pruijn, G.J.M. Citrullination and Carbamylation in the Pathophysiology of Rheumatoid Arthritis. Front. Immunol. 2015, 6, 192. [Google Scholar] [CrossRef] [PubMed]
  148. Sharma, S.; Minhas, R.; Shankar, S.; Taha, O. Rheumatoid arthritis-associated interstitial lung disease. QJM 2019, 112, 815–816. [Google Scholar] [CrossRef] [PubMed]
  149. Douglas, D.; Keating, L.; Strykowski, R.; Lee, C.T.; Garcia, N.; Selvan, K.; Kaushik, N.; Bauer Ventura, I.; Jablonski, R.; Vij, R. Tobacco smoking is associated with combined pulmonary fibrosis and emphysema and worse outcomes in interstitial lung disease. Am. J. Physiol. Lung Cell. Mol. Physiol. 2023, 325, L233–L243. [Google Scholar] [CrossRef] [PubMed]
  150. Wells, A.U.; Denton, C.P. Interstitial lung disease in connective tissue disease--mechanisms and management. Nat. Rev. Rheumatol. 2014, 10, 728–739. [Google Scholar] [CrossRef]
  151. Kondo, N.; Kuroda, T.; Kobayashi, D. Cytokine networks in the pathogenesis of rheumatoid arthritis. Int. J. Mol. Sci. 2021, 22, 10922. [Google Scholar] [CrossRef]
  152. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [PubMed]
  153. Ciobanu, D.A.; Poenariu, I.S.; Crînguș, L.I.; Vreju, F.A.; Turcu-Stiolica, A.; Tica, A.A.; Padureanu, V.; Dumitrascu, R.M.; Banicioiu-Covei, S.; Dinescu, S.C.; et al. JAK/STAT pathway in pathology of rheumatoid arthritis. Exp. Ther. Med. 2020, 20, 498–3503. [Google Scholar] [CrossRef] [PubMed]
  154. Alam, J.; Jantan, I.; Bukhari, S.N.A. Rheumatoid arthritis: Recent advances on its etiology, role of cytokines and pharmacotherapy. Biomed. Pharmacother. 2017, 92, 615–633. [Google Scholar] [CrossRef] [PubMed]
  155. Györfi, A.H.; Matei, A.-E.; Distler, J.H. Targeting TGF-β signaling for the treatment of fibrosis. Matrix Biol. 2018, 68, 8–27. [Google Scholar] [CrossRef] [PubMed]
  156. Huang, Y.; Lin, W.; Chen, Z.; Wang, Y.; Huang, Y.; Tu, S. Effect of tumor necrosis factor inhibitors on interstitial lung disease in rheumatoid arthritis: Angel or demon? Drug Des. Dev. Ther. 2019, 1, 2111–2125. [Google Scholar] [CrossRef] [PubMed]
  157. Marahleh, A.; Kitaura, H.; Ohori, F.; Kishikawa, A.; Ogawa, S.; Shen, W.-R.; Qi, J.; Noguchi, T.; Nara, Y.; Mizoguchi, I. TNF-α Directly Enhances Osteocyte RANKL Expression and Promotes Osteoclast Formation. Front. Immunol. 2019, 10, 2925. [Google Scholar] [CrossRef] [PubMed]
  158. Mena-Vázquez, N.; Godoy-Navarrete, F.J.; Lisbona-Montañez, J.M.; Redondo-Rodriguez, R.; Manrique-Arija, S.; Rioja, J.; Mucientes, A.; Ruiz-Limón, P.; Garcia-Studer, A.; Ortiz-Márquez, F.; et al. Inflammatory Biomarkers in the Diagnosis and Prognosis of Rheumatoid Arthritis–Associated Interstitial Lung Disease. Int. J. Mol. Sci. 2023, 24, 6800. [Google Scholar] [CrossRef] [PubMed]
  159. Shen, H.; Xia, L.; Lu, J. Interleukin-4 in rheumatoid arthritis patients with interstitial lung disease: A pilot study. Indian J. Med. Res. 2013, 138, 919–921. [Google Scholar]
  160. Wang, X.; Zhu, G.; Ren, Q.; Wu, J.; Gu, B.; Su, D.; Shen, M. Increased interleukin-11 associated with disease activity and development of interstitial lung disease in patients with rheumatoid arthritis. Clin. Exp. Rheumatol. 2022, 40, 135–141. [Google Scholar] [CrossRef]
  161. Xiangyang, Z.; Lutian, Y.; Lin, Z.; Liping, X.; Hui, S.; Jing, L. Increased levels of interleukin-33 associated with bone erosion and interstitial lung diseases in patients with rheumatoid arthritis. Cytokine 2012, 58, 6–9. [Google Scholar] [CrossRef] [PubMed]
  162. Hussein, M.S.; El-Barbary, A.M.; Nada, D.W.; Gaber, R.A.; Elkolaly, R.M.; Aboelhawa, M.A. Identification of serum interleukin-13 and interleukin-13 receptor subunit expressions: Rheumatoid arthritis–associated interstitial lung disease. Int. J. Rheum. Dis. 2021, 24, 591–598. [Google Scholar] [CrossRef] [PubMed]
  163. Matsuo, T.; Hashimoto, M.; Ito, I.; Kubo, T.; Uozumi, R.; Furu, M.; Ito, H.; Fujii, T.; Tanaka, M.; Terao, C.; et al. Interleukin-18 is associated with the presence of interstitial lung disease in rheumatoid arthritis: A cross-sectional study. Scand. J. Rheumatol. 2019, 48, 87–94. [Google Scholar] [CrossRef] [PubMed]
  164. Zhang, J.; Wang, D.; Wang, L.; Wang, S.; Roden, A.C.; Zhao, H.; Li, X.; Prakash, Y.S.; Matteson, E.L.; Tschumperlin, D.J.; et al. Profibrotic effect of IL-17A and elevated IL-17RA in idiopathic pulmonary fibrosis and rheumatoid arthritis-associated lung disease support a direct role for IL-17A/IL-17RA in human fibrotic interstitial lung disease. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L487–L497. [Google Scholar] [CrossRef] [PubMed]
  165. Zhang, C.; Wang, S.; Lau, J.; Roden, A.C.; Matteson, E.L.; Sun, J.; Luo, F.; Tschumperlin, D.J.; Vassallo, R. IL-23 amplifies the epithelial-mesenchymal transition of mechanically conditioned alveolar epithelial cells in rheumatoid arthritis-associated interstitial lung disease through mTOR/S6 signaling. Am. J. Physiol. Lung Cell. Mol. Physiol. 2021, 321, L1006–L1022. [Google Scholar] [CrossRef] [PubMed]
  166. Lubberts, E. The IL-23-IL-17 axis in inflammatory arthritis. Nat. Rev. Rheumatol. 2015, 11, 415–429. [Google Scholar] [CrossRef] [PubMed]
  167. Robert, M.; Miossec, P. IL-17 in Rheumatoid Arthritis and Precision Medicine: From Synovitis Expression to Circulating Bioactive Levels. Front. Med. 2019, 5, 364. [Google Scholar] [CrossRef]
  168. Schinocca, C.; Rizzo, C.; Fasano, S.; Grasso, G.; Barbera, L.L.; Ciccia, F.; Guggino, G. Role of IL-23/IL-17 pathway in rheumatic diseases: An overview. Front. Immunol. 2021, 12, 637829. [Google Scholar] [CrossRef] [PubMed]
  169. Kolahian, S.; Fernandez, I.E.; Eickelberg, O.; Hartl, D. Immune mecha- nisms in pulmonary fibrosis. Am. J. Respir. Cell. Mol. Biol. 2016, 55, 309–322. [Google Scholar] [CrossRef]
  170. Przybylo, J.A.; Radisky, D.C. Matrix metalloproteinase-induced epithelial–mesenchymal transition: Tumor progression at Snail’s pace. Int. J. Biochem. Cell Biol. 2007, 39, 1082–1088. [Google Scholar] [CrossRef]
  171. Yamashita, C.M.; Dolgonos, L.; Zemans, R.L.; Young, S.K.; Robertson, J.; Briones, N.; Suzuki, T.; Campbell, M.N.; Gauldie, J.; Radisky, D.C. Matrix metalloproteinase 3 is a mediator of pulmonary fibrosis. Am. J. Pathol. 2011, 179, 1733–1745. [Google Scholar] [CrossRef] [PubMed]
  172. Xue, J.; Hu, W.; Wu, S.; Wang, J.; Chi, S.; Liu, X. Development of a risk nomogram model for identifying interstitial lung disease in patients with rheumatoid arthritis. Front. Immunol. 2022, 13, 823669. [Google Scholar] [CrossRef]
  173. Kollar, B.; Uffing, A.; Borges, T.J.; Shubin, A.V.; Aoyama, B.T.; Dagot, C.; Haug, V.; Kauke, M.; Safi, A.-F.; Talbot, S.G. MMP3 is a non-invasive biomarker of rejection in skin-bearing vascularized composite allotransplantation: A multicenter validation study. Front. Immunol. 2019, 10, 2771. [Google Scholar] [CrossRef] [PubMed]
  174. Chen, J.; Doyle, T.J.; Liu, Y.; Aggarwal, R.; Wang, X.; Shi, Y.; Ge, S.X.; Huang, H.; Lin, Q.; Liu, W. Biomarkers of rheumatoid arthritis–associated interstitial lung disease. Arthritis Rheumatol. 2015, 67, 28–38. [Google Scholar] [CrossRef]
  175. Doyle, T.J.; Lee, J.S.; Dellaripa, P.F.; Lederer, J.A.; Matteson, E.L.; Fischer, A.; Ascherman, D.P.; Glassberg, M.K.; Ryu, J.H.; Danoff, S.K. A roadmap to promote clinical and translational research in rheumatoid arthritis-associated interstitial lung disease. Chest 2014, 145, 454–463. [Google Scholar] [CrossRef] [PubMed]
  176. Mohan, C.; Assassi, S. Biomarkers in Rheumatic Diseases: How Can They Facilitate Diagnosis and Assessment of Disease Activity? BMJ 2015, 351, h5079. [Google Scholar] [CrossRef]
  177. Kass, D.J.; Nouraie, M.; Glassberg, M.K.; Ramreddy, N.; Fernandez, K.; Harlow, L.; Zhang, Y.; Chen, J.; Kerr, G.S.; Reimold, A.M.; et al. Comparative Profiling of Serum Protein Biomarkers in Rheumatoid Arthritis–Associated Interstitial Lung Disease and Idiopathic Pulmonary Fibrosis. Arthritis Rheumatol. 2020, 72, 409–419. [Google Scholar] [CrossRef]
  178. Mahalanobish, S.; Saha, S.; Dutta, S.; Sil, P.C. Matrix Metalloproteinase: An Upcoming Therapeutic Approach for Idiopathic Pulmonary Fibrosis. Pharmacol. Res. 2020, 152, 104591. [Google Scholar] [CrossRef]
  179. Ma, X.; Chen, R.; Liu, X.; Xie, J.; Si, K.; Duan, L. Effects of matrine on JAK-STAT signaling transduction pathways in bleomycin-induced pulmonary fibrosis. Afr. J. Tradit. Complement. Altern. Med. 2013, 10, 442–448. [Google Scholar] [CrossRef]
  180. Simon, L.S.; Taylor, P.C.; Choy, E.H.; Sebba, A.; Quebe, A.; Knopp, K.L.; Porreca, F. The Jak/STAT pathway: A focus on pain in rheumatoid arthritis. Semin. Arthritis Rheum. 2021, 51, 278–284. [Google Scholar] [CrossRef]
  181. Montero, P.; Milara, J.; Roger, I.; Cortijo, J. Role of JAK/STAT in interstitial lung diseases; molecular and cellular mechanisms. Int. J. Mol. Sci. 2021, 22, 6211. [Google Scholar] [CrossRef] [PubMed]
  182. Lee, J.S.; Lee, E.Y.; Ha, Y.J.; Kang, E.H.; Lee, Y.J.; Song, Y.W. Serum Kl-6 Levels Reflect the Severity of Interstitial Lung Disease Associated with Connective Tissue Disease. Arthritis Res. Ther. 2019, 21, 58. [Google Scholar] [CrossRef] [PubMed]
  183. Oguz, E.O.; Kucuksahin, O.; Turgay, M.; Yildizgoren, M.T.; Ates, A.; Demir, N.; Kumbasar, O.O.; Kinikli, G.; Duzgun, N. Association of Serum KL-6 Levels with Interstitial Lung Disease in Patients with Connective Tissue Disease: A Cross-Sectional Study. Clin. Rheumatol. 2016, 35, 663–666. [Google Scholar] [CrossRef] [PubMed]
  184. Kim, H.C.; Choi, K.H.; Jacob, J.; Song, J.W. Prognostic role of blood KL-6 in rheumatoid arthritis–associated interstitial lung disease. PLoS ONE 2020, 15, e0229997. [Google Scholar] [CrossRef]
Figure 1. Risk factors associated with RA-ILD. SO2—sulfur dioxide, NO2—nitrogen dioxide, Si—silica, NH4—ammonium, PAHs—polycyclic aromatic hydrocarbons, DAS-28—disease activity score in 28 joints, CDAI—clinical disease activity index.
Figure 1. Risk factors associated with RA-ILD. SO2—sulfur dioxide, NO2—nitrogen dioxide, Si—silica, NH4—ammonium, PAHs—polycyclic aromatic hydrocarbons, DAS-28—disease activity score in 28 joints, CDAI—clinical disease activity index.
Ijms 25 06460 g001
Figure 2. The role of macrophages, neutrophils, fibroblasts, dendritic cells, and T and B cells in the pathogenesis of RA-ILD. MMP—matrix metalloproteinase, TGFβ—transforming growth factor beta, IL—interleukin, GF—growth factor, TNF-α—tumor necrosis factor alpha, RoS—reactive oxygen species, NET—neutrophil extracellular trap, MHC II—major histocompatibility complex class II, DC—dendritic cells, LyT—lymphocyte T, LyB—lymphocyte B.
Figure 2. The role of macrophages, neutrophils, fibroblasts, dendritic cells, and T and B cells in the pathogenesis of RA-ILD. MMP—matrix metalloproteinase, TGFβ—transforming growth factor beta, IL—interleukin, GF—growth factor, TNF-α—tumor necrosis factor alpha, RoS—reactive oxygen species, NET—neutrophil extracellular trap, MHC II—major histocompatibility complex class II, DC—dendritic cells, LyT—lymphocyte T, LyB—lymphocyte B.
Ijms 25 06460 g002
Figure 3. The role of IL-17 and TNF-α in RA-ILD. IL—interleukin, MMP—matrix metalloproteinase, PDGF-β—platelet-derived growth factor beta, TGF-β—transforming growth factor beta, RANK-L—receptor activator of nuclear factor KB ligand, ↑—increased.
Figure 3. The role of IL-17 and TNF-α in RA-ILD. IL—interleukin, MMP—matrix metalloproteinase, PDGF-β—platelet-derived growth factor beta, TGF-β—transforming growth factor beta, RANK-L—receptor activator of nuclear factor KB ligand, ↑—increased.
Ijms 25 06460 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Anton, M.-L.; Cardoneanu, A.; Burlui, A.M.; Mihai, I.R.; Richter, P.; Bratoiu, I.; Macovei, L.A.; Rezus, E. The Lung in Rheumatoid Arthritis—Friend or Enemy? Int. J. Mol. Sci. 2024, 25, 6460. https://doi.org/10.3390/ijms25126460

AMA Style

Anton M-L, Cardoneanu A, Burlui AM, Mihai IR, Richter P, Bratoiu I, Macovei LA, Rezus E. The Lung in Rheumatoid Arthritis—Friend or Enemy? International Journal of Molecular Sciences. 2024; 25(12):6460. https://doi.org/10.3390/ijms25126460

Chicago/Turabian Style

Anton, Maria-Luciana, Anca Cardoneanu, Alexandra Maria Burlui, Ioana Ruxandra Mihai, Patricia Richter, Ioana Bratoiu, Luana Andreea Macovei, and Elena Rezus. 2024. "The Lung in Rheumatoid Arthritis—Friend or Enemy?" International Journal of Molecular Sciences 25, no. 12: 6460. https://doi.org/10.3390/ijms25126460

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