Next Article in Journal
BaGdF5 Nanophosphors Doped with Different Concentrations of Eu3+ for Application in X-ray Photodynamic Therapy
Next Article in Special Issue
Adult-Onset Still’s Disease: Novel Biomarkers of Specific Subsets, Disease Activity, and Relapsing Forms
Previous Article in Journal
Developmental Stressors Induce Innate Immune Memory in Microglia and Contribute to Disease Risk
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Update on the Pathogenic Role of Neutrophils in Systemic Juvenile Idiopathic Arthritis and Adult-Onset Still’s Disease

Department of Rheumatology, Ajou University School of Medicine, Suwon 16499, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(23), 13038; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222313038
Submission received: 17 November 2021 / Revised: 30 November 2021 / Accepted: 30 November 2021 / Published: 2 December 2021

Abstract

:
Neutrophils are innate immune phagocytes that play a key role in immune defense against invading pathogens. The main offensive mechanisms of neutrophils are the phagocytosis of pathogens, release of granules, and production of cytokines. The formation of neutrophil extracellular traps (NETs) has been described as a novel defense mechanism in the literature. NETs are a network of fibers assembled from chromatin deoxyribonucleic acid, histones, and neutrophil granule proteins that have the ability to kill pathogens, while they can also cause toxic effects in hosts. Activated neutrophils with NET formation stimulate autoimmune responses related to a wide range of inflammatory autoimmune diseases by exposing autoantigens in susceptible individuals. The association between increased NET formation and autoimmunity was first reported in antineutrophil cytoplasmic antibody-related vasculitis, and the role of NETs in various diseases, including systemic lupus erythematosus, rheumatoid arthritis, and psoriasis, has since been elucidated in research. Herein, we discuss the mechanistic role of neutrophils, including NETs, in the pathogenesis of systemic juvenile idiopathic arthritis (SJIA) and adult-onset Still’s disease (AOSD), and provide their clinical values as biomarkers for monitoring and prognosis.

1. Introduction

Neutrophils are innate immune phagocytes with a short lifespan that arise from hematopoietic stem cells that account for 40% to 70% of circulating white blood cells in humans [1]. As the first line of defense in the innate immune system, they play a key role in eliminating invading pathogens [2]. The main offensive mechanisms of neutrophils are phagocytosis, release of granules, and production of reactive oxygen species (ROS). Furthermore, the formation of neutrophil extracellular traps (NETs) has been described as a novel defense mechanism in the literature [3]. NETs are a network of fibers assembled from chromatin deoxyribonucleic acid (DNA), histones, serine proteases, cytoskeletal proteins, and antimicrobial peptides, which are formed and released from the extracellular space during the elimination of pathogens that have entered the body [4,5,6]. With the discovery of NETs, shifting the perception that neutrophils are far more heterogeneous than initially estimated, it is becoming clear that these cells have functional aspects beyond mere immune defense [7]. In addition to NET formation, neutrophils exhibit a wide range of phenotypes according to nuclear morphology, migratory and phagocytic capacity, and immunomodulatory functions, reflecting variability from the developmental stage to the activation status. The heterogeneity of the rapidly changing phenotypic and functional properties of neutrophils under pathophysiological conditions represents an opportunity to intervene in a variety of diseases, including vascular and immune-related diseases [8].
Currently, increasing evidence shows that neutrophil heterogeneity and NETosis could be strong inducers of autoimmunity [9]. In terms of neutrophil heterogeneity, one possible explanation might be that putative pathogenic neutrophil subsets are a key aspect in the development of autoimmune responses and the most widely studied in the context of autoimmunity is low-density granulocytes (LDGs) [10]. Moreover, the composition of NETs may vary depending on the stimulus, which means that there is a disease associated with NETs [11]. The association between increased disease-specific NETs and autoimmunity was first reported in antineutrophil cytoplasmic antibody-related vasculitis, and the role of NETs in various diseases, including systemic lupus erythematosus, rheumatoid arthritis (RA), and psoriasis, has since been elucidated in the literature [12,13,14,15]. Compared to studies on autoimmune diseases, relatively few studies have shown that activated neutrophils and NETosis contribute to pathogenicity in rare systemic autoinflammatory diseases such as systemic juvenile idiopathic arthritis (SJIA) and adult-onset Still’s disease (AOSD).
Autoinflammatory diseases are a group of rare diseases characterized by seemingly unexplained fever and inflammation affecting multiple organs, which belong to an area of clinical diseases other than autoimmune diseases [16]. Both SJIA and AOSD are autoinflammatory diseases with similar clinical features and laboratory findings, with a high spiking fever, skin rash, generalized lymphadenopathy, hepatosplenomegaly, and leukocytosis [17,18]. Since AOSD is known as the manifestation of SJIA in adults, SJIA and AOSD are two expressions of the same disease in different hosts [19]. Diagnosis of SJIA and AOSD is difficult because it is essential to rule out similar conditions such as infection or malignancy, and it uses criteria based on clinical evaluation without specific tests or laboratory results [20,21]. Several biomarkers, including acute phase reactants, inflammatory cytokines, and chemokines, are intended to be used for diagnosis in routine practice; however, their clinical utility is still unclear and nonspecific [22]. The sharing of several features implies that similar pathogenic mechanisms may underlie in the development of these two diseases. Unprovoked activation of the innate immune system and overproduction of proinflammatory cytokines play a pivotal role in disease pathogenesis, while the role of adaptive immunity is limited [23,24,25,26].
Despite thousands of reports available on SJIA and AOSD, our understanding of the exact pathogenesis is not yet complete. New research results on how neutrophil activation and NETosis cause amplification of inflammation in SJIA and AOSD are also gradually being provided, although the progress is slower than it is for other autoimmune and autoinflammatory disorders [27,28]. A better understanding of the role of neutrophils and NETs in the pathogenesis of SJIA and AOSD may lead to the discovery of new biomarkers. Herein, we discuss the mechanistic role of neutrophils and NETs in the development and progression of SJIA and AOSD, and provide their clinical values as biomarkers for monitoring and prognosis.

2. Neutrophils and NETs in Sterile Inflammation

Neutrophils are the first cells recruited at an inflammatory site during invasion of microorganisms or tissue damage, and they seem to orchestrate both the role of acting against pathogens and facilitating resolution of inflammation [29]. In the early stages of inflammation, the initiating stimulus is sensed by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), leading to the release of pro-inflammatory mediators, including lipid mediators such as prostaglandins, leukotriene B4, and complement anaphylatoxins C3a and C5a along with many cytokines and chemokines (e.g., tumor necrosis factor (TNF)-α, interleukin–1 (IL–1), and CXC chemokines) [30]. Multiple pro-inflammatory mediators act as potent chemoattractants to alert innate immunity and contribute to neutrophil recruitment [31].
Pro-inflammatory mediators attract monocytes and macrophages in addition to neutrophils to the site of inflammation to eliminate the stimulus [32]. Simultaneously, with the recruitment of these phagocytic cells, a switch in lipid mediator type occurs, resulting in the biosynthesis of pro-inflammatory lipid mediators into anti-inflammatory lipid mediators, such as lipoxins, resolvins, and protectins, to attenuate inflammation [33]. These novel mediators initiate the resolution of inflammation, in which neutrophils restrict further infiltration and undergo apoptosis, and this cellular debris generated during apoptosis is removed by macrophages [34]. Through this series of processes, neutrophils must be cleared before they cause some subsidiary damage to host tissues and unwanted inflammation in the inflamed tissues, which is essential for the maintenance of normal tissue homeostasis [35].
The traditional view of neutrophils as simple scavengers of pathogens has radically expanded to new perspectives, as the unrecognized complexity of neutrophils is uncovered in research [36]. Until 2004, when NETs were first reported, phagocytosis, ROS production, and protease activity were thought to be the major neutrophil effector mechanisms [37]. NETs have brought neutrophils back to the spotlight of immunological research, as they are at the forefront of new interest due to their broadly effective pathogen clearance. NETosis is a program for the formation of NETs, which begins with activation of neutrophils through the recognition of stimuli, and the process is divided into lytic NETosis and non-lytic NETosis. [38]. In lytic NETosis, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase is activated via the Raf-MEK-ERK signaling pathway, thereby generating ROS and activating protein-arginine deiminase type 4, inducing NET formation through chromatin decondensation [39]. NETosis ends with the release of chromatin from neutrophils along with associated neutrophil proteins, resulting in neutrophil cell death. It is also referred to as suicidal NETosis, and this process takes approximately 2–4 h [40]. Non-lytic NETosis, called vital NETosis, releases NETs within 5–60 min without cell death via Toll-like receptor (TLR) 2, TLR4, or complement receptors independent of NADPH oxidase activation [41].
Initially, neutrophils and NETs were considered to be involved in antimicrobials, while a number of studies had shown that they also played a pivotal role in the development of sterile inflammation under several chronic inflammatory conditions [42]. Sterile inflammation occurs in the absence of microorganisms, and it can be induced by physical, chemical, metabolic, or antigenic stimuli. When these non-infectious stimuli persist and cannot be eliminated, they promote sterile inflammation, becoming a part of the pathophysiology of numerous human diseases, such as metabolic disorders, autoimmune diseases, autoinflammatory diseases, and cancer [43,44,45,46]. Delayed neutrophil apoptosis or prolonged NET formation is the root cause of inflammatory disorders [47,48].

3. Neutrophils and NETs in Autoinflammatory Diseases

The term “autoinflammatory disease” was first coined in 1999 to represent an emerging family of clinical disorders that are different from autoimmune diseases [49]. Autoinflammatory and autoimmune diseases have similarities in that self-tissue-directed inflammation occurs without obvious infectious triggers or injuries; however, the two categories can be distinguished by pathways of innate or adaptive immune responses [50]. Autoinflammatory diseases are characterized by episodes of recurrent unprovoked inflammation that cause aberrant activation of the innate immune system without autoantibodies or auto-reactive antigen-specific T cells [51]. While most of these have strong genetic factors with mutations in a single gene, there are also polygenic or multifactorial origins without identified genetic mutations, such as SJIA, AOSD, and inflammatory bowel disease (IBD). More than 30 new genes associated with autoinflammatory diseases have been identified since the mutation of Mediterranean fever (MEFV) gene was first discovered as the cause of familial Mediterranean fever (FMF) [52,53,54]. The important pathogenic mechanisms have been observed to be driven by mutations in genes involving inflammasome activation or cytokine pathways, including NOD-like receptor pyrin domain-containing protein 3, TNF receptor 1, and IL-1 receptor antagonist [55].
As neutrophils are the key cells explaining the development of autoinflammation, it has been significantly elucidated that NETs contribute to certain aspects of the pathogenesis of systemic autoinflammatory diseases. Several autoinflammatory diseases mediated by a sustained influx of neutrophils and NET release and their proposed roles are discussed accordingly in the literature.

3.1. Familial Mediterranean Fever

FMF is the most common hereditary autoinflammatory disorder, and it mainly affects individuals in the eastern Mediterranean region. Clinically, it is characterized by recurrent, self-limited episodes of fever caused by neutrophil-induced serosal inflammation, including synovitis, pleuritis, and pericarditis [56,57]. FMF is associated with mutations in the MEFV gene encoding the protein pyrin, which is highly expressed primarily in neutrophils. Neutrophils are the main effector cells of acute inflammatory attacks in patients with FMF, and disease attacks result in neutrophilia and a massive influx of neutrophils into the inflamed sites [58,59]. Mutated pyrin reduces autophagic function by impairing its role as a selective autophagy receptor targeting inflammasome components, leading to the release of IL–1β, a key cytokine in this disease [60,61,62]. Recent experimental evidence suggests that neutrophils release large amounts of NETs in an autophagy-dependent manner during FMF inflammation. These NETs are transmitted to extracellular space bioactive IL–1β, which amplifies IL–1β release during the first 24 h of acute inflammation attacks. At the same time, FMF inflammation is suppressed by a negative feedback mechanism of NETs that interferes with IL–1β activity by dismantling the NET chromatin scaffold by DNase. Compensatively, the autophagy of neutrophils is lowered, thereby attenuating the release of NETs and protecting them from crisis [63]. In addition, regulated in development and DNA damage response 1 (REDD1), a modulator of neutrophil function upstream of pyrin, is involved in the release of NETs and regulation of IL–1β, and it has emerged as a novel link in the mechanisms leading to FMF attacks [64].

3.2. Gout

Gout is classified as an autoinflammatory disease that is caused by monosodium urate (MSU) crystals, which act as DAMPs and can contribute to the initiation of acute joint inflammation by activating the innate immune system [65]. Uptake of MSU crystals through phagocytosis induces the activation of NLRP3 inflammasome and release of IL–1β, which not only promotes the recruitment of neutrophils and other immune cells, but also secretes more inflammatory mediators such as IL–6, IL–8, and TNF–α [66,67,68]. Early studies of gout and NETs revealed that NETs were induced in synovial cells and peripheral neutrophils during the acute phase of gouty inflammation to assemble pro-inflammatory responses, whereas recent studies have shown that aggregated NETs can inhibit inflammatory responses through the degradation of inflammatory cytokines and chemokines [69,70,71]. In this process, acute gout inflammation is naturally resolved within a few days, leaving the affected tissue with an abscess-like creamy lump containing MSU crystals, dead immune cells, and NETs, defined as tophi [72]. Although tophi formation is a mechanism to resolve inflammation, it accumulates in joints and parts of the body for long periods of time, contributing to chronic gout inflammation, leading to the destruction of surrounding tissue [71,73].

3.3. Inflammatory Bowel Diseases

Crohn’s disease (CD) and ulcerative colitis (UC), the most widely known IBD, are a group of relapsing and chronic diseases that cause damage to the gastrointestinal tract due to a dysregulated immune response to unknown stimuli [74]. Neutrophils, regarded as first responders to inflammation, are important entities that influence the gut mucosal inflammatory milieu in IBD [75]. In the early stages of intestinal inflammation in patients with IBD, neutrophils flow into the intestinal mucosa and recognize and phagocytose pathogens to heal the mucous membrane. If this process is not tightly regulated, a vicious cycle of self-amplification loop occurs in which a large number of neutrophils accumulate in the inflamed mucous membrane, producing excessive ROS and proinflammatory cytokines, thereby maintaining persistent intestinal inflammation and eventually impairing intestinal barrier function [76,77,78]. Several studies have demonstrated an increased presence of NETs in inflamed gut mucosa, stool, or blood in patients with IBD, and there have also been reports that the abundance of NETs is positively correlated with disease activity [79,80,81]. Among IBD, the response of neutrophils to autoinflammatory properties, such as REDD1, autophagy, and the NETosis pathway, is strongly featured, especially in UC. Studies have reported that ROS generation is enhanced in CD, whereas neutrophil migration is reduced, phagocytic functions are impaired, and evidence of NET formation is scarce therein [79,82,83]. This suggests that the activity of neutrophils is also different in the two diseases as seen by clinical symptoms, and the location and layer affected in the gut are different.

4. Neutrophils in SJIA and AOSD

Neutrophil activation, a hallmark of the pathogenesis of SJIA and AOSD, is responsible for the initiation and development of inflammation by releasing a wide variety of granular enzymes and antimicrobial proteins. In an acute flare of the disease, more than 80% of patients have neutrophilic leukocytosis, which can be a differentiating feature of SJIA and AOSD from other rheumatic diseases. Several lines of evidence indicate the crucial role of various activating cell surface receptors and intracellular signaling mechanisms expressed in neutrophils involved in the pathogenesis of SJIA and AOSD (Figure 1). Studies related to the role of neutrophils in SJIA and AOSD are summarized in Table 1.
Initially, levels of C–X–C motif chemokine ligand (CXCL)–8, known to be critical for neutrophil recruitment and activation, significantly increase during the inflammatory response associated with AOSD [84]. Similarly, Kasama et al. detected that CX3CL1, whose primary role is to promote neutrophil binding, adhesion, and activation of target cells, is positively correlated with disease activity of AOSD and that high CX3CL1 reflects the presence of hemophagocytic syndrome [85]. Another molecule facilitating adhesion and retention of neutrophils at the inflamed sites, intracellular adhesion molecule 1, and neutrophil activation marker CD64 (FcγR1) were upregulated in active untreated AOSD patients, and these were correlated with disease activity scores [86,87]. C–type lectin domain family 5–member A (CLEC5A), which combines with adapter proteins to form receptor complexes and which is involved in inflammatory responses through differentiation and activation of neutrophils, has been demonstrated to be overexpressed in AOSD patients. A positive correlation between CLEC5A levels and inflammatory parameters or disease activity of AOSD has also been reported in the literature [88]. The frequencies of circulating CD11b, the molecules of complement receptor 3, and CD32, most widely expressed in FcγR, were observed in whole blood cells of patients with AOSD than in patients with RA or healthy controls [89]. CD11b and CD32 are mainly expressed in neutrophils, suggesting that neutrophils contribute significantly to the onset of AOSD. Other receptors related to neutrophil activation, such as granulocyte-macrophage colony-stimulating factor receptor, are being mentioned as targets for new therapies for AOSD; however, evidence for their pathological roles in AOSD is still lacking [98].
LDGs, known as the key drivers of inflammation in various autoimmune diseases, cancer, and sepsis, are separated by density from normal density granulocytes [99]. LDGs with pro-inflammatory features were also found to be increased in active SJIA and AOSD. Increased IL-6 producing LDGs contributes to the pathophysiology of AOSD, and activation of neutrophils due to elevated transcription of genes encoding LDGs with pro-inflammatory features has potential implications for the mechanism of SJIA [90,91]. Additionally, evidence of functional and gene expression of neutrophil alterations has been reported in SJIA patients, and persistent proinflammatory activation in neutrophils has been shown not only in active disease but also in long-standing clinically inactive disease in SJIA [90,92].
Significantly higher levels of phagocyte-secreted pro-inflammatory cytokines IL–1, IL–6, and IL–18 in patients with SJIA and AOSD compared to all other inflammatory diseases can also support the role of neutrophils [93,94,95]. Moreover, a large amount of data on the elevation of various molecules expressed and secreted by neutrophils in response to inflammatory stimuli suggest the importance of neutrophils in the pathogenesis of SJIA and AOSD. Chemokines, including CXCL–9, 10, 11, 12, 13, and CXC receptor 4, were found in high concentrations in the serum of patients with SJIA and AOSD [94,96,97].

5. NETs in SJIA and AOSD

Recently, growing evidence has suggested that the release of NETs is explained by the mechanisms of SJIA and AOSD (Table 2). Figure 2 shows the mechanisms of NET involvement in SJIA and AOSD. The main protein components of NETs are histones, followed by granule-derived peptides and enzymes such as neutrophilic elastase, myeloperoxidase, calprotectin, cathepsin G, leukocyte proteinase 3, lysozyme C, and neutrophil defensins [100]. In SJIA and AOSD, the most frequently investigated field related to NETs is the S100 protein. The term calprotectin is described as a complex of two calcium-binding proteins of the S100 family of proteins, S100A8 and S100A9 [101]. In addition, S100A8/S100A9 complex, S100A8, S100A9, and S100A12 belonging to the S100 family are released from neutrophils as part of NETs and they function as DAMPs [102]. The S100 family of proteins, which are mainly derived from neutrophils and macrophages, are significantly higher in patients with SJIA and AOSD than in healthy controls. They act as ligands of TLR4 or receptor for advanced glycation end products (RAGE) to accelerate neutrophils and activate the release of pro-inflammatory cytokines, contributing to systemic inflammation of AOSD through enhancement of the feedback loop [103]. Serum levels of S100A8/A9 and S100A12 are significantly higher in patients with SJIA and AOSD, and they are strongly correlated with peripheral blood neutrophil levels [92,104,105,106]. Serum S100A8/A9 levels have been proven to have a positive correlation with inflammatory markers of AOSD, and they are associated with neutrophil infiltration in the skin and lymph nodes affected by AOSD. Serum S100A12 was also found to be strongly correlated with disease activity markers of SJIA and AOSD [105,106,107].
The levels of cell–free DNA, α–defensin, and MPO–DNA complex, which are remnants of NETs, have been found to increase significantly in the serum of patients with AOSD compared to healthy controls. These NET molecules were associated with skin rash and lymphadenopathy, indicating the induction of IL–1β production in monocytes as a pathogenesis of AOSD [27]. In another study, the levels of cell–free DNA and NET–DNA complexes were significantly increased in patients with AOSD compared with healthy controls, and NET formation accelerated pathogenic mechanisms through activation of NLRP3 inflammasomes and macrophages for pro-inflammatory cytokine release [28]. Moreover, urinary proteins enriched in neutrophil degranulation and neutrophil-derived lipocalin–2 were highly expressed in patients with AOSD [108,109].
A previous study demonstrated that IL–18, a pivotal cytokine of AOSD, induces NET by enhancing calcium influx into neutrophils. Another molecule involved in this process is microRNA–223, which modulates the immune response of the host. In this study, a fine-tuned mechanism was proposed in which IL–18, microRNA–223, and NET formation regulate inflammation in AOSD, in which NET formation inhibits calcium influx into neutrophils by upregulation of microRNA–223, and it eventually inhibits IL–18 mediated NET formation [110]. The role of LDGs and NET formation in AOSD was also identified, and it has been reported that the concentration of high mobility group box–1 protein (HMGB–1) and cathelicidin LL–37 in NETs was increased in patients with AOSD [111]. HMGB–1 is a nuclear protein released from various cells and is a DAMP that interacts with TLR4, TLR2, and RAGE to act as a major mediator of the immune response [118,119]. Serum HMGB–1 level was the most elevated at the time of diagnosis in patients with AOSD and SJIA, and most of the follow-up patients showed decreased levels after the disease showed an improvement. In patients with AOSD, serum HMGB–1 may be correlated with clinical disease activity, such as C–reactive protein (CRP), systemic score, skin rash, and sore throat [112,113,114]. LL–37 is released in the form of NETs via TLR 8/13 and formyl peptide receptor-like 1 in neutrophils, inducing the production of inflammatory cytokines, leading to the generation of interferon-α by dendritic cells. A positive loop was identified in which LL–37 activates the NLRP3 inflammasome and releases IL–1β and IL–18 through caspase-1, which stimulates NET release [120]. Differential expression of LL–37 was not found according to the disease activity status of AOSD [111]. In addition, circulating NETs have the potential to discriminate the involvement of the hepatic and cardiopulmonary systems among patients with AOSD, and they may help to provide therapeutic strategies by predicting their glucocorticoid response [115].
Recently, there has been a report of genetic susceptibility to neutrophil activation and NETosis in AOSD. Evidence has emerged that LILR3, a gene of the leukocyte immunoglobulin–like receptor (LIR)–A3, plays a pathogenic role in AOSD. Plasma levels of LIR–A3 were elevated in patients with LILRA3 +/+, and they correlated with disease activity measures and circulating NET–DNA complex levels [116]. Although various roles of NETs have been identified in AOSD, data on the role of NETs in SJIA are still limited. In one study, Hu et al. demonstrated that active SJIA patients release a large amount of histones from NETs and that induced-serum histones promote the propagation of NETs, resulting in malignant feedback mechanisms, which may be the pathogenesis of SJIA [117]. A recent study showed that secondary neutrophil granule proteins, such as human neutrophil lipocalin and myeloperoxidase, were significantly increased in patients with SJIA [108]. Currently, many studies are underway to elucidate the role of NETs in the pathogenesis of SJIA; thus, a clearer mechanism is expected to be identified in the future.

6. Clinical Values as Biomarkers Related to Neutrophils for Monitoring and Prognosis in SJIA and AOSD

Several studies have evaluated the role of cellular or serum markers related to neutrophils as biomarkers of SJIA and AOSD. A biomarker is defined as an objective measurement of a molecular or biological event, whose changes can reflect disease pathogenesis [121]. Therefore, several markers related to neutrophils and NETs could be good biomarkers for the diagnosis and evaluation of disease activities in SJIA and AOSD. In this section, we present a review of the potential use of markers related to neutrophils for the diagnosis, evaluation of disease activities, and prognosis in SJIA and AOSD patients.
Several proinflammatory cytokines and chemokines related to neutrophil recruitment or activation and NET have been evaluated in the serum of patients with SJIA and AOSD [18,95,96,97,122,123,124]. Significantly higher levels of IL–6, CXCL8, and IL–18 in sera or plasma were found in patients with active untreated AOSD or SJIA than in healthy controls [123,124,125]. Furthermore, the mRNA expression levels of IL–18 and CXCL8 were significantly higher in the biopsy tissue of Still’s rash and synovial membranes of AOSD patients compared with those in controls [124]. Interestingly, AOSD patients with chronic articular patterns had significantly higher levels of serum CXCL8 than those with a monocyclic systemic pattern [124]. Therefore, CXCL8 could be a good marker for predicting chronic articular patterns in AOSD. Several chemokines and receptors, including CXCL9, 10, 11, 12, 13, and CXCR3 and CXCR4, were found in high concentrations in the serum of patients with SJIA or AOSD, and these correlated with several disease activity markers of SJIA or AOSD [96,97,126,127]. In particular, the levels of CXCR9, CXCL10, CXCL11, and IL–18 were elevated in SJIA patients with macrophage activation syndrome compared with active SJIA without macrophage activation syndrome [125,126]. Therefore, interferon-γ-related chemokines and IL–18 could predict poor prognosis related to macrophage activation syndrome in patients with SJIA. A recent study aimed to identify biomarkers in a prospective cohort of patients with bloodstream infection and AOSD for differential diagnosis, and found that a combination of plasma IL–18 and ferritin levels could be used to distinguish bloodstream infections from AOSD (sensitivity: 96.15% and specificity: 100%) [128].
S100 proteins, such as S100A8/A9 and S100A12, have become reliable biomarkers for differential diagnosis, evaluation of current disease activity, and prediction of further disease flares in SJIA or AOSD [104,105,106,107]. Serum S100A8/A9 levels were elevated in active AOSD or SJIA, and they correlated with disease activity markers, such as CRP, ferritin, and systemic scores. S100 protein was also suggested to be a predictive marker of further disease flares during clinical remission in patients with SJIA [104,105,129]. Furthermore, S100A8/A9 was superior to CRP in differentiating systemic JIA from other fever syndromes, such as systemic undifferentiated recurring fever syndrome [130]. In a study with AOSD, the sensitivity and specificity of S100A8/A9 using receiver operating characteristic curves for the diagnosis of AOSD from other rheumatic diseases including RA, OA, and SLE was evaluated, and a sensitivity of 63% and a specificity of 80% were reported accordingly [131]. Studies on the role of S100A12 as a biomarker were similar to those on S100A8/A9 in SJIA and AOSD patients, with these levels being elevated in active SJIA or AOSD patients as compared to HC and associated with inflammatory markers, such as ESR, CRP, and ferritin, and the systemic score [106,107]. Furthermore, S100A12 levels were elevated in patients with SJIA or FMF compared to those in patients with other fever diseases, such as acute myeloblastic leukemia, acute lymphoblastic leukemia, or systemic infection, and their sensitivity and specificity to distinguish between SJIA and infections were 66% and 94%, respectively [106]. Another study showed that S100A12 levels were increased in patients with SJIA, compared to systemic undifferentiated recurring fever syndrome, other defined autoinflammatory syndromes, and non-systemic JIA. The area under the receiver operating characteristic curve (AUC = 0.9) of S100 proteins for differentiating SJIA from other fever syndromes was superior to that of CRP (AUC = 0.7) [130]. Therefore, these data suggest that serum S100 proteins, such as S100A8/A9 and S100A12, are good biomarkers for the differential diagnosis of systemic JIA and AOSD, and for the evaluation of disease activity.
Several recent studies showed elevated cell-free DNA and NET–related molecules in patients with active AOSD or SJIA, compared to those with inactive status or HC [27,28,63,109,117]. Furthermore, these levels, such as MPO–DNA and α–defensin, were correlated with the levels of disease activity markers of AOSD. In a recent study, patients with active SJIA had more extracellular histones released by activated neutrophils than those with inactive SJIA and HC. They also correlated significantly with clinical disease activity [117]. However, these studies did not evaluate the associations between NET molecules and the prognosis of AOSD or SJIA. Therefore, further studies are needed to evaluate the role of NET molecules as prognostic markers in patients with AOSD or SJIA.

7. Conclusions

This review describes the roles of neutrophils and their release of NETs, as DAMPs, that contribute to aggravation of inflammation in the pathogenesis of AOSD and SJIA. Although the initial trigger of inflammation in SJIA or AOSD is not well known, viral or bacterial infection and environmental trigger factors as danger signals could lead to the activation of neutrophils, leading to the transmigration into inflamed tissues and the production of proinflammatory cytokines and chemokines. Several specific receptors on neutrophils, including pattern-recognition receptors and Fc receptors interacting with danger signals, further increase neutrophil recruitment and activation. Neutrophil activation and mediator release form a positive-feedback loop that enhances neutrophil recruitment and amplifies inflammatory responses, which may act as an important axis of pathogenesis in AOSD and SJIA. In addition, activated neutrophils produce and release NETs, proinflammatory cytokines, and chemokines and activate and interact with macrophages in the inflamed tissue. Furthermore, several DAMPs, such as HMGB–1, S100 proteins, LL37, and MPO–DNA complex, are elevated in the blood of patients with SJIA or AOSD, accelerating systemic inflammation with proinflammatory cascades through several pattern recognition receptors of neutrophils.
Despite the continuous research conducted so far on this subject, undoubtedly, further research in this regard will provide greater clarity in understanding the pathogenesis of AOSD and SJIA. The first question is whether the initial danger signal for activation of neutrophils is truly provoked, or whether increased danger signaling is due to a loss of inhibitory signal in neutrophil activation during the initial inflammatory stages of AOSD. The second is whether neutrophil activation and NETs are associated with chronic articular forms of AOSD, while they are thought to contribute clearly to the initial acute inflammation of AOSD. The third is how much of the inflammation of SJIA and AOSD is caused by neutrophils, and how effective it would be when it is blocked. Ongoing research into the role of neutrophils and NETs in the pathogenesis of SJIA and AOSD will provide a better understanding of disease pathogenesis.
The current findings enhance our understanding of the pathogenesis of systemic JIA and AOSD, and they will facilitate the development of novel therapeutics targeting neutrophils and NET molecules and the discovery of biomarkers related to neutrophils that could be used to diagnose and predict prognosis in systemic JIA and AOSD.

Author Contributions

Conceptualization, J.-W.K. and H.-A.K.; methodology, J.-W.K. and H.-A.K.; resources, J.-W.K.; data curation, J.-W.K., M.-H.A., H.-A.K.; writing—original draft preparation, J.-W.K., H.-A.K.; writing—review and editing, J.-Y.J., M.-H.A., H.-A.K. and C.-H.S.; supervision, C.-H.S.; project administration, J.-W.K. and H.-A.K..; funding acquisition, H.-A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Education, Science, and Technology [grant number 2019R1A2C1005988].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All available data are reported in the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Silvestre-Roig, C.; Fridlender, Z.G.; Glogauer, M.; Scapini, P. Neutrophil diversity in health and disease. Trends Immunol. 2019, 40, 565–583. [Google Scholar] [CrossRef]
  2. Kobayashi, S.D.; Malachowa, N.; De Leo, F.R. Neutrophils and bacterial immune evasion. J. Innate Immun. 2018, 10, 432–441. [Google Scholar] [CrossRef] [PubMed]
  3. Fousert, E.; Toes, R.; Desai, J. Neutrophil extracellular traps (NETs) take the central stage in driving autoimmune responses. Cells 2020, 9, 915. [Google Scholar] [CrossRef] [Green Version]
  4. Dąbrowska, D.; Jabłońska, E.; Garley, M.; Ratajczak-Wrona, W.; Iwaniuk, A. New aspects of the biology of neutrophil extracellular traps. Scand. J. Immunol. 2016, 84, 317–322. [Google Scholar] [CrossRef]
  5. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef] [PubMed]
  6. Ravindran, M.; Khan, M.A.; Palaniyar, N. Neutrophil extracellular trap formation: Physiology, pathology, and pharmacology. Biomolecules 2019, 9. [Google Scholar] [CrossRef] [Green Version]
  7. Ng, L.G.; Ostuni, R.; Hidalgo, A. Heterogeneity of neutrophils. Nat. Rev. Immunol. 2019, 19, 255–265. [Google Scholar] [CrossRef]
  8. Grieshaber-Bouyer, R.; Nigrovic, P.A. Neutrophil heterogeneity as therapeutic opportunity in immune-mediated disease. Front. Immunol. 2019, 10, 346. [Google Scholar] [CrossRef] [Green Version]
  9. Granger, V.; Peyneau, M.; Chollet-Martin, S.; De Chaisemartin, L. Neutrophil extracellular traps in autoimmunity and allergy: Immune complexes at work. Front. Immunol. 2019, 10, 2824. [Google Scholar] [CrossRef] [Green Version]
  10. Nakazawa, D.; Kudo, T. Novel therapeutic strategy based on neutrophil subset and its function in autoimmune disease. Front. Pharmacol. 2021, 12, 684886. [Google Scholar] [CrossRef] [PubMed]
  11. Petretto, A.; Bruschi, M.; Pratesi, F.; Croia, C.; Candiano, G.; Ghiggeri, G.; Migliorini, P. Neutrophil extracellular traps (NET) induced by different stimuli: A comparative proteomic analysis. PLoS ONE 2019, 14, e0218946. [Google Scholar] [CrossRef] [PubMed]
  12. Söderberg, D.; Segelmark, M. Neutrophil extracellular traps in ANCA-associated vasculitis. Front. Immunol. 2016, 7, 256. [Google Scholar] [CrossRef]
  13. Lee, K.H.; Kronbichler, A.; Park, D.D.; 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]
  14. Hakkim, A.; Fürnrohr, B.G.; Amann, K.; Laube, B.; Abed, U.A.; Brinkmann, V.; Herrmann, M.; Voll, R.E.; Zychlinsky, A. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc. Natl. Acad. Sci. USA 2010, 107, 9813–9818. [Google Scholar] [CrossRef] [Green Version]
  15. Spengler, J.; Lugonja, B.; Ytterberg, A.J.; Zubarev, R.A.; Creese, A.J.; Pearson, M.J.; Grant, M.M.; Milward, M.; Lundberg, K.; Buckley, C.D.; et al. Release of active peptidyl arginine deiminases by neutrophils can explain production of extracellular citrullinated autoantigens in rheumatoid arthritis synovial fluid. Arthritis Rheumatol. 2015, 67, 3135–3145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kastner, D.L.; Aksentijevich, I.; Goldbach-Mansky, R. Autoinflammatory disease reloaded: A clinical perspective. Cell 2010, 140, 784–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Pay, S.; Türkçapar, N.; Kalyoncu, M.; Simşek, I.; Beyan, E.; Ertenli, I.; Oztürk, M.A.; Düzgün, N.; Erdem, H.; Ozbalkan, Z.; et al. A multicenter study of patients with adult-onset still’s disease compared with systemic juvenile idiopathic arthritis. Clin. Rheumatol. 2006, 25, 639–644. [Google Scholar]
  18. Inoue, N.; Shimizu, M.; Tsunoda, S.; Kawano, M.; Matsumura, M.; Yachie, A. Cytokine profile in adult-onset still’s disease: Comparison with systemic juvenile idiopathic arthritis. Clin. Immunol. 2016, 169, 8–13. [Google Scholar] [CrossRef] [Green Version]
  19. Jamilloux, Y.; Gerfaud-Valentin, M.; Martinon, F.; Belot, A.; Henry, T.; Sève, P. Pathogenesis of adult-onset still’s disease: New insights from the juvenile counterpart. Immunol. Res. 2015, 61, 53–62. [Google Scholar] [CrossRef]
  20. Pardeo, M.; Bracaglia, C.; De Benedetti, F. Systemic juvenile idiopathic arthritis: New insights into pathogenesis and cytokine directed therapies. Best Pract. Res. Clin. Rheumatol. 2017, 31, 505–516. [Google Scholar] [CrossRef]
  21. Yamaguchi, M.; Ohta, A.; Tsunematsu, T.; Kasukawa, R.; Mizushima, Y.; Kashiwagi, H.; Kashiwazaki, S.; Tanimoto, K.; Matsumoto, Y.; Ota, T.; et al. Preliminary criteria for classification of adult still’s disease. J. Rheumatol. 1992, 19, 424–430. [Google Scholar] [PubMed]
  22. Feist, E.; Mitrovic, S.; Fautrel, B. Mechanisms, biomarkers and targets for adult-onset still’s disease. Nat. Rev. Rheumatol. 2018, 14, 603–618. [Google Scholar] [CrossRef] [PubMed]
  23. Giacomelli, R.; Ruscitti, P.; Shoenfeld, Y. A comprehensive review on adult onset still’s disease. J. Autoimmun. 2018, 93, 24–36. [Google Scholar] [CrossRef] [PubMed]
  24. Giaglis, S.; Hahn, S.; Hasler, P. “The NET outcome”: Are neutrophil extracellular traps of any relevance to the pathophysiology of autoimmune disorders in childhood? Front. Pediatr. 2016, 4, 97. [Google Scholar] [CrossRef] [Green Version]
  25. Gerfaud-Valentin, M.; Jamilloux, Y.; Iwaz, J.; Sève, P. Adult-onset still’s disease. Autoimmun. Rev. 2014, 13, 708–722. [Google Scholar] [CrossRef] [Green Version]
  26. Cimaz, R. Systemic-onset juvenile idiopathic arthritis. Autoimmun. Rev. 2016, 15, 931–934. [Google Scholar] [CrossRef]
  27. Ahn, M.H.; Han, J.H.; Chwae, Y.J.; Jung, J.Y.; Suh, C.H.; Kwon, J.E.; Kim, H.A. Neutrophil Extracellular Traps May Contribute to the Pathogenesis in Adult-Onset Still Disease. J. Rheumatol. 2019, 46, 1560–1569. [Google Scholar] [CrossRef]
  28. Hu, Q.; Shi, H.; Zeng, T.; Liu, H.; Su, Y.; Cheng, X.; Ye, J.; Yin, Y.; Liu, M.; Zheng, H.; et al. Increased Neutrophil Extracellular Traps Activate NLRP3 and Inflammatory Macrophages in Adult-Onset Still’s Disease. Arthritis Res. Ther. 2019, 21, 9. [Google Scholar] [CrossRef] [Green Version]
  29. Liew, P.X.; Kubes, P. The Neutrophil’s Role during Health and Disease. Physiol. Rev. 2019, 99, 1223–1248. [Google Scholar] [CrossRef]
  30. Metzemaekers, M.; Gouwy, M.; Proost, P. Neutrophil Chemoattractant Receptors in Health and Disease: Double-Edged Swords. Cell. Mol. Immunol. 2020, 17, 433–450. [Google Scholar] [CrossRef]
  31. Abdolmaleki, F.; Kovanen, P.T.; Mardani, R.; Gheibi-Hayat, S.M.; Bo, S.; Sahebkar, A. Resolvins: Emerging Players in Autoimmune and Inflammatory Diseases. Clin. Rev. Allergy Immunol. 2020, 58, 82–91. [Google Scholar] [CrossRef] [PubMed]
  32. Rossaint, J.; Margraf, A.; Zarbock, A. Role of Platelets in Leukocyte Recruitment and Resolution of Inflammation. Front. Immunol. 2018, 9, 2712. [Google Scholar] [CrossRef]
  33. Serhan, C.N.; Chiang, N.; Dalli, J.; Levy, B.D. Lipid Mediators in the Resolution of Inflammation. Cold Spring Harb. Perspect. Biol. 2014, 7, a016311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Rosales, C. Neutrophils at the Crossroads of Innate and Adaptive Immunity. J. Leukoc. Biol. 2020, 108, 377–396. [Google Scholar] [CrossRef]
  35. Greenlee-Wacker, M.C. Clearance of Apoptotic Neutrophils and Resolution of Inflammation. Immunol. Rev. 2016, 273, 357–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Hahn, J.; Knopf, J.; Maueröder, C.; Kienhöfer, D.; Leppkes, M.; Herrmann, M. Neutrophils and Neutrophil Extracellular Traps Orchestrate Initiation and Resolution of Inflammation. Clin. Exp. Rheumatol. 2016, 34, 6–8. [Google Scholar]
  37. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil Extracellular Traps Kill Bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef] [PubMed]
  38. Delgado-Rizo, V.; Martínez-Guzmán, M.A.; Iñiguez-Gutierrez, L.; García-Orozco, A.; Alvarado-Navarro, A.; Fafutis-Morris, M. Neutrophil Extracellular Traps and Its Implications in Inflammation: An Overview. Front. Immunol. 2017, 8, 81. [Google Scholar] [CrossRef] [Green Version]
  39. Hakkim, A.; Fuchs, T.A.; Martinez, N.E.; Hess, S.; Prinz, H.; Zychlinsky, A.; Waldmann, H. Activation of the Raf-MEK-ERK Pathway Is Required for Neutrophil Extracellular Trap Formation. Nat. Chem. Biol. 2011, 7, 75–77. [Google Scholar] [CrossRef]
  40. Van Avondt, K.; Hartl, D. Mechanisms and Disease Relevance of Neutrophil Extracellular Trap Formation. Eur. J. Clin. Investig. 2018, 48, e12919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Yipp, B.G.; Kubes, P. NETosis: How Vital Is It? Blood 2013, 122, 2784–2794. [Google Scholar] [CrossRef] [PubMed]
  42. Mulay, S.R.; Anders, H.-J. Neutrophils and Neutrophil Extracellular Traps Regulate Immune Responses in Health and Disease. Cells 2020, 9, 2130. [Google Scholar] [CrossRef]
  43. Rubartelli, A.; Lotze, M.T.; Latz, E.; Manfredi, A.A. Mechanisms of Sterile Inflammation. Front. Immunol. 2013, 4, 398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Rossaint, J.; Herter, J.M.; Van Aken, H.; Napirei, M.; Döring, Y.; Weber, C.; Soehnlein, O.; Zarbock, A. Synchronized Integrin Engagement and Chemokine Activation Is Crucial in Neutrophil Extracellular Trap–Mediated Sterile Inflammation. Blood 2014, 123, 2573–2584. [Google Scholar] [CrossRef]
  45. Chen, G.Y.; Nuñez, G. Sterile Inflammation: Sensing and Reacting to Damage. Nat. Rev. Immunol. 2010, 10, 826–837. [Google Scholar] [CrossRef] [Green Version]
  46. Rock, K.L.; Latz, E.; Ontiveros, F.; Kono, H. The Sterile Inflammatory Response. Annu. Rev. Immunol. 2010, 28, 321–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Amulic, B.; Cazalet, C.; Hayes, G.L.; Metzler, K.D.; Zychlinsky, A. Neutrophil Function: From Mechanisms to Disease. Annu. Rev. Immunol. 2012, 30, 459–489. [Google Scholar] [CrossRef]
  48. De Oliveira, S.; Rosowski, E.E.; Huttenlocher, A. Neutrophil Migration in Infection and Wound Repair: Going Forward in Reverse. Nat. Rev. Immunol. 2016, 16, 378–391. [Google Scholar] [CrossRef] [Green Version]
  49. McDermott, M.F.; Aksentijevich, I.; Galon, J.; McDermott, E.M.; Ogunkolade, B.W.; Centola, M.; Mansfield, E.; Gadina, M.; Karenko, L.; Pettersson, T.; et al. Germline Mutations in the Extracellular Domains of the 55 kDa TNF Receptor, TNFR1, Define a Family of Dominantly Inherited Autoinflammatory Syndromes. Cell 1999, 97, 133–144. [Google Scholar] [CrossRef]
  50. Savic, S.; Dickie, L.J.; Wittmann, M.; McDermott, M.F. Autoinflammatory Syndromes and Cellular Responses to Stress: Pathophysiology, Diagnosis and New Treatment Perspectives. Best Pract. Res. Clin. Rheumatol. 2012, 26, 505–533. [Google Scholar] [CrossRef]
  51. Ciccarelli, F.; De Martinis, M.; Ginaldi, L. An Update on Autoinflammatory Diseases. Curr. Med. Chem. 2014, 21, 261–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Beck, D.B.; Aksentijevich, I. Biochemistry of Autoinflammatory Diseases: Catalyzing Monogenic Disease. Front. Immunol. 2019, 10, 101. [Google Scholar] [CrossRef] [Green Version]
  53. Krainer, J.; Siebenhandl, S.; Weinhäusel, A. Systemic Autoinflammatory Diseases. J. Autoimmun. 2020, 109, 102421. [Google Scholar] [CrossRef]
  54. Navallas, M.; Inarejos Clemente, E.J.; Iglesias, E.; Rebollo-Polo, M.; Hernández, J.C.; Navarro, O.M. Autoinflammatory Diseases in Childhood, part 2: Polygenic Syndromes. Pediatr. Radiol. 2020, 50, 431–444. [Google Scholar] [CrossRef]
  55. Arakelyan, A.; Nersisyan, L.; Poghosyan, D.; Khondkaryan, L.; Hakobyan, A.; Löffler-Wirth, H.; Melanitou, E.; Binder, H. Autoimmunity and Autoinflammation: A Systems View on Signaling Pathway Dysregulation Profiles. PLoS ONE 2017, 12, e0187572. [Google Scholar]
  56. Ozen, S.; Bilginer, Y. A Clinical Guide to Autoinflammatory Diseases: Familial Mediterranean Fever and Next-of-Kin. Nat. Rev. Rheumatol. 2014, 10, 135–147. [Google Scholar] [CrossRef]
  57. Shohat, M.; Halpern, G.J. Familial Mediterranean Fever—A Review. Genet. Med. 2011, 13, 487–498. [Google Scholar] [CrossRef] [Green Version]
  58. Manukyan, G.; Petrek, M.; Kriegova, E.; Ghazaryan, K.; Fillerova, R.; Boyajyan, A. Activated Phenotype of Circulating Neutrophils in Familial Mediterranean Fever. Immunobiology 2013, 218, 892–898. [Google Scholar] [CrossRef]
  59. Skendros, P.; Papagoras, C.; Mitroulis, I.; Ritis, K. Autoinflammation: Lessons from the Study of Familial Mediterranean Fever. J. Autoimmun. 2019, 104, 102305. [Google Scholar] [CrossRef]
  60. Kimura, T.; Jain, A.; Choi, S.W.; Mandell, M.A.; Schroder, K.; Johansen, T.; Deretic, V. TRIM-Mediated Precision Autophagy Targets Cytoplasmic Regulators of Innate Immunity. J. Cell Biol. 2015, 210, 973–989. [Google Scholar] [CrossRef] [Green Version]
  61. Gohar, F.; Orak, B.; Kallinich, T.; Jeske, M.; Lieber, M.; Von Bernuth, H.; Giese, A.; Weissbarth-Riedel, E.; Haas, J.P.; Dressler, F.; et al. Correlation of Secretory Activity of Neutrophils With Genotype in Patients With Familial Mediterranean Fever. Arthritis Rheumatol. 2016, 68, 3010–3022. [Google Scholar] [CrossRef] [Green Version]
  62. Chae, J.J.; Cho, Y.H.; Lee, G.S.; Cheng, J.; Liu, P.P.; Feigenbaum, L.; Katz, S.I.; Kastner, D.L. Gain-of-Function Pyrin Mutations Induce NLRP3 Protein-Independent Interleukin-1β Activation and Severe Autoinflammation in Mice. Immunity 2011, 34, 755–768. [Google Scholar] [CrossRef] [Green Version]
  63. Apostolidou, E.; Skendros, P.; Kambas, K.; Mitroulis, I.; Konstantinidis, T.; Chrysanthopoulou, A.; Nakos, K.; Tsironidou, V.; Koffa, M.; Boumpas, D.T.; et al. Neutrophil Extracellular Traps Regulate IL-1β-Mediated Inflammation in Familial Mediterranean Fever. Ann. Rheum. Dis. 2016, 75, 269–277. [Google Scholar] [CrossRef]
  64. Skendros, P.; Chrysanthopoulou, A.; Rousset, F.; Kambas, K.; Arampatzioglou, A.; Mitsios, A.; Bocly, V.; Konstantinidis, T.; Pellet, P.; Angelidou, I.; et al. Regulated in Development and DNA Damage responses 1 (REDD1) Links Stress With IL-1β–Mediated Familial Mediterranean Fever Attack Through Autophagy-Driven Neutrophil Extracellular Traps. J. Allergy Clin. Immunol. 2017, 140, 1378–1387.e13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Oliviero, F.; Bindoli, S.; Scanu, A.; Feist, E.; Doria, A.; Galozzi, P.; Sfriso, P. Autoinflammatory Mechanisms in Crystal-Induced Arthritis. Front. Med. (Lausanne). 2020, 7, 166. [Google Scholar] [CrossRef] [PubMed]
  66. Malawista, S.E.; de Boisfleury, A.C.; Naccache, P.H. Inflammatory Gout: Observations over a Half-Century. FASEB J. 2011, 25, 4073–4078. [Google Scholar] [CrossRef] [Green Version]
  67. Mitroulis, I.; Kambas, K.; Ritis, K. Neutrophils, IL-1β, and Gout: Is There a Link? Semin. Immunopathol. 2013, 35, 501–512. [Google Scholar] [CrossRef]
  68. Busso, N.; So, A. Gout. Mechanisms of Inflammation in Gout. Arthritis Res. Ther. 2010, 12, 1–8. [Google Scholar] [CrossRef] [Green Version]
  69. Mitroulis, I.; Kambas, K.; Chrysanthopoulou, A.; Skendros, P.; Apostolidou, E.; Kourtzelis, I.; Drosos, G.I.; Boumpas, D.T.; Ritis, K. Neutrophil Extracellular Trap Formation Is Associated With IL-1β and Autophagy-Related Signaling in Gout. PLoS ONE 2011, 6, e29318. [Google Scholar] [CrossRef]
  70. Jeong, J.H.; Choi, S.J.; Ahn, S.M.; Oh, J.S.; Kim, Y.G.; Lee, C.K.; Yoo, B.; Hong, S. Neutrophil Extracellular Trap Clearance by Synovial Macrophages in Gout. Arthritis Res. Ther. 2021, 23, 88. [Google Scholar] [CrossRef]
  71. Schauer, C.; Janko, C.; Munoz, L.E.; Zhao, Y.; Kienhöfer, D.; Frey, B.; Lell, M.; Manger, B.; Rech, J.; Naschberger, E.; et al. Aggregated Neutrophil Extracellular Traps Limit Inflammation by Degrading Cytokines and Chemokines. Nat. Med. 2014, 20, 511–517. [Google Scholar] [CrossRef] [PubMed]
  72. Desai, J.; Steiger, S.; Anders, H.J. Molecular Pathophysiology of Gout. Trends Mol. Med. 2017, 23, 756–768. [Google Scholar] [CrossRef] [PubMed]
  73. De Luca, A.; Smeekens, S.P.; Casagrande, A.; Iannitti, R.; Conway, K.L.; Gresnigt, M.S.; Begun, J.; Plantinga, T.S.; Joosten, L.A.; Van Der Meer, J.W.; et al. IL-1 Receptor Blockade Restores Autophagy and Reduces Inflammation in Chronic Granulomatous Disease in Mice and in Humans. Proc. Natl. Acad. Sci. USA 2014, 111, 3526–3531. [Google Scholar] [CrossRef] [Green Version]
  74. De Lange, K.M.; Barrett, J.C. Understanding Inflammatory Bowel Disease via Immunogenetics. J. Autoimmun. 2015, 64, 91–100. [Google Scholar] [CrossRef] [Green Version]
  75. Zhang, Y.Z.; Li, Y.Y. Inflammatory Bowel Disease: Pathogenesis. World J. Gastroenterol. 2014, 20, 91–99. [Google Scholar] [CrossRef]
  76. Naito, Y.; Takagi, T.; Yoshikawa, T. Molecular Fingerprints of Neutrophil-Dependent Oxidative Stress in Inflammatory Bowel Disease. J. Gastroenterol. 2007, 42, 787–798. [Google Scholar] [CrossRef]
  77. Zhou, G.X.; Liu, Z.J. Potential Roles of Neutrophils in Regulating Intestinal Mucosal Inflammation of Inflammatory Bowel Disease. J. Dig. Dis. 2017, 18, 495–503. [Google Scholar] [CrossRef] [Green Version]
  78. Biasi, F.; Leonarduzzi, G.; Oteiza, P.I.; Poli, G. Inflammatory Bowel Disease: Mechanisms, Redox Considerations, and Therapeutic Targets. Antioxid. Redox Signal. 2013, 19, 1711–1747. [Google Scholar] [CrossRef] [Green Version]
  79. Bennike, T.B.; Carlsen, T.G.; Ellingsen, T.; Bonderup, O.K.; Glerup, H.; Bøgsted, M.; Christiansen, G.; Birkelund, S.; Stensballe, A.; Andersen, V. Neutrophil Extracellular Traps in Ulcerative Colitis: A Proteome Analysis of Intestinal Biopsies. Inflamm. Bowel Dis. 2015, 21, 2052–2067. [Google Scholar] [CrossRef] [Green Version]
  80. He, Z.; Si, Y.; Jiang, T.; Ma, R.; Zhang, Y.; Cao, M.; Li, T.; Yao, Z.; Zhao, L.; Fang, S.; et al. Phosphotidylserine Exposure and Neutrophil Extracellular Traps Enhance Procoagulant Activity in Patients With Inflammatory Bowel Disease. Thromb. Haemost. 2016, 115, 738–751. [Google Scholar]
  81. Cao, M.; Yu, M.; Zhang, Y.; Tong, D.; Guo, L.; Wang, C.; Li, T.; Yang, X.; Zhang, C.; Kou, J. Neutrophil Extracellular Traps Exacerbate Inflammatory Responses and Thrombotic Tendency in Both a Murine Colitis Model and Patients With Inflammatory Bowel Disease; American Society of Hematology: Washington, DC, USA, 2017. [Google Scholar]
  82. Angelidou, I.; Chrysanthopoulou, A.; Mitsios, A.; Arelaki, S.; Arampatzioglou, A.; Kambas, K.; Ritis, D.; Tsironidou, V.; Moschos, I.; Dalla, V.; et al. REDD1/autophagy Pathway Is Associated With Neutrophil-Driven IL-1β Inflammatory Response in Active Ulcerative Colitis. J. Immunol. 2018, 200, 3950–3961. [Google Scholar] [CrossRef]
  83. Drury, B.; Hardisty, G.; Gray, R.D.; Ho, G.T. Neutrophil Extracellular Traps in Inflammatory Bowel Disease: Pathogenic Mechanisms and Clinical Translation. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 321–333. [Google Scholar] [CrossRef] [PubMed]
  84. Choi, J.H.; Suh, C.H.; Lee, Y.M.; Suh, Y.J.; Lee, S.K.; Kim, S.S.; Nahm, D.H.; Park, H.S. Serum Cytokine Profiles in Patients With Adult Onset Still’s Disease. J. Rheumatol. 2003, 30, 2422–2427. [Google Scholar] [PubMed]
  85. Kasama, T.; Furuya, H.; Yanai, R.; Ohtsuka, K.; Takahashi, R.; Yajima, N.; Miwa, Y.; Kobayashi, K. Correlation of Serum CX3CL1 Level With Disease Activity in Adult-Onset Still’s Disease and Significant Involvement in Hemophagocytic Syndrome. Clin. Rheumatol. 2012, 31, 853–860. [Google Scholar] [CrossRef]
  86. Chen, D.Y.; Lan, J.L.; Lin, F.J.; Hsieh, T.Y. Association of Intercellular Adhesion molecule-1 With Clinical Manifestations and Interleukin-18 in Patients With Active, Untreated Adult-Onset Still’s Disease. Arthritis Rheum. 2005, 53, 320–327. [Google Scholar] [CrossRef] [PubMed]
  87. Komiya, A.; Matsui, T.; Nogi, S.; Iwata, K.; Futami, H.; Takaoka, H.; Arinuma, Y.; Hashimoto, A.; Shimada, K.; Ikenaka, T.; et al. Neutrophil CD64 Is Upregulated in Patients With Active Adult-Onset Still’s Disease. Scand. J. Rheumatol. 2012, 41, 156–158. [Google Scholar] [CrossRef]
  88. Chen, P.K.; Hsieh, S.L.; Lan, J.L.; Lin, C.C.; Chang, S.H.; Chen, D.Y. Elevated Expression of C-Type Lectin Domain family 5-Member A (CLEC5A) and Its Relation to Inflammatory Parameters and Disease Course in Adult-Onset Still’s Disease. J. Immunol. Res. 2020, 2020, 9473497. [Google Scholar] [CrossRef]
  89. Kim, H.A.; Choi, B.; Suh, C.H.; Han, M.H.; Jung, J.Y.; Sayeed, H.M.; Kim, Y.W.; Sohn, S. Highly Expression of CD11b and CD32 on Peripheral Blood Mononuclear Cells From Patients With Adult-Onset Still’s Disease. Int. J. Mol. Sci. 2017, 18, 202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Ramanathan, K.; Glaser, A.; Lythgoe, H.; Ong, J.; Beresford, M.W.; Midgley, A.; Wright, H.L. Neutrophil Activation Signature in Juvenile Idiopathic Arthritis Indicates the Presence of Low-Density Granulocytes. Rheumatology 2018, 57, 488–498. [Google Scholar] [CrossRef] [Green Version]
  91. Liu, Y.; Xia, C.; Chen, J.; Fan, C.; He, J. Elevated Circulating Pro-Inflammatory Low-Density Granulocytes in Adult-Onset Still’s Disease. Rheumatology 2021, 60, 297–303. [Google Scholar] [CrossRef] [PubMed]
  92. Brown, R.A.; Henderlight, M.; Do, T.; Yasin, S.; Grom, A.A.; DeLay, M.; Thornton, S.; Schulert, G.S. Neutrophils From Children With Systemic Juvenile Idiopathic Arthritis Exhibit Persistent Proinflammatory Activation Despite Long-Standing Clinically Inactive Disease. Front. Immunol. 2018, 9, 2995. [Google Scholar] [CrossRef] [PubMed]
  93. De Benedetti, F.; Massa, M.; Robbioni, P.; Ravelli, A.; Burgio, G.R.; Martini, A. Correlation of Serum Interleukin-6 Levels With Joint Involvement and Thrombocytosis in Systemic Juvenile Rheumatoid Arthritis; Wiley: Hoboken, NJ, USA, 1991. [Google Scholar]
  94. De Jager, W.; Hoppenreijs, E.P.; Wulffraat, N.M.; Wedderburn, L.R.; Kuis, W.; Prakken, B.J. Blood and Synovial Fluid Cytokine Signatures in Patients with Juvenile Idiopathic Arthritis: A Cross-Sectional Study. Ann. Rheum. Dis. 2007, 66, 589–598. [Google Scholar] [CrossRef] [Green Version]
  95. Pascual, V.; Allantaz, F.; Arce, E.; Punaro, M.; Banchereau, J. Role of Interleukin-1 (IL-1) in the Pathogenesis of Systemic Onset Juvenile Idiopathic Arthritis and Clinical Response to IL-1 Blockade. J. Exp. Med. 2005, 201, 1479–1486. [Google Scholar] [CrossRef] [PubMed]
  96. Han, J.H.; Suh, C.H.; Jung, J.Y.; Nam, J.Y.; Kwon, J.E.; Yim, H.; Kim, H.A. Association of CXCL10 and CXCL13 Levels With Disease Activity and Cutaneous Manifestation in Active Adult-Onset Still’s Disease. Arthritis Res. Ther. 2015, 17, 260. [Google Scholar] [CrossRef] [Green Version]
  97. Han, J.H.; Ahn, M.H.; Jung, J.Y.; Suh, C.H.; Kwon, J.E.; Yim, H.; Kim, H.A. The Levels of CXCL12 and Its Receptor, CXCR4, as a Biomarker of Disease Activity and Cutaneous Manifestation in Adult-Onset Still’s Disease. Clin. Exp. Rheumatol. 2019, 37, 67–73. [Google Scholar]
  98. Ma, Y.; Meng, J.; Jia, J.; Wang, M.; Teng, J.; Zhu, D.; Yang, C.; Hu, Q. Current and Emerging Biological Therapy in Adult-Onset Still’s Disease. Rheumatology 2021, 60, 3986–4000. [Google Scholar] [CrossRef] [PubMed]
  99. Carmona-Rivera, C.; Kaplan, M.J. Low-Density Granulocytes: A Distinct Class of Neutrophils in Systemic Autoimmunity. Semin. Immunopathol. 2013, 35, 455–463. [Google Scholar] [CrossRef] [PubMed]
  100. Brinkmann, V. Neutrophil Extracellular Traps in the Second Decade. J. Innate Immun. 2018, 10, 414–421. [Google Scholar] [CrossRef]
  101. Roth, J.; Goebeler, M.; Sorg, C. S100A8 and S100A9 in Inflammatory Diseases. Lancet 2001, 357, 1041. [Google Scholar] [CrossRef]
  102. Austermann, J.; Spiekermann, C.; Roth, J. S100 Proteins in Rheumatic Diseases. Nat. Rev. Rheumatol. 2018, 14, 528–541. [Google Scholar] [CrossRef]
  103. Wang, S.; Song, R.; Wang, Z.; Jing, Z.; Wang, S.; Ma, J. S100A8/A9 in Inflammation. Front. Immunol. 2018, 9, 1298. [Google Scholar] [CrossRef] [PubMed]
  104. Frosch, M.; Ahlmann, M.; Vogl, T.; Wittkowski, H.; Wulffraat, N.; Foell, D.; Roth, J. The Myeloid-Related proteins 8 and 14 Complex, a Novel Ligand of Toll-Like Receptor 4, and Interleukin-1Beta Form a Positive Feedback Mechanism in Systemic-Onset Juvenile Idiopathic Arthritis. Arthritis Rheum. 2009, 60, 883–891. [Google Scholar] [CrossRef]
  105. Kim, H.A.; Han, J.H.; Kim, W.J.; Noh, H.J.; An, J.M.; Yim, H.; Jung, J.Y.; Kim, Y.S.; Suh, C.H. TLR4 Endogenous Ligand S100A8/A9 Levels in Adult-Onset Still’s Disease and Their Association With Disease Activity and Clinical Manifestations. Int. J. Mol. Sci. 2016, 17, 1342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Wittkowski, H.; Frosch, M.; Wulffraat, N.; Goldbach-Mansky, R.; Kallinich, T.; Kuemmerle-Deschner, J.; Frühwald, M.C.; Dassmann, S.; Pham, T.H.; Roth, J.; et al. S100A12 Is a Novel Molecular Marker Differentiating Systemic-Onset Juvenile Idiopathic Arthritis From Other Causes of Fever of Unknown Origin. Arthritis Rheum. 2008, 58, 3924–3931. [Google Scholar] [CrossRef] [PubMed]
  107. Bae, C.B.; Suh, C.H.; An, J.M.; Jung, J.Y.; Jeon, J.Y.; Nam, J.Y.; Kim, H.A. Serum S100A12 May Be a Useful Biomarker of Disease Activity in Adult-Onset Still’s Disease. J. Rheumatol. 2014, 41, 2403–2408. [Google Scholar] [CrossRef] [PubMed]
  108. Backlund, M.; Venge, P.; Berntson, L. A Cross-Sectional Cohort Study of the Activity and Turnover of Neutrophil Granulocytes in Juvenile Idiopathic Arthritis. Pediatr. Rheumatol. Online J. 2021, 19, 102. [Google Scholar] [CrossRef]
  109. Jia, J.; Yang, L.; Cao, Z.; Wang, M.; Ma, Y.; Ma, X.; Liu, Q.; Teng, J.; Shi, H.; Liu, H.; et al. Neutrophil-Derived lipocalin-2 in Adult-Onset Still’s Disease: A Novel Biomarker of Disease Activity and Liver Damage. Rheumatology 2021, 60, 304–315. [Google Scholar] [CrossRef] [PubMed]
  110. Liao, T.L.; Chen, Y.M.; Tang, K.T.; Chen, P.K.; Liu, H.J.; Chen, D.Y. MicroRNA-223 Inhibits Neutrophil Extracellular Traps Formation Through Regulating Calcium Influx and Small Extracellular Vesicles Transmission. Sci. Rep. 2021, 11, 15676. [Google Scholar] [CrossRef]
  111. Torres-Ruiz, J.; Carrillo-Vázquez, D.A.; Tapia-Rodríguez, M.; Garcia-Galicia, J.A.; Alcocer-Varela, J.; Gómez-Martín, D. The Role of Low Density Granulocytes and NETosis in the Pathogenesis of Adult-Onset Still’s Disease. Clin. Exp. Rheumatol. 2019, 37, 74–82. [Google Scholar]
  112. Jung, J.Y.; Suh, C.H.; Sohn, S.; Nam, J.Y.; Kim, H.A. Elevated High-Mobility Group B1 Levels in Active Adult-Onset Still’s Disease Associated With Systemic Score and Skin Rash. Clin. Rheumatol. 2016, 35, 1937–1942. [Google Scholar] [CrossRef]
  113. Xu, D.; Zhang, Y.; Zhang, Z.Y.; Tang, X.M. Association Between High Mobility Group Box 1 Protein and Juvenile Idiopathic Arthritis: A Prospective Longitudinal Study. Pediatr. Rheumatol. Online J. 2021, 19, 112. [Google Scholar] [CrossRef]
  114. Bobek, D.; Grčević, D.; Kovačić, N.; Lukić, I.K.; Jelušić, M. The Presence of High Mobility Group box-1 and Soluble Receptor for Advanced Glycation End-Products in Juvenile Idiopathic Arthritis and Juvenile Systemic Lupus Erythematosus. Pediatr. Rheumatol. Online J. 2014, 12, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Jia, J.; Wang, M.; Ma, Y.; Teng, J.; Shi, H.; Liu, H.; Sun, Y.; Su, Y.; Meng, J.; Chi, H.; et al. Circulating Neutrophil Extracellular Traps Signature for Identifying Organ Involvement and Response to Glucocorticoid in Adult-Onset Still’s Disease: A Machine Learning Study. Front. Immunol. 2020, 11, 563335. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, M.; Liu, M.; Jia, J.; Shi, H.; Teng, J.; Liu, H.; Sun, Y.; Cheng, X.; Ye, J.; Su, Y.; et al. Association of the Leukocyte Immunoglobulin-Like Receptor A3 Gene With Neutrophil Activation and Disease Susceptibility in Adult-Onset Still’s Disease. Arthritis Rheumatol. 2021, 73, 1033–1043. [Google Scholar] [CrossRef] [PubMed]
  117. Hu, X.; Xie, Q.; Mo, X.; Jin, Y. The Role of Extracellular Histones in Systemic-Onset Juvenile Idiopathic Arthritis. Ital. J. Pediatr. 2019, 45, 14. [Google Scholar] [CrossRef] [PubMed]
  118. Tsung, A.; Tohme, S.; Billiar, T.R. High-Mobility Group box-1 in Sterile Inflammation. J. Intern. Med. 2014, 276, 425–443. [Google Scholar] [CrossRef]
  119. Ge, Y.; Huang, M.; Yao, Y.M. The Effect and Regulatory Mechanism of High Mobility Group Box-1 Protein on Immune Cells in Inflammatory Diseases. Cells 2021, 10, 1044. [Google Scholar] [CrossRef]
  120. Pahar, B.; Madonna, S.; Das, A.; Albanesi, C.; Girolomoni, G. Immunomodulatory Role of the Antimicrobial LL-37 Peptide in Autoimmune Diseases and Viral Infections. Vaccines 2020, 8, 517. [Google Scholar] [CrossRef] [PubMed]
  121. Illei, G.G.; Tackey, E.; Lapteva, L.; Lipsky, P.E. Biomarkers in Systemic Lupus Erythematosus. I. General Overview of Biomarkers and Their Applicability. Arthritis Rheum. 2004, 50, 1709–1720. [Google Scholar] [CrossRef] [PubMed]
  122. Yilmaz, M.; Kendirli, S.G.; Altintas, D.; Bingöl, G.; Antmen, B. Cytokine Levels in Serum of Patients With Juvenile Rheumatoid Arthritis. Clin. Rheumatol. 2001, 20, 30–35. [Google Scholar] [CrossRef]
  123. De Benedetti, F.; Pignatti, P.; Bernasconi, S.; Gerloni, V.; Matsushima, K.; Caporali, R.; Montecucco, C.M.; Sozzani, S.; Fantini, F.; Martini, A. Interleukin 8 and Monocyte Chemoattractant protein-1 in Patients With Juvenile Rheumatoid Arthritis. Relation to Onset Types, Disease Activity, and Synovial Fluid Leukocytes. J. Rheumatol. 1999, 26, 425–431. [Google Scholar]
  124. Chen, D.Y.; Lan, J.L.; Lin, F.J.; Hsieh, T.Y. Proinflammatory Cytokine Profiles in Sera and Pathological Tissues of Patients With Active Untreated Adult Onset Still’s Disease. J. Rheumatol. 2004, 31, 2189–2198. [Google Scholar] [PubMed]
  125. Yasin, S.; Fall, N.; Brown, R.A.; Henderlight, M.; Canna, S.W.; Girard-Guyonvarc’h, C.; Gabay, C.; Grom, A.A.; Schulert, G.S. IL-18 as a Biomarker Linking Systemic Juvenile Idiopathic Arthritis and Macrophage Activation Syndrome. Rheumatology 2020, 59, 361–366. [Google Scholar] [CrossRef] [PubMed]
  126. Bracaglia, C.; De Graaf, K.; Pires Marafon, D.; Guilhot, F.; Ferlin, W.; Prencipe, G.; Caiello, I.; Davì, S.; Schulert, G.; Ravelli, A.; et al. Elevated Circulating Levels of Interferon-γ and Interferon-γ-Induced Chemokines Characterise Patients With Macrophage Activation Syndrome Complicating Systemic Juvenile Idiopathic Arthritis. Ann. Rheum. Dis. 2017, 76, 166–172. [Google Scholar] [CrossRef]
  127. Han, J.H.; Suh, C.H.; Jung, J.Y.; Ahn, M.H.; Han, M.H.; Kwon, J.E.; Yim, H.; Kim, H.A. Elevated Circulating Levels of the Interferon-γ-Induced Chemokines Are Associated With Disease Activity and Cutaneous Manifestations in Adult-Onset Still’s Disease. Sci. Rep. 2017, 7, 46652. [Google Scholar] [CrossRef] [Green Version]
  128. Zhang, W.; Yang, T.; Zhang, H.; Xu, Y.; Yang, Q.; Liu, Q.; Gao, Y.; Wu, J.; Shao, L.; Zhang, W. Biomarker Screening and Validation for the Differentiation of Bloodstream Infection From Adult-Onset Still’s Disease: A Prospective Cohort Study. Cytokine 2021, 146, 155642. [Google Scholar] [CrossRef] [PubMed]
  129. Holzinger, D.; Frosch, M.; Kastrup, A.; Prince, F.H.; Otten, M.H.; Van Suijlekom-Smit, L.W.; Ten Cate, R.; Hoppenreijs, E.P.; Hansmann, S.; Moncrieffe, H.; et al. The Toll-Like Receptor 4 Agonist MRP8/14 Protein Complex Is a Sensitive Indicator for Disease Activity and Predicts Relapses in Systemic-Onset Juvenile Idiopathic Arthritis. Ann. Rheum. Dis. 2012, 71, 974–980. [Google Scholar] [CrossRef]
  130. Aljaberi, N.; Tronconi, E.; Schulert, G.; Grom, A.A.; Lovell, D.J.; Huggins, J.L.; Henrickson, M.; Brunner, H.I. The Use of S100 Proteins Testing in Juvenile Idiopathic Arthritis and Autoinflammatory Diseases in a Pediatric Clinical Setting: A Retrospective Analysis. Pediatr. Rheumatol. Online J. 2020, 18, 7. [Google Scholar] [CrossRef] [Green Version]
  131. Guo, Q.; Zha, X.; Li, C.; Jia, Y.; Zhu, L.; Guo, J.; Su, Y. Serum Calprotectin--A Promising Diagnostic Marker for Adult-Onset Still’s Disease. Clin. Rheumatol. 2016, 35, 73–79. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Overview of the role of neutrophils in AOSD and SJIA. Circulating neutrophils are triggered by the release of pathogen-associated molecular pattern molecules (PAMPs) or damage-associated molecular pattern molecules (DAMPs) during inflammation and transmigrate into inflamed tissues. In AOSD and SJIA, a variety of specific receptors on neutrophils, including pattern-recognition receptors and Fc receptors interacting with PAMPs and DAMPs increase, further promoting neutrophil recruitment and activation. Activated neutrophils release more cytokines/chemokines and communicate with macrophages in the innate immune system. Neutrophil activation and mediator release form a positive-feedback loop that enhances neutrophil recruitment and amplifies inflammatory responses, acting as an axis of pathogenesis in AOSD and SJIA.
Figure 1. Overview of the role of neutrophils in AOSD and SJIA. Circulating neutrophils are triggered by the release of pathogen-associated molecular pattern molecules (PAMPs) or damage-associated molecular pattern molecules (DAMPs) during inflammation and transmigrate into inflamed tissues. In AOSD and SJIA, a variety of specific receptors on neutrophils, including pattern-recognition receptors and Fc receptors interacting with PAMPs and DAMPs increase, further promoting neutrophil recruitment and activation. Activated neutrophils release more cytokines/chemokines and communicate with macrophages in the innate immune system. Neutrophil activation and mediator release form a positive-feedback loop that enhances neutrophil recruitment and amplifies inflammatory responses, acting as an axis of pathogenesis in AOSD and SJIA.
Ijms 22 13038 g001
Figure 2. Neutrophil extracellular traps and its implications in AOSD and SJIA. Several neutrophil extracellular traps (NETs) released through neutrophils such as S100 protein, high mobility group box-1 protein, and LL–37 accelerate inflammation with a proinflammatory cascade through toll–like receptor 4 or receptor for advanced glycation end products or inflammasome activation. NET formation promotes cytokine storms by linking neutrophils and macrophages.
Figure 2. Neutrophil extracellular traps and its implications in AOSD and SJIA. Several neutrophil extracellular traps (NETs) released through neutrophils such as S100 protein, high mobility group box-1 protein, and LL–37 accelerate inflammation with a proinflammatory cascade through toll–like receptor 4 or receptor for advanced glycation end products or inflammasome activation. NET formation promotes cytokine storms by linking neutrophils and macrophages.
Ijms 22 13038 g002
Table 1. List of studies indicating association between neutrophils and SJIA/AOSD.
Table 1. List of studies indicating association between neutrophils and SJIA/AOSD.
Study PopulationMain FindingsCountryAuthor, Year [ref.]
Analysis of serum from 14 patients with AOSD Overproduction of CXCL–8 may contribute to the pathogenic mechanism of AOSD.South KoreaChoi J-H et al., 2003 [84]
Analysis of serum from 19 patients with AOSD and 19 HCsSerum CX3CL1 level may be used as a clinical marker to assess the disease activity of AOSD, and high serum CXCL–8 and ferritin reflected the presence of hemophagocytic syndrome.JapanKasama T et al., 2012 [85]
Analysis of serum from 50 patients with untreated AOSD, 20 with RA, and 20 HCsSerum levels of ICAM–1 were significantly elevated in patients with active untreated AOSD compared with those with active RA and HCs. TaiwanChen DY et al., 2005 [86]
Analysis of serum from 10 patients with AOSD Neutrophil CD64 is upregulated in patients with active AOSDJapanKomiya A et al., 2012 [87]
Analysis of serum from 34 patients with AOSD and 12 HCsC-type lectin domain family 5–member A is involved in the pathogenesis and may serve as an activity indicator of AOSD.TaiwanChen P-K et al., 2020 [88]
Analysis of serum from 13 patients with AOSD, 19 with RA, and 19 HCsSignificantly higher frequencies of cells presenting CD11b and CD32 from whole blood cells in patients with AOSD than in patients with RA or HCKoreaKim HA et al., 2017 [89]
Analysis of serum from 16 patients with JIA (3 SJIA) and 19 HCsNeutrophils from JIA patients have elevated transcription of genes encoding granule proteins, a major cause of inflammation.UKRamanathan K et al., 2018 [90]
Analysis of serum from 56 patients with AOSD (active 32 and inactive 24) and 26 HCsActive AOSD is associated with elevated levels of low-density granulocytes that produce IL–6ChinaLiu Y et al., 2021 [91]
Analysis of serum from 23 patients with active SJIA and 22 with inactive SJIANeutrophil activations, S100 alarmin release, and proinflammatory gene expression were seen in SJIA patients with both active disease and clinically inactive disease.United StatesBrown RA et al., 2018 [92]
Analysis of serum from 39 patients with SJIA (active 25 and inactive 14) and 17 HCsIL-6 plays a significant role in the pathogenesis of SJIA.ItalyDe Benedetti F et al., 1991 [93]
Analysis of serum and synovial fluid from 65 patients with JIA (20 SJIA), 9 with type I diabetes, and 20 HCsSeveral cytokines including IL18 may correspond to the activation status during inflammation in JIA The NetherlandsDe Jager W et al., 2007 [94]
Analysis of serum from 23 patients with SJIA, and 12 HCsIL-1 is a major mediator of the inflammatory cascade that underlies SJIAUnited StatesPascual V et al., 2005 [95]
Analysis of serum from 39 patients with active AOSD, 32 with RA, and 40 HCsSerum CXCL10 and CXCL13 levels may serve as clinical markers for assessment of disease activity, especially skin manifestations, in AOSDSouth KoreaHan JH et al., 2015 [96]
Analysis of skin biopsy materials of 40 patients with AOSD, 10 with eczema, 10 with psoriasis, and 10 HCsCXCR4 could be a clinical biomarker of evaluation for disease activity in AOSD. CXCR4/CXCL12 may influence skin manifestations of AOSD.South KoreaHan JH et al., 2019 [97]
SJIA, systemic juvenile idiopathic arthritis; AOSD, adult–onset Still’s disease; HC, healthy control; RA, rheumatoid arthritis, ICAM–1, intracellular adhesion molecule–1; IL, interleukin; SLE, systemic lupus erythematosus.
Table 2. List of studies indicating association between NETs and SJIA/AOSD.
Table 2. List of studies indicating association between NETs and SJIA/AOSD.
Study PopulationMain FindingsCountryAuthor, Year [ref.]
Analysis of serum from 60 patients with SJIA, 148 with other inflammatory disease, and 50 HCsS100A9/S100A9 and IL–1β represent a novel positive feedback mechanism activating phagocytes during the pathogenesis of systemic-onset JIA.GermanyFrosch M et al., 2009 [104]
Analysis of serum from 20 patients with AOSD and 20 HCsS100A8/A9 may be involved in the inflammatory response with induction of proinflammatory cytokines and may serve as a clinicopathological marker for disease activity in AOSD.South KoreaKim HA et al., 2016 [105]
Analysis of serum from 240 patients (60 with SJIA, 17 with FMF, 18 with neonatal–onset multisystem inflammatory disease, 17 with Muckle–Wells syndrome, 45 with leukemia, 83 with systemic infection), and 45 HCsS100A12, a marker of granulocyte activation, is highly overexpressed in patients with systemic-onset JIA or FMF.GermanyWittkowski H et al., 2008 [106]
Analysis of serum from 37 patients with SLE and 38 HCsS100A12 levels showed strong correlations with known disease activity markers.South KoreaBae C-B et al., 2014 [107]
Analysis of serum from 35 patients with AOSD and 20 HCsSerum levels of cell-free DNA, myeloperoxidase-DNA complex, and α-defensin were significantly increased in patients with AOSD compared to HCs.South KoreaAhn MH et al., 2019 [27]
Analysis of serum from 37 SJIA patients without treatment, 32 with SJIA on treatment, and 16 HCsLevels of neutrophil granulocytes in serum reflect underlying disease activities of JIA.SwedenBacklund M et al., 2021 [108]
Analysis of 109 patients with AOSD (active 78 and inactive 31), 29 with SLE, 29 with RA, and 62 HCsNeutrophils-derived lipocalin–22 is higher in plasma and liver tissue in AOSD patients than in healthy controls. ChinaJia J et al., 2021 [109]
Analysis of serum from 38 patients with AOSD and 26 HCsFine-tuned mechanism between inflammatory (IL–18 induced NETs) and anti-inflammatory (microRNA–223) factors in AOSDTaiwanLiao T-L et al., 2021 [110]
Analysis of 30 patients with AOSDLDGs and NETs (HMGB–1 and LL–37) are increased in patients with active AOSD and correlate with cutaneous manifestations, arthritis and fever.MexicoTorres-Ruiz J et al., 2019 [111]
Analysis of serum from 40 patients with AOSD and 40 HCsSerum HMGB–1 levels were elevated in AOSD patients compared to the HCs and were correlated with CRP and systemic score.South KoreaJung J-Y et al., 2016 [112]
Analysis of serum from 12 patients with SJIA and 28 with other JIASerum HMGB–1 can be associated with clinical disease activity in JIA and is particularly at the highest level at the time of diagnosis.TaiwanXu D et al., 2021 [113]
Analysis of serum and synovial fluids from 99 patients with SJIA, 19 with SLE, and 27 HCsHMGB–1 and its soluble receptor RAGE in the blood and SF indicate that inflammation is triggered by alarmins in SJIA and SLECroatiaBobek D et al., 2014 [114]
Analysis of serum from 66 patients with AOSD and 40 HCsDemonstrated a close association between the increased levels of circulating NETs and organic involvement, as well as glucocorticoid responses in AOSD patients.ChinaJia Jin et al., 2020 [115]
Analysis of genotype from 164 patients with AOSD and 305 HCsFunctional LILRA3 is a novel genetic risk factor for the development of AOSD.ChinaWang M et al., 2021 [116]
Analysis of serum from 26 patients with SJIAThe level of serum histones extracted from NETs released by activated neutrophils has a positive correlation with the activity of SJIA.ChinaHu X et al., 2019 [117]
NET, neutrophil extracellular trap; SJIA, systemic juvenile idiopathic arthritis; AOSD, adult–onset Still’s disease; HC, healthy control; IL, interleukin; FMF, familial Mediterranean fever; DNA, deoxyribonucleic acid; SLE, systemic lupus erythematosus; RA, rheumatoid arthritis; RNA, ribonucleic acid; LDG, low–density granulocytes; HMGB–1, high mobility group box–1; RAGE, receptor for advanced glycation end products; CRP, C–reactive protein.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, J.-W.; Ahn, M.-H.; Jung, J.-Y.; Suh, C.-H.; Kim, H.-A. An Update on the Pathogenic Role of Neutrophils in Systemic Juvenile Idiopathic Arthritis and Adult-Onset Still’s Disease. Int. J. Mol. Sci. 2021, 22, 13038. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222313038

AMA Style

Kim J-W, Ahn M-H, Jung J-Y, Suh C-H, Kim H-A. An Update on the Pathogenic Role of Neutrophils in Systemic Juvenile Idiopathic Arthritis and Adult-Onset Still’s Disease. International Journal of Molecular Sciences. 2021; 22(23):13038. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222313038

Chicago/Turabian Style

Kim, Ji-Won, Mi-Hyun Ahn, Ju-Yang Jung, Chang-Hee Suh, and Hyoun-Ah Kim. 2021. "An Update on the Pathogenic Role of Neutrophils in Systemic Juvenile Idiopathic Arthritis and Adult-Onset Still’s Disease" International Journal of Molecular Sciences 22, no. 23: 13038. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222313038

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