Next Article in Journal
PD-L1 Expression in NSCLC: Clouds in a Bright Sky
Previous Article in Journal
Towards Effective Helicobacter pylori Eradication: Emerging Therapies in the Wake of Antibiotic Resistance
Previous Article in Special Issue
The Relationship Between Cholesterol Level, Cytokine Profile, and Arterial Stiffness in Young Patients with Uncomplicated Type 1 Diabetes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunopathology of Antineutrophil Cytoplasmic Antibody (ANCA)-Associated Vasculitis

by
Katarzyna Aleksandra Lisowska
* and
Anna Wardowska
*
Department of Pathophysiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(13), 6065; https://doi.org/10.3390/ijms26136065
Submission received: 15 May 2025 / Revised: 19 June 2025 / Accepted: 24 June 2025 / Published: 24 June 2025
(This article belongs to the Special Issue The Molecular Basis of Vascular Pathology)

Abstract

Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) constitutes a group of rare diseases characterized by autoimmune-associated inflammation and vessel damage. Based on the clinical manifestations and involvement of immune components, three disease syndromes are distinguished: granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and eosinophilic granulomatosis with polyangiitis (EGPA). In this review, we present the current data on the epidemiology, the clinical manifestations of each syndrome, and the most up-to-date classification criteria. The role of the underlying genetic and epigenetic abnormalities, as well as their interplay, is described. The immunological diversification of AAV is also described, with a focus on the immune cell dysfunctions detected in patients. In conclusion, we emphasize the urgent need to unravel the sophisticated mechanisms of this disease, which would enable the development of new, effective therapeutic strategies.

1. Introduction

Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a group of rare disorders [1,2] associated with the inflammation and damage of predominantly small- and medium-sized vessels [3,4]. Based on the clinical picture, AAV is divided into three disease syndromes: granulomatosis with polyangiitis (GPA), formerly known as Wegener’s granulomatosis; microscopic polyangiitis (MPA); and eosinophilic GPA (EGPA), formerly known as Churg–Strauss syndrome [3]. Each subtype has unique clinical and pathological features. MPA typically presents with glomerulonephritis and pulmonary capillaritis [4], while GPA and EGPA are predominantly characterized by respiratory tract involvement. Notably, GPA and EGPA are further distinguished by the presence of granulomatous inflammation [5]. The AAV pathogenesis is highly complex and results from a combination of overlapping factors that lead to immune tolerance breakdown. The multifactorial background of vasculitis includes genetic predisposition, environmental factors, sex hormones, and immunological mechanisms.
Genetic factors, such as variations in the HLA (human leukocyte antigen) or PRTN3 (proteinase 3) genes, play a crucial role [6]. However, epigenetic factors also contribute significantly to disease susceptibility. Among environmental factors, ultraviolet (UV) radiation [7], smoking [8], pesticides, fertilizers [9], and some other occupational exposures have been implicated as potential triggers.
This review aims to explore the mechanisms underlying the clinical manifestation of AAV, with a particular focus on genetic and cellular processes. In recent years, epigenetic mechanisms have been studied extensively, especially regarding their role in linking altered epigenetic patterns to pathological processes in the human body [10]. The dysregulation of these mechanisms can lead to changes in gene expression, disrupting immune homeostasis and promoting pathological autoimmune processes. Finally, we discuss the mechanisms contributing to the development of vasculitis cases without ANCA, highlighting the heterogeneity of these conditions and the need for further research.

2. Epidemiology

Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a relatively rare disease [1,2]. It is classified as an orphan disease under the number ORPHA:156152 on Orphanet [11]. However, the incidence of AAV has increased over time, plausibly due to the improvements in ANCA testing, disease classification, and clinical recognition [12,13]. Presently, AAV has an estimated prevalence of 200 cases per million people [1,2,14,15] and an incidence of about 20 per million person-years in Europe and North America [14,15].
According to the latest meta-analysis by Redondo-Rodriguez et al. [14], the highest incidence of AAV is observed in Minnesota and the lowest in Turkey. The AAV prevalence is also the highest in Minnesota. The pooled incidence and prevalence are greater for GPA than for MPA and EGPA. According to Watts et al. [15], MPA is the most common in Southern Europe and Japan, while GPA is more common in other populations. These studies show that there are differences regarding the variability between the northern and southern hemispheres and between continents, thus highlighting the ethnic and genetic differences. Some studies suggest that the incidence and prevalence may depend on the latitude. In 2009, Gatenby et al. [7] demonstrated that the incidence of AAV, especially GPA and EGPA, increases with increasing latitude and decreasing ambient UV radiation. A similar observation was made by Li et al. [1], which would suggest that low UV radiation exposure, possibly by influencing vitamin D levels, could, combined with other factors, stimulate AAV development [16].
In Poland, in 2014, a register of primary vasculitis was established as part of the Scientific Consortium of the Polish Vasculitis Registry (POLVAS). It is one of several such initiatives in Europe. The Consortium aims to collect epidemiological and clinical data on all forms of primary and secondary vasculitis, both retrospectively and prospectively. It conducted a multicenter retrospective study of all adult patients diagnosed with AAV between 1990 and 2016, identifying 625 AAV adults. It was found that 66.7% of patients from the POLVAS registry were diagnosed with GPA, 17.0% with MPA, and 16.3% with EGPA [17].

3. Clinical Manifestation

The clinical manifestations of AAV largely depend on the vascular bed affected, with the lungs and kidneys being the most common sites of involvement [18]. The disease can be limited to one organ at the time of presentation or involve multiple organs. Kidney involvement in AAV is widespread and can follow a slow or rapid progressive disease course, in time leading to dialysis.
All three disorders share symptoms of systemic inflammation, including weight loss, malaise, and fatigue. Approximately 50% of AAV patients also present arthralgia and myalgia [19]. Although various organs may be affected by AAV, and there is a significant clinical overlap between these diseases, each AAV demonstrates distinct pathological profiles. MPA typically presents with glomerulonephritis and pulmonary capillaritis, leading to interstitial pneumonitis and lung hemorrhage, which often worsens rapidly. Skin, nerves, and gastrointestinal tract involvement is common [19].
GPA and EGPA are characterized by a predominant respiratory tract involvement and can be further distinguished by granulomatous inflammation, primarily neutrophilic in GPA and eosinophilic in EGPA [5]. Patients frequently report symptoms of chronic rhinitis, sinusitis, or laryngitis [20]. Asthma is a typical early feature of EGPA [21]. Some studies have shown that heart and neurological involvement is highly prevalent in EGPA patients [17]. Meanwhile, 10–60% of GPA patients suffer from hyperplastic gingivitis and frequently experience ocular symptoms, including uveitis, ocular ulcers, episcleritis, and scleritis [19].

4. AAV Classification

The most widely used systemic vasculitis classification system was established at the 2012 International Chapel Hill Consensus Conference (CHCC). This system stratified vasculitis according to vessel size [3,22]. According to this system, AAV was then divided into three clinical diseases: granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and eosinophilic GPA (EGPA). These clinical phenotypes are characterized by vasculitic necrotizing lesions that predominantly affect arterioles, small arteries, capillaries, and venules [23,24].
The current classification criteria for GPA, MPA, and EGPA, developed by the ACR/EULAR (American College of Rheumatology/European Alliance of Associations for Rheumatology), were published in 2022 [25,26,27]. These criteria were the result of many years of collaborative work with rheumatologists, nephrologists, and immunologists, leading to the creation of weighted criteria for MPA [26], GPA [25], and EGPA [27].
In addition to the clinical symptoms and histopathological findings, ANCA antibodies are a helpful tool in recognizing the specific type of AAV (Table 1). ANCAs primarily target myeloperoxidase (MPO-ANCA) or proteinase 3 (PR3-ANCA) [28,29]. MPO-ANCAs may also react with other enzymes such as cathepsin G, elastase, lactoferrin, and lysozyme [30]. In immunofluorescence staining, PR3-ANCA shows a cytoplasmic pattern, hence the term c-ANCAs, while MPO-ANCA displays a perinuclear pattern (p-ANCA) [31]. c-ANCAs are mainly associated with GPA (75%) or renal-limited vasculitis, while p-ANCAs are more frequently found in MPA (60%). Several studies have shown that ANCAs, especially MPO-ANCA, are also present in approximately 50% of EGPA patients [3,28,32]. Despite this, the authors of the ACR/EULAR criteria did not include MPO-ANCA or PR3-ANCA positivity in the final criteria of EGPA, because these antibodies are significantly more prevalent in MPA and GPA [27]. Recent studies suggest that patients can also be positive for both types of antibodies [33]. These patients exhibit a combination of clinical characteristics of patients with both antibody types but are generally more similar to patients with MPO-ANCA.
Though ANCAs are crucial for the diagnosis of AAV, there are cases where antibodies are not detected [34]. Approximately 10% of GPA patients are ANCA-negative, mainly those with the disease limited to the upper and lower respiratory tract [35]. A similar case concerns MPA; ANCA-negative MPA patients usually present only renal symptoms or a less severe systemic disease [36]. Moreover, both GPA and MPA may become ANCA-positive in the course of the disease, suggesting a dynamic relationship between the autoantibody presence and disease progression [37]. Only half of EGPA patients are ANCA-positive, with positivity often correlating with a more vasculitic phenotype (vasculitic EGPA), while ANCA-negative cases are more frequently associated with eosinophilic tissue infiltration (non-vasculitic EGPA) [38]. Therefore, vasculitic EGPA usually presents with kidney involvement that quickly progresses and an alveolar hemorrhage. Meanwhile, non-vasculitic EGPA is characterized by cardiac involvement, pleural infusion, and gastroenteritis.
These observations imply that ANCAs are not essential for the initiation or pathogenesis of AAV. Additionally, their absence does not exclude the diagnosis, underscoring the importance of clinical evaluation and complementary diagnostic tools such as histopathology and imaging.

5. Genetic Background of AAV

5.1. HLA Genes and Their Association with AAV

Familial AAV cases have been reported for GPA [39] and EGPA [40]. Additionally, overlapping syndromes, mainly between GPA and EGPA, have been described [41]. However, a putative hereditary factor has never been definitively demonstrated. Studies show that the relative familial risk (RR 1.56) for GPA [42,43] is similar to that for rheumatoid arthritis (RA) (RR 1.5–5.0) but lower than that for other immune-mediated diseases [44].
Several studies have shown associations between HLA (human leukocyte antigen) class II loci and AAV, with variations dependent on the ethnic group and geographic region. The association has been particularly noted with HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQB1, and HLA-DRB1 [45]. For example, HLA-DPB1*0401 alleles have been associated with GPA in German patients [46,47]. As for the Chinese population, HLA-DRB1*1202 has been associated with GPA, while HLA-DRB1*1101 has been associated with MPA [48]. In Japanese patients, MPA was associated with HLA-DRB1*0901 [49]. HLA-DRB1*07 was associated with EGPA in the Italian population [50] and the German population [51]. However, in German patients, a stronger correlation was found for HLA-DRB1*04 [51].

5.2. Role of Non-HLA Genes in AAV

Several non-HLA gene associations with AAV have been found, particularly genes involved in immune responses and ANCA generation. These include polymorphisms in CTLA4 (cytotoxic T lymphocyte-associated antigen-4), TLRs (Toll-like receptors), PTPN22 (protein tyrosine phosphatase, non-receptor type 22), SERPINA1 (serine protease inhibitor), and PRTN3 (proteinase 3) [6].
CTLA-4, a member of the CD28 receptor family, is a regulatory molecule expressed on the surface of T cells. It plays a significant role in maintaining peripheral tolerance by inhibiting autoreactive T cells, thus preventing autoimmune diseases [52]. Inhibiting T-cell activity is mediated through the binding with CD80 (B7-1) and CD86 (B7-2) present on antigen-presenting cells (APCs) [53]. Several studies have linked single-nucleotide polymorphisms (SNPs) in the CTLA4 gene to AAV, especially GPA. A British study found that the allele (A) of rs3087243 SNP has a protective effect against AAV [54]. Polymorphisms in exon 1 (+49) and 4 (CT60) were also associated with AAV in a separate British cohort [55].
TLRs, which are present on immune cells and are involved in pathogen recognition and antigen presentation, have been implicated to be associated with several autoimmune diseases, including AAV. Different TLRs recognize different pathogen-associated molecular patterns (PAMPs). Evidence suggests that TLR9, which recognizes unmethylated CpG motifs in bacterial nucleic acid, may be involved in AAV development. In a German study, four different SNPs in the TLR9 gene (rs352162, rs352140, rs352139, and rs5743836) were strongly associated with GPA [56]. While similar findings were observed for MPA, the associated SNPs had the contrary alleles. No association was found for EGPA. The authors could not confirm their results in Dutch and British AAV cohorts.
Polymorphisms in PTPN22, a gene encoding an intracellular tyrosine phosphatase that inhibits T-cell activation, are also linked with autoimmune diseases. The first study investigating the effect of SNPs in this gene revealed a strong association between rs2476601 and GPA, especially in ANCA-positive patients from the German population [57]. Similar results were later reported in British GPA and MPA patients [54]. In an Italian study, the 620 W SNP was found to be significantly associated with ANCA-positive GPA [58].
PRTN3, which encodes proteinase 3 (PR3), one of the main ANCA targets, is another gene linked to AAV. In Swedish patients, the A564G variation in the promoter region was found to be associated with GPA [59]. Additionally, in the Egyptian population, SNP rs62132295 and rs62132293 were significantly associated with AAV, especially PR3-AAV and GPA [60].
Finally, SERPINA1, which encodes α1-antitrypsin (α1-AT), the primary inhibitor of PR3, has been also implicated in AAV. Variants leading to reduced α1-AT function may result in persistent PR3 activity, promoting tissue damage and AAV development. Some studies showed that heterozygous patients for the Z variant (Glu342Lys) of SERPINA1 have an increased risk of developing GPA compared to the general population [61,62].

5.3. Insights from Genome-Wide Association Studies (GWASs)

Genome-wide association studies (GWASs) have provided insights into the genetic background of AAV, particularly GPA and MPA [63,64,65]. Several studies have confirmed associations between AAV and specific genetic loci, with a stronger correlation observed for antibody subtypes than for clinical manifestations. In one of the first GWAS studies, associations with GPA and MPA were identified at three loci: HLA-DPB1, SERPINA 1, and PRTN3 [64]. Among these, PR3-ANCA positivity emerged as the strongest factor linked to these SNPs. This finding highlighted the role of the immune system and genetic predisposition in shaping disease subtypes. The second study by Xie et al. [65] showed an association between the HLA-DPB1 and HLA-DPA1 loci and a novel locus, SEMA6A, particularly in GPA patients. As in the earliest studies, these associations were most potent in the PR3-ANCA-positive patients, emphasizing the distinct genetic signatures of the PR3-associated disease.
Interestingly, despite early hypotheses, GWAS failed to confirm the association between CTLA4 SNPs and AAV, suggesting a more limited role for this gene in disease susceptibility.
Further expanding this genetic framework, most recent GWAS studies identified a strong association between HLA-DQ and EGPA, particularly MPO-ANCA EGPA [63]. This highlights the heterogeneity within AAV subtypes and underscores the importance of integrating genetic data with clinical and serological profiles for improved disease classification and understanding.
In recent years, epigenetic mechanisms have been extensively studied, leading to a deeper understanding of the complex relationship between altered epigenetic patterns and pathological processes [10,66,67]. Epigenetics, broadly defined, refers to mechanisms that may alter the gene expression without changes in the DNA nucleotide sequence. Three essential modifications, collectively referred to as the epigenetic triad, form the foundation of epigenetics: DNA methylation, chromatin remodeling through histone modifications, and non-coding RNAs [68]. Dysregulated epigenetic mechanisms can disrupt gene expression and destabilize immune homeostasis, contributing to the development of autoimmune diseases [10,69].
The intricate interplay between these epigenetic mechanisms and their contributions to autoimmune pathogenesis is exemplified by studies on AAV [70,71]. Although the role of epigenetics in AAV remains relatively underexplored, initial findings highlight its significance in investigating epigenetic modifications in neutrophils from AAV patients and observed that the levels of the chromatin modification H3K27me3 were depleted at PRTN3 and MPO loci. This finding suggests that the epigenetic modifications of ANCA autoantigen-encoding genes may contribute to the inappropriate expression of PR3 and MPO in AAV. A complementary work by Yang et al. investigated the expression of proteins responsible for histone modification, euchromatic histone-lysine N-methyltransferase 1 and 2 (EHMT1, EHMT2), and male sex lethal 1 homolog and insulin growth factor (MSL1 and ING4) [72]. They found that EHMT1 and EHMT2 are associated with H3K9me2, responsible for gene silencing, and their expression in granulocytes from AAV patients is decreased compared with healthy people. Meanwhile, the expression of MSL1, associated with H4K16ac which activates genes, was overexpressed. Jones et al. [70] demonstrated that AAV patients with an active disease display the hypomethylation of MPO and PRTN3 genes, leading to the increased expression of these autoantigens, while, in remission, gene methylation increases.
In general, these works represent a first and significant step in linking the epigenetic modifications and disease phenotype in AAV. Unfortunately, there is too little research to precisely determine the importance of epigenetics in the development of AAV.

6. Immunopathomechanism in AAV

Immunologically, GPA and MPA are driven by aberrantly activated neutrophils, while EGPA is characterized by eosinophilic infiltration [5]. These cells are primed by specific autoantibodies targeted at neutrophil autoantigens—PR3 and MPO [28,29]. After reaching the vascular endothelium, activated cells release pro-inflammatory mediators. However, immunologically, AAVs are much more complex. Not only innate but also acquired response mechanisms are responsible for the development of the disease.

6.1. The Role of ANCA and Neutrophils

ANCA antigens, specifically PR3 and MPO, are primarily found in the granules of neutrophils. Eosinophils also express PR3 [73], together with eosinophil peroxidase (EPO), which shares a high structural similarity (69.8% amino acid) to MPO [74]. MPO and EPO are enzymes that catalyze the formation of hypochlorous acid, a potent chemical compound with strong antibacterial and antiviral properties [75]. PR3 is a serine protease enzyme responsible for degrading extracellular proteins, playing a role in immune defense and tissue remodeling [76].
Under healthy conditions, PR3 and MPO are stored in intracellular granules of neutrophils. However, in response to specific stimuli, these enzymes can be expressed on the surface of neutrophils [77] or monocytes [78]. The PR3 expression on neutrophils is bimodal with two distinct populations: one with a low and one with a high PR3 expression [78]. When neutrophils with a high PR3 expression are stimulated with PR3-ANCA, they generate higher amounts of superoxide, a reactive oxygen species (ROS), compared to cells with a lower PR3 expression. Neutrophils and monocytes can also express minimal surface MPO, with its expression increasing following cell mobilization [77,79].
For AAV to develop, there must first be the loss of the immunological tolerance of T and B cells towards MPO, which is necessary for the production of autoantibodies that play a role in vasculitis (Figure 1). Tan et al. showed that the deletion of autoreactive anti-MPO T cells in the thymus and the presence of peripheral regulatory T cells (Tregs) that suppress potentially autoreactive MPO-specific CD4+ T cells protect mice against glomerulonephritis (GN) [80]. MPO-ANCA-induced GN in mice is more aggressive in the presence of MPO-specific CD4+ T cells [81]. The role of PR3 is more complicated due to its presence on the surface of neutrophils. According to some studies, when non-obese diabetic (NOD) mice are injected with PR3, this results in the increased production of PR3-ANCA without inducing AAV manifestation, which suggests that the presence of antibodies alone is not sufficient to cause the disease [82]. Recently, Sharma et al. identified in the blood of AAV patients CD4+ T cells producing interferon-gamma (IFN-γ) in response to stimulation with PR3 [83].
ANCAs activate neutrophils and monocytes by engaging their Fc receptors after binding MPO or PR3 [84], or by Fab’2 binding MPO and PR3 expressed on their surface [85]. Additionally, the aberrant glycosylation of MPO- and PR3-specific ANCAs has been shown to enhance their ability to interact with activating Fc receptors on neutrophils, thereby increasing their immune-activating potential [86].
In vitro studies have demonstrated that ANCAs stimulate neutrophils to release ROS and undergo degranulation [87]. However, in response to ANCA binding, neutrophils can also release neutrophil extracellular traps (NETs), which are part of a defense mechanism against infections. In healthy conditions, NETs are released from activated neutrophils in response to pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) [88]. NETs are unique net-like structures composed of DNA strands, histones, and antimicrobial proteins such as MPO, cathepsin, and elastase [89], which are all autoantigens recognized by ANCA [30]. In vitro studies confirmed that ANCA-induced NETs are responsible for the damage of endothelial cells (ECs) [90,91] mediated through complement activation [91]. Moreover, in a mouse model, it was shown that PR3 and MPO from NETs can also be transferred to dendritic cells (DCs) [92]. The immunization of mice with these NET-loaded DCs led to ANCA production and subsequent kidney damage, suggesting a potential pathophysiological role for NETs in the development of AAV.
Kessenbrock et al. observed NET formation in the kidneys of patients with small-vessel vasculitis (SVV) and further demonstrated that ANCA-stimulated neutrophils release NETs containing PR3 and MPO [93]. Some authors demonstrated the presence of NETs in other organs affected by AAV, like the lungs [94], or the nervous system [95]. In healthy people, NET-DNA is processed by deoxyribonuclease (DNase) I [96]. NETs, mainly their remnants, are found in the blood of AAV patients [97]. At the same time, studies showed that the activity of DNase I is reduced in AAV patients which decreases NET degradation and makes PR3 and MPO available for ANCA [98].
At this point, it should be added that MPO-ANCA, which is so important for the pathogenesis of AAV, has also been found in other autoimmune diseases, including systemic lupus erythematosus (SLE) and Sjögren’s syndrome (SS) [99]. However, in the majority of those patients, ANCA did not recognize p-ANCA-related antigens, and their role in systemic autoimmune diseases is currently unknown [99]. Both types of ANCA were also shown in infective endocarditis, tuberculosis, primary sclerosing cholangitis, and interstitial lung diseases [100]. Moreover, studies showed that patients with idiopathic pulmonary fibrosis who were MPO-ANCA-positive can develop MPA [101]. Such observations suggest that the presence of ANCA in certain clinical situations may predict the development of AAV.
In vivo, AAV patients exhibit an increased percentage of polymorphonuclear leukocytes (PMNs) in their blood compared with healthy people [102]. Interestingly, there are no significant differences in the expression of certain surface antigens, such as CD88, CD11c, or CD62L, between patients and healthy controls. CD88 plays the role of a receptor for 5a, which is a protein fragment released from the cleavage of the complement component C5. C5a is an anaphylatoxin that causes an increased expression of adhesion molecules on the endothelium, the contraction of smooth muscle, and increased vascular permeability [103]. It also stimulates the release of inflammatory mediators. In AA, C5a with its receptor plays an essential role in ANCA-mediated neutrophil recruitment and activation [104]. CD62L is a cell surface protein that plays a crucial role in the adhesion and migration of leukocytes, including neutrophils, which can contribute to leucocyte endothelial migration [105]. CD11c is an integrin present in dendritic cells, monocytes, macrophages, and neutrophils, and is associated with antigen presentation [106]. Although there is no indication of changes in the expression of these antigens in AAV patients, they still may mediate antibody-dependent vascular injury.

6.2. The Role of Monocytes in AAV

In healthy individuals, monocytes express low levels of ANCA antigens on their surface [24]. However, in response to pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), the surface expression of ANCA antigens can be upregulated during infection, which makes them available for ANCA antibodies. Indeed, Hattar et al. [107] demonstrated that the pre-incubation of monocytes with PR3-ANCA enhanced the production of interleukin 8 (IL-8), tumor necrosis factor-alpha (TNFα), and IL-6 in response to lipopolysaccharide (LPS) and lipoteichoic acid (LTA), both of which are components of the bacterial cell wall and, as such, serve as PAMPs. This response was associated with the upregulation of the CD14 antigen, a receptor detecting LPS, and a co-receptor of TLR4, suggesting that infections may promote AAV development.
Monocytes can also respond to ANCA by producing ROS [108]. However, a notable reduction in ROS production is observed in AAV patients. Moreover, decreased ROS production correlates with disease activity; patients in remission show higher ROS production than those with an active disease. Furthermore, PMNs from MPA patients showed a lower oxidative burst capacity than cells from GPA and EGPA patients [102].
Monocytes are categorized into three subsets based on the expression of CD14 and CD16 antigen (Fc gamma receptor III): classical (CD14++CD16−), intermediate (CD14++CD16+), and non-classical (CD14+CD16++). Tarzi et al. [109] demonstrated that monocytes of AAV patients with an active disease showed an increased expression of CD14 antigen compared to those in remission, without any changes in proportions between these three subsets. The expression of CD14 was strongly correlated with the expression of ANCA antigens, particularly PR3. Additionally, Matsumoto et al. [110] observed a higher proportion of intermediate monocytes in AAV patients compared to healthy people, regardless of the underlying cause of AAV. AAV patients also exhibited elevated levels of CD62L in monocytes compared to healthy individuals [102]. Moreover, an increased expression of CD11b and CD64 (a high-affinity Fcγ receptor) was observed in monocytes from patients with active GPA, suggesting heightened monocyte activation [111]. Furthermore, these patients demonstrated an increased expression of integrins such as CD29, CD18, CD11a, and CD11b, which are crucial for leukocyte–endothelium interaction and the process of leukocyte diapedesis [112].

6.3. Eosinophils as Essential Cells in EGPA

In the case of EGPA, the type of cells involved in inflammatory processes depends on the presence of ANCA [113]. While, in ANCA-positive EGPA, similar to GPA and MPA, neutrophils play an important role, in ANCA-negative EGPA, eosinophils are the predominant immune cells driving inflammation [114]. Eosinophil activation in ANCA-negative EGPA is influenced by type 2 innate lymphoid cells (ILC2) and Th2 cells, which produce IL-5 [115]. IL-5 is a key cytokine responsible for the survival, activation, and recruitment of eosinophils [116]. The pivotal role of IL-5 in EGPA pathogenesis is supported by the clinical efficacy of mepolizumab, a humanized monoclonal antibody targeting IL-5, which prevents eosinophil activation and reduces inflammation [117]. In recognition of its therapeutic benefits, mepolizumab was approved by the US Food and Drug Administration (FDA) in 2017 for the treatment of EGPA.
Upon activation, eosinophils release their granule contents, which include a variety of cytotoxic proteins that contribute to tissue damage and disease progression. These proteins include the major basic protein (MBP), eosinophil cationic protein (ECP), eosinophil peroxidase (EPO), and eosinophil-derived neurotoxin (EDN). Their combined effects lead to cellular damage, endothelial dysfunction, and organ involvement, particularly in the lungs and heart [114]. Moreover, recent studies show that eosinophils release eosinophil extracellular traps (EETs) [40] that can be observed in inflamed tissues in the course of EGPA [118]. EETs, similar to NETs, contain mitochondrial DNA and granule proteins, as mentioned above, and galectin-10. Galectin-10 forms Charcot–Leyden crystals that persist in tissues where they activate inflammasome and sustain inflammation [119].

6.4. Importance of DCs

In the vasculitis granuloma, neutrophils and monocytes containing ANCA-specific antigens are surrounded by antigen-presenting cells (APCs), T cells, and B cells [120]. DCs appear to play a central role in the pathogenesis of AAV, as they can drive an inflammatory response to autoantigens by presenting them. The dysregulated activity of circulating DCs has been closely linked to disease progression.
After taking up antigens from neutrophil extracellular traps (NETs), DCs become activated and can initiate autoimmune inflammation and ANCA production [92]. NET-induced DC activation is characterized by an increased TLR4 surface expression and enhanced release of pro-inflammatory cytokines [121]. As TLR4 is responsible for recognizing damage-associated molecular patterns (DAMPs), its expression abnormalities may significantly impact inflammation and tissue damage in the course of AAV. In the experimental animal model, mice immunized with DCs loaded with NETs showed an increase in serum ANCA [92]. The induction of ANCA was accompanied by glomerulonephritis or tubulointerstitial nephritis with vascular neutrophilic infiltration.
Increased spontaneous NET production and the upregulation of the TLR4 receptor have been reported in patients with active MPO-AAV. Interestingly, it was linked with the decreased expression of TIM-3, a co-inhibitory receptor that regulates the T-cell immune response. Su et al. confirmed the regulatory role of TIM-3 in the NET-mediated DC activation in AAV by introducing a TIM-3 blockade in animal experiments [121].
The reduced numbers of circulating DCs (DCs) [122] and plasmacytoid DCs (pDCs) are a commonly reported feature of patients with an active disease [123]. Braudeau et al. [124] demonstrated that conventional DCs (cDCs) from patients with GPA and MPA in the active stage produce significantly lower levels of IL-12 and IL-23p40 following stimulation via TLR3, 4, or 7/8, compared with those from patients in remission or healthy individuals. No such differences were observed in plasmacytoid DCs (pDCs) activity. However, pDCs from MPO-ANCA patients with rapidly progressive glomerulonephritis exhibited a reduced ability to produce IFN-α upon stimulation. This alteration in pDC function was described as a long-term phenomenon, as it persisted even after several years of AAV treatment. The authors attempted to explain the reduced number of circulating pDCs by the migration of this subpopulation to the kidneys, as revealed by an immunohistological analysis [123]. Wilde et al. [125] also reported the presence of both mature (CD208) and immature (CD209) DCs in renal biopsies from AAV patients, emphasizing the critical role of these cells in the inflammatory process.
The study by Tsurikisawa et al. reported that monocyte-derived DCs may be associated with the activity of EGPA, as this group of patients exhibited higher percentages of CD83+ DCs in remission. The elevated CD83+ DCs showed a positive correlation with naturally occurring (nTreg) and inducible Tregs (iTreg), thus implicating a plausible way to induce remission in patients with EGPA [126]. Other literature data also highlight the therapeutic role of DCs in AAV management, including MPO-presenting tolerogenic DCs. However, further studies are required to confirm the precise mechanisms of DCs’ action in AAV and the potential effectiveness of DC-based therapy.

6.5. Lymphocytes in AAV

The loss of immunological tolerance towards PR3 or MPO, key autoantigens in AAV, is fundamental to the disease’s development. This breakdown in tolerance is perpetuated by a positive feedback loop of autoreactivity, wherein primed neutrophils or eosinophils release autoantigens while causing vascular damage. The deposition of these autoantigens at injury sites makes them accessible for APCs, which further facilitates antigen recognition by autoreactive T cells. These activated T cells then mediate additional tissue damage. Moreover, AAV patients exhibit a dysfunction of regulatory T cells (Tregs), which exacerbates the loss of tolerance and the emergence of pathogenic ANCA responses.
The course of the immune reaction in AAV, in particular, the type of cells involved in the process of tissue destruction, largely depends on the type of T cells. Naïve helper T cells (Th cells), depending on the cytokines produced by DCs, can differentiate into various subpopulations, including Th1, Th2, Th17, and Tregs (regulatory T cells). This is important in EGPA, where, depending on the type of Th cells, other mechanisms are involved in the development of tissue inflammation. In ANCA-positive EGPA, Th1 and Th17 dominate [127]; they release pro-inflammatory cytokines that activate neutrophils [128] and B cells [129]. The activated neutrophils then expose MPO, and, less frequently, PR3, which is recognized by ANCA. The binding of MPO-ANCA causes NET formation and the release of ROS and proteolytic enzymes that trigger endothelial damage [93]. Meanwhile, in ANCA-negative EGPA, Th2 cells and their cytokines, especially IL-5, play a central role in tissue destruction [130]. As mentioned above, IL-5 is responsible for priming eosinophils to produce enzymesm causing tissue damage, especially in the lungs and heart.
In GPA, the type of Th cells involved in pathogenesis determines whether the disease is localized or generalized [130,131]. In GPA limited to nasal granulomatous lesions, IFN-γ, a marker of the Th1 subset, dominates [105], while, in generalized disease, a high IL-4 expression was observed, suggesting Th2 involvement [132]. The type of Th response is also associated with the disease phase; several authors demonstrated that GPA remission is characterized by Th2 dominance [133,134]. Less is known about T-cell polarization in MPA but it was suggested that it is rather associated with Th1 dominance, especially in patients with the active phase of the disease [135].
Additionally, several studies have highlighted the altered immune cell composition in AAV patients. For instance, AAV patients exhibit increased proportions of activated T cells with HLA-DR [116] or CD25 [136,137] expression compared to healthy controls. In GPA patients, the expression of CD28 antigen, which is an essential co-stimulatory molecule, is significantly decreased in T cells [138]. T cells without any CD28 expression present in those patients are a source of pro-inflammatory cytokines [139] and have the killer cell phenotype [140].
A significant expansion of Th17 cells and a decrease in Tregs have been demonstrated in AAV patients [141]. Th17 expansion may be driven by activated monocytes through TLR ligand interactions, a phenomenon observed in other autoimmune diseases [142]. Increased frequencies of Th17 have been reported in EGPA patients during disease relapse [143]. In GPA, the presence of Th17 was independent of disease activity [144]. The suppressive function of Tregs is significantly impaired, especially during an active disease, as reported by Kitching et al. [42], but tends to normalize during periods of remission [134].
The role of natural killer (NK) cells in AAV remains uncertain [145], with some studies showing a reduction in NK cell numbers or percentages, while others report an increase. Some authors found that NK cells from GPA patients are characterized by a higher expression of the early activation marker CD69, suggesting an altered NK cell response in AAV [146].
B cells are integral to the pathogenesis of AAV, primarily through their role as precursors of antibody-secreting plasma cells [147]. These cells regulate immune responses by inhibiting T-cell proliferation and the production of pro-inflammatory cytokines, such as interferon-γ (IFN-γ), TNF-α, and IL-17 [148]. Additionally, B cells function as APCs, initiating T-cell responses by providing costimulatory signals and producing cytokines and growth factors [149]. The significance of B cells in AAV pathogenesis is further supported by the success of B-cell depletion therapies, such as rituximab (RTX). RTX is as effective as CYC treatment in inducing remission in severe AAV cases and maybe even more effective in preventing relapses [150,151]. Importantly, the reappearance of B cells and the return of ANCA positivity following RTX treatment are associated with a higher risk of AAV relapse [152]. Specific changes in the frequencies of B cells were also reported, including a decrease in the percentage of transitional B cells and an increase in switched memory B cells and plasmablasts, reflecting an adaptive immune response that sustains the inflammatory process in AAV.

7. Conclusions

The comparison of all three AAV subtypes is summarized in Table 2. As shown, although considerable progress has been made in elucidating the genetic and cellular mechanisms underlying AAV, many critical aspects of its pathogenesis remain poorly understood. In particular, the role of epigenetic regulation in the initiation and progression of the disease is still largely unexplored. Additionally, the contribution of specific immune cell populations—such as dendritic cells (DCs) and natural killer (NK) cells—remains insufficiently characterized, despite the growing evidence of their involvement in other autoimmune and inflammatory conditions. A deeper understanding of these factors could significantly enhance our ability to identify biomarkers for disease activity and prognosis, as well as uncover novel therapeutic targets. Ultimately, continued research into these underexplored areas holds promise for the development of more precise and effective treatment strategies for patients with AAV.

Author Contributions

Conceptualization, K.A.L. and A.W.; writing—review and editing, K.A.L. and A.W. 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. Li, J.; Cui, Z.; Long, J.; Huang, W.; Wang, J.; Wang, H.; Zhang, L.; Chen, M.; Zhao, M. The Frequency of ANCA-Associated Vasculitis in a National Database of Hospitalized Patients in China. Arthritis Res. Ther. 2018, 20, 226. [Google Scholar] [CrossRef] [PubMed]
  2. Berti, A.; Cornec, D.; Crowson, C.S.; Specks, U.; Matteson, E.L. The Epidemiology of Antineutrophil Cytoplasmic Autoantibody–Associated Vasculitis in Olmsted County, Minnesota: A Twenty-Year US Population–Based Study. Arthritis Rheumatol. 2017, 69, 2338–2350. [Google Scholar] [CrossRef]
  3. Coattrenec, Y.; Muller, Y.D.; Spoerl, D.; Lobrinus, J.A.; Seebach, J.D. Prevalence of large vessel vasculitis in ANCA-associated vasculitis: A retrospective cohort study. Rheumatol. Int. 2021, 41, 2147–2156. [Google Scholar] [CrossRef] [PubMed]
  4. Kaymakci, M.S.; Elfishawi, M.M.; Langenfeld, H.E.; Hanson, A.C.; Crowson, C.S.; Bois, M.C.; Ghaffar, U.; Koster, M.J.; Specks, U.; Warrington, K.J. Large vessel involvement in antineutrophil cytoplasmic antibody-associated vasculitis. Rheumatology 2024, 63, 1682–1689. [Google Scholar] [CrossRef]
  5. Cartin-Ceba, R.; Peikert, T.; Specks, U. Pathogenesis of ANCA-Associated Vasculitis. Curr. Rheumatol. Rep. 2012, 14, 481–493. [Google Scholar] [CrossRef]
  6. Bonatti, F.; Reina, M.; Neri, T.M.; Martorana, D. Genetic Susceptibility to ANCA-Associated Vasculitis: State of the Art. Front. Immunol. 2014, 5, 577. [Google Scholar] [CrossRef] [PubMed]
  7. Gatenby, P.A.; Lucas, R.M.; Engelsen, O.; Ponsonby, A.; Clements, M. Antineutrophil Cytoplasmic Antibody–Associated Vasculitides: Could Geographic Patterns Be Explained by Ambient Ultraviolet Radiation? Arthritis Rheum. 2009, 61, 1417–1424. [Google Scholar] [CrossRef]
  8. McDermott, G.; Fu, X.; Stone, J.H.; Wallwork, R.; Zhang, Y.; Choi, H.K.; Wallace, Z.S. Association of Cigarette Smoking With Antineutrophil Cytoplasmic Antibody–Associated Vasculitis. JAMA Intern. Med. 2020, 180, 870. [Google Scholar] [CrossRef]
  9. Lane, S.E.; Watts, R.A.; Bentham, G.; Innes, N.J.; Scott, D.G.I. Are Environmental Factors Important in Primary Systemic Vasculitis?: A Case–Control Study. Arthritis Rheum. 2003, 48, 814–823. [Google Scholar] [CrossRef]
  10. Mazzone, R.; Zwergel, C.; Artico, M.; Taurone, S.; Ralli, M.; Greco, A.; Mai, A. The Emerging Role of Epigenetics in Human Autoimmune Disorders. Clin. Epigenetics 2019, 11, 34. [Google Scholar] [CrossRef]
  11. Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis. Available online: https://www.orpha.net/en/disease/detail/156152 (accessed on 11 June 2025).
  12. Rathmann, J.; Segelmark, M.; Englund, M.; Mohammad, A.J. Stable Incidence but Increase in Prevalence of ANCA-Associated Vasculitis in Southern Sweden: A 23-Year Study. RMD Open 2023, 9, e002949. [Google Scholar] [CrossRef]
  13. Pearce, F.A.; Lanyon, P.C.; Grainge, M.J.; Shaunak, R.; Mahr, A.; Hubbard, R.B.; Watts, R.A. Incidence of ANCA-Associated Vasculitis in a UK Mixed Ethnicity Population. Rheumatology 2016, 55, 1656–1663. [Google Scholar] [CrossRef]
  14. Redondo-Rodriguez, R.; Mena-Vázquez, N.; Cabezas-Lucena, A.M.; Manrique-Arija, S.; Mucientes, A.; Fernández-Nebro, A. Systematic Review and Metaanalysis of Worldwide Incidence and Prevalence of Antineutrophil Cytoplasmic Antibody (ANCA) Associated Vasculitis. J. Clin. Med. 2022, 11, 2573. [Google Scholar] [CrossRef]
  15. Watts, R.A.; Mahr, A.; Mohammad, A.J.; Gatenby, P.; Basu, N.; Flores-Suárez, L.F. Classification, Epidemiology and Clinical Subgrouping of Antineutrophil Cytoplasmic Antibody (ANCA)-Associated Vasculitis. Nephrol. Dial. Transplant. 2015, 30, i14–i22. [Google Scholar] [CrossRef]
  16. Gatenby, P.A. Anti-neutrophil Cytoplasmic Antibody-associated Systemic Vasculitis: Nature or Nurture? Intern. Med. J. 2012, 42, 351–359. [Google Scholar] [CrossRef]
  17. Wójcik, K.; Wawrzycka-Adamczyk, K.; Włudarczyk, A.; Sznajd, J.; Zdrojewski, Z.; Masiak, A.; Czuszyńska, Z.; Majdan, M.; Jeleniewicz, R.; Klinger, M.; et al. Clinical Characteristics of Polish Patients with ANCA-Associated Vasculitides—Retrospective Analysis of POLVAS Registry. Clin. Rheumatol. 2019, 38, 2553–2563. [Google Scholar] [CrossRef]
  18. Almaani, S.; Fussner, L.A.; Brodsky, S.; Meara, A.S.; Jayne, D. ANCA-Associated Vasculitis: An Update. J. Clin. Med. 2021, 10, 1446. [Google Scholar] [CrossRef]
  19. Jayne, D. The diagnosis of vasculitis. Best Pract. Res. Clin. Rheumatol. 2009, 23, 445–453. [Google Scholar] [CrossRef]
  20. Solans-Laqué, R.; Fraile, G.; Rodriguez-Carballeira, M.; Caminal, L.; Castillo, M.J.; Martínez-Valle, F.; Sáez, L.; Rios, J.J.; Solanich, X.; Oristrell, J.; et al. Clinical characteristics and outcome of Spanish patients with ANCA-associated vasculitides: Impact of the vasculitis type, ANCA specificity, and treatment on mortality and morbidity. Medicine 2017, 96, e6083. [Google Scholar] [CrossRef]
  21. Puan, Y.; Ong, K.Y.; Tiew, P.Y.; Wen Chen, G.X.; Teo, N.W.Y.; Low, A.H.L.; Wechsler, M.E.; Koh, M.S. Characteristics of Severe Asthma Clinic Patients with Eosinophilic Granulomatosis with Polyangiitis. J. Allergy Clin. Immunol. Pract. 2025, 13, 361–368.e2. [Google Scholar] [CrossRef]
  22. Jennette, J.C.; Falk, R.J.; Bacon, P.A.; Basu, N.; Cid, M.C.; Ferrario, F.; Flores-Suarez, L.F.; Gross, W.L.; Guillevin, L.; Hagen, E.C.; et al. 2012 Revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum. 2013, 65, 1–11. [Google Scholar] [CrossRef]
  23. Renauer, P.; Coit, P.; Sawalha, A.H. Epigenetics and Vasculitis: A Comprehensive Review. Clin. Rev. Allergy Immunol. 2016, 50, 357–366. [Google Scholar] [CrossRef] [PubMed]
  24. Brunini, F.; Page, T.H.; Gallieni, M.; Pusey, C.D. The Role of Monocytes in ANCA-Associated Vasculitides. Autoimmun. Rev. 2016, 15, 1046–1053. [Google Scholar] [CrossRef] [PubMed]
  25. Robson, J.C.; Grayson, P.C.; Ponte, C.; Suppiah, R.; Craven, A.; Judge, A.; Khalid, S.; Hutchings, A.; Watts, R.A.; Merkel, P.A.; et al. 2022 American College of Rheumatology/European Alliance of Associations for Rheumatology Classification Criteria for Granulomatosis with Polyangiitis. Ann. Rheum. Dis. 2022, 81, 315–320. [Google Scholar] [CrossRef] [PubMed]
  26. Suppiah, R.; Robson, J.C.; Grayson, P.C.; Ponte, C.; Craven, A.; Khalid, S.; Judge, A.; Hutchings, A.; Merkel, P.A.; Luqmani, R.A.; et al. 2022 American College of Rheumatology/European Alliance of Associations for Rheumatology Classification Criteria for Microscopic Polyangiitis. Ann. Rheum. Dis. 2022, 81, 321–326. [Google Scholar] [CrossRef]
  27. Grayson, P.C.; Ponte, C.; Suppiah, R.; Robson, J.C.; Craven, A.; Judge, A.; Khalid, S.; Hutchings, A.; Luqmani, R.A.; Watts, R.A.; et al. 2022 American College of Rheumatology/European Alliance of Associations for Rheumatology Classification Criteria for Eosinophilic Granulomatosis with Polyangiitis. Ann. Rheum. Dis. 2022, 81, 309–314. [Google Scholar] [CrossRef]
  28. Geetha, D.; Jefferson, J.A. ANCA-Associated Vasculitis: Core Curriculum 2020. Am. J. Kidney Dis. 2020, 75, 124–137. [Google Scholar] [CrossRef]
  29. Cohen Tervaert, J.W.; Damoiseaux, J. Antineutrophil Cytoplasmic Autoantibodies: How Are They Detected and What Is Their Use for Diagnosis, Classification and Follow-Up? Clin. Rev. Allerg. Immunol. 2012, 43, 211–219. [Google Scholar] [CrossRef]
  30. Chejfec, G. Advanced Atlas of Autoantibody Patterns. Arch. Pathol. Lab. Med. 2000, 124, 472. [Google Scholar] [CrossRef]
  31. Radice, A.; Sinico, R.A. Antineutrophil Cytoplasmic Antibodies (ANCA). Autoimmunity 2005, 38, 93–103. [Google Scholar] [CrossRef]
  32. Elmér, E.; Smargianaki, S.; Pettersson, Å.; Skattum, L.; Ohlsson, S.; Hellmark, T.; Johansson, Å.C.M. Increased Frequencies of Switched Memory B Cells and Plasmablasts in Peripheral Blood from Patients with ANCA-Associated Vasculitis. J. Immunol. Res. 2020, 2020, 8209737. [Google Scholar] [CrossRef] [PubMed]
  33. Gong, Y.; Shen, C.; Meng, T.; Lin, W.; Hu, X.; Tang, R.; Xiong, Q.; Ooi, J.D.; Eggenhuizen, P.J.; Chen, J.; et al. Clinical Features and Prognosis of ANCA-Associated Vasculitis Patients Who Were Double-Seropositive for Myeloperoxidase-ANCA and Proteinase 3-ANCA. Clin. Exp. Med. 2024, 24, 66. [Google Scholar] [CrossRef] [PubMed]
  34. Falk, R.J.; Hoffman, G.S. Controversies in Small Vessel Vasculitis—Comparing the Rheumatology and Nephrology Views. Curr. Opin. Rheumatol. 2007, 19, 1–9. [Google Scholar] [CrossRef]
  35. Ohteru, Y.; Hamada, K.; Oishi, K.; Suizu, J.; Harada, M.; Murakawa, K.; Chikumoto, A.; Matsuda, K.; Uehara, S.; Ohata, S.; et al. Antineutrophil Cytoplasmic Antibody-Negative Granulomatosis with Polyangiitis Localized to the Lungs. Respir. Med. Case Rep. 2022, 36, 101600. [Google Scholar] [CrossRef]
  36. Eisenberger, U.; Fakhouri, F.; Vanhille, P.; Beaufils, H.; Mahr, A.; Guillevin, L.; Lesavre, P.; Noël, L.-H. ANCA-Negative Pauci-Immune Renal Vasculitis: Histology and Outcome. Nephrol. Dial. Transplant. 2005, 20, 1392–1399. [Google Scholar] [CrossRef]
  37. Falde, S.D.; Fussner, L.A.; Tazelaar, H.D.; O’Brien, E.K.; Lamprecht, P.; Konig, M.F.; Specks, U. Proteinase 3-Specific Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Lancet Rheumatol. 2024, 6, e314–e327. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, Y.; Wan, Q.; Liu, B. ANCA-Negative Eosinophilic Granulomatosis with Polyangiitis Complicated by Peripheral Nerve Damage: A Case Report. Medicine 2023, 102, e34450. [Google Scholar] [CrossRef]
  39. Tanna, A.; Salama, A.D.; Brookes, P.; Pusey, C.D. Familial Granulomatosis with Polyangiitis: Three Cases of This Rare Disorder in One Indoasian Family Carrying an Identical HLA DPB1 Allele. BMJ Case Rep. 2012, 2012, bcr0120125502. [Google Scholar] [CrossRef]
  40. Ueki, Y.; Oshikata, C.; Asai, Y.; Kaneko, T.; Tsurikisawa, N. Familial Eosinophilic Granulomatosis with Polyangiitis in a Sister and Brother. Intern. Med. 2020, 59, 991–995. [Google Scholar] [CrossRef]
  41. Quan, M.V.; Frankel, S.K.; Maleki-Fischbach, M.; Tan, L.D. A Rare Case Report of Polyangiitis Overlap Syndrome: Granulomatosis with Polyangiitis and Eosinophilic Granulomatosis with Polyangiitis. BMC Pulm. Med. 2018, 18, 181. [Google Scholar] [CrossRef]
  42. Kitching, A.R.; Anders, H.-J.; Basu, N.; Brouwer, E.; Gordon, J.; Jayne, D.R.; Kullman, J.; Lyons, P.A.; Merkel, P.A.; Savage, C.O.S.; et al. ANCA-Associated Vasculitis. Nat. Rev. Dis. Primers 2020, 6, 71. [Google Scholar] [CrossRef] [PubMed]
  43. Knight, A.; Sandin, S.; Askling, J. Risks and Relative Risks of Wegener’s Granulomatosis among Close Relatives of Patients with the Disease. Arthritis Rheum. 2008, 58, 302–307. [Google Scholar] [CrossRef] [PubMed]
  44. Hemminki, K.; Li, X.; Sundquist, J.; Sundquist, K. Familial Associations of Rheumatoid Arthritis with Autoimmune Diseases and Related Conditions. Arthritis Rheum. 2009, 60, 661–668. [Google Scholar] [CrossRef]
  45. Li, W.; Huang, H.; Cai, M.; Yuan, T.; Sheng, Y. Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Update: Genetic Pathogenesis. Front. Immunol. 2021, 12, 624848. [Google Scholar] [CrossRef]
  46. Jagiello, P.; Gencik, M.; Arning, L.; Wieczorek, S.; Kunstmann, E.; Csernok, E.; Gross, W.L.; Epplen, J.T. New Genomic Region for Wegener?S Granulomatosis as Revealed by an Extended Association Screen with 202 Apoptosis-Related Genes. Hum. Genet. 2004, 114, 468–477. [Google Scholar] [CrossRef] [PubMed]
  47. Heckmann, M.; Holle, J.U.; Arning, L.; Knaup, S.; Hellmich, B.; Nothnagel, M.; Jagiello, P.; Gross, W.L.; Epplen, J.T.; Wieczorek, S. The Wegener’s Granulomatosis Quantitative Trait Locus on Chromosome 6p21.3 as Characterised by tagSNP Genotyping. Ann. Rheum. Dis. 2008, 67, 972–979. [Google Scholar] [CrossRef]
  48. Luo, H.; Chen, M.; Yang, R.; Xu, P.-C.; Zhao, M.-H. The Association of HLA-DRB1 Alleles with Antineutrophil Cytoplasmic Antibody-Associated Systemic Vasculitis in Chinese Patients. Hum. Immunol. 2011, 72, 422–425. [Google Scholar] [CrossRef]
  49. Tsuchiya, N.; Kobayashi, S.; Hashimoto, H.; Ozaki, S.; Tokunaga, K. Association of HLA-DRB1*0901-DQB1*0303 Haplotype with Microscopic Polyangiitis in Japanese. Genes Immun. 2006, 7, 81–84. [Google Scholar] [CrossRef]
  50. Vaglio, A.; Martorana, D.; Maggiore, U.; Grasselli, C.; Zanetti, A.; Pesci, A.; Garini, G.; Manganelli, P.; Bottero, P.; Tumiati, B.; et al. HLA–DRB4 as a Genetic Risk Factor for Churg-Strauss Syndrome. Arthritis Rheum. 2007, 56, 3159–3166. [Google Scholar] [CrossRef]
  51. Wieczorek, S.; Hellmich, B.; Gross, W.L.; Epplen, J.T. Associations of Churg-Strauss Syndrome with the HLA–DRB1 Locus, and Relationship to the Genetics of Antineutrophil Cytoplasmic Antibody–Associated Vasculitides: Comment on the Article by Vaglio et Al. Arthritis Rheum. 2008, 58, 329–330. [Google Scholar] [CrossRef]
  52. Parry, R.V.; Chemnitz, J.M.; Frauwirth, K.A.; Lanfranco, A.R.; Braunstein, I.; Kobayashi, S.V.; Linsley, P.S.; Thompson, C.B.; Riley, J.L. CTLA-4 and PD-1 Receptors Inhibit T-Cell Activation by Distinct Mechanisms. Mol. Cell. Biol. 2005, 25, 9543–9553. [Google Scholar] [CrossRef] [PubMed]
  53. Collins, A.V.; Brodie, D.W.; Gilbert, R.J.C.; Iaboni, A.; Manso-Sancho, R.; Walse, B.; Stuart, D.I.; Van Der Merwe, P.A.; Davis, S.J. The Interaction Properties of Costimulatory Molecules Revisited. Immunity 2002, 17, 201–210. [Google Scholar] [CrossRef]
  54. Carr, E.J.; Niederer, H.A.; Williams, J.; Harper, L.; Watts, R.A.; Lyons, P.A.; Smith, K.G. Confirmation of the Genetic Association of CTLA4 and PTPN22 with ANCA-Associated Vasculitis. BMC Med. Genet. 2009, 10, 121. [Google Scholar] [CrossRef]
  55. Kamesh, L.; Heward, J.M.; Williams, J.M.; Gough, S.C.L.; Chavele, K.-M.; Salama, A.; Pusey, C.; Savage, C.O.S.; Harper, L. CT60 and +49 Polymorphisms of CTLA 4 Are Associated with ANCA-Positive Small Vessel Vasculitis. Rheumatology 2009, 48, 1502–1505. [Google Scholar] [CrossRef]
  56. Husmann, C.A.; Holle, J.U.; Moosig, F.; Mueller, S.; Wilde, B.; Cohen Tervaert, J.W.; Harper, L.; Assmann, G.; Gross, W.L.; Epplen, J.T.; et al. Genetics of Toll like Receptor 9 in ANCA Associated Vasculitides. Ann. Rheum. Dis. 2014, 73, 890–896. [Google Scholar] [CrossRef] [PubMed]
  57. Jagiello, P.; Aries, P.; Arning, L.; Wagenleiter, S.E.N.; Csernok, E.; Hellmich, B.; Gross, W.L.; Epplen, J.T. The PTPN22 620W Allele Is a Risk Factor for Wegener’s Granulomatosis. Arthritis Rheum. 2005, 52, 4039–4043. [Google Scholar] [CrossRef] [PubMed]
  58. Martorana, D.; Maritati, F.; Malerba, G.; Bonatti, F.; Alberici, F.; Oliva, E.; Sebastio, P.; Manenti, L.; Brugnano, R.; Catanoso, M.G.; et al. PTPN22 R620W Polymorphism in the ANCA-Associated Vasculitides. Rheumatology 2012, 51, 805–812. [Google Scholar] [CrossRef]
  59. Gencik, M.; Meller, S.; Borgmann, S.; Fricke, H. Proteinase 3 Gene Polymorphisms and Wegener’s Granulomatosis. Kidney Int. 2000, 58, 2473–2477. [Google Scholar] [CrossRef]
  60. Merkel, P.A.; Xie, G.; Monach, P.A.; Ji, X.; Ciavatta, D.J.; Byun, J.; Pinder, B.D.; Zhao, A.; Zhang, J.; Tadesse, Y.; et al. Identification of Functional and Expression Polymorphisms Associated with Risk for Antineutrophil Cytoplasmic Autoantibody–Associated Vasculitis. Arthritis Rheumatol. 2017, 69, 1054–1066. [Google Scholar] [CrossRef]
  61. Elzouki, A.-N.Y.; Segelmark, M.; Wieslander, J.; Eriksson, S. Strong Link between the Alpha1 -antitrypsin PiZ Allele and Wegener’s Granulomatosis. J. Intern. Med. 1994, 236, 543–548. [Google Scholar] [CrossRef]
  62. Borgmann, S.; Endisch, G.; Urban, S.; Sitter, T.; Fricke, H. A Linkage Disequilibrium between Genes at the Serine Protease Inhibitor Gene Cluster on Chromosome 14q32.1 Is Associated with Wegener’s Granulomatosis. Clin. Immunol. 2001, 98, 244–248. [Google Scholar] [CrossRef]
  63. Lyons, P.A.; Peters, J.E.; Alberici, F.; Liley, J.; Coulson, R.M.R.; Astle, W.; Baldini, C.; Bonatti, F.; Cid, M.C.; Elding, H.; et al. Genome-Wide Association Study of Eosinophilic Granulomatosis with Polyangiitis Reveals Genomic Loci Stratified by ANCA Status. Nat. Commun. 2019, 10, 5120. [Google Scholar] [CrossRef] [PubMed]
  64. Lyons, P.A.; Rayner, T.F.; Trivedi, S.; Holle, J.U.; Watts, R.A.; Jayne, D.R.W.; Baslund, B.; Brenchley, P.; Bruchfeld, A.; Chaudhry, A.N.; et al. Genetically Distinct Subsets within ANCA-Associated Vasculitis. N. Engl. J. Med. 2012, 367, 214–223. [Google Scholar] [CrossRef] [PubMed]
  65. Xie, G.; Roshandel, D.; Sherva, R.; Monach, P.A.; Lu, E.Y.; Kung, T.; Carrington, K.; Zhang, S.S.; Pulit, S.L.; Ripke, S.; et al. Association of Granulomatosis with Polyangiitis (Wegener’s) With HLA–DPB1*04 and SEMA6A Gene Variants: Evidence From Genome-Wide Analysis. Arthritis Rheum. 2013, 65, 2457–2468. [Google Scholar] [CrossRef] [PubMed]
  66. Moosavi, A.; Motevalizadeh Ardekani, A. Role of Epigenetics in Biology and Human Diseases. Iran. Biomed. J. 2016, 20, 246–258. [Google Scholar] [CrossRef] [PubMed]
  67. Bird, A. Perceptions of Epigenetics. Nature 2007, 447, 396–398. [Google Scholar] [CrossRef]
  68. Wardowska, A. The Epigenetic Face of Lupus: Focus on Antigen-Presenting Cells. Int. Immunopharmacol. 2020, 81, 106262. [Google Scholar] [CrossRef]
  69. Karagianni, P.; Tzioufas, A.G. Epigenetic Perspectives on Systemic Autoimmune Disease. J. Autoimmun. 2019, 104, 102315. [Google Scholar] [CrossRef]
  70. Jones, B.E.; Yang, J.; Muthigi, A.; Hogan, S.L.; Hu, Y.; Starmer, J.; Henderson, C.D.; Poulton, C.J.; Brant, E.J.; Pendergraft, W.F.; et al. Gene-Specific DNA Methylation Changes Predict Remission in Patients with ANCA-Associated Vasculitis. J. Am. Soc. Nephrol. 2017, 28, 1175–1187. [Google Scholar] [CrossRef]
  71. Ciavatta, D.J.; Yang, J.; Preston, G.A.; Badhwar, A.K.; Xiao, H.; Hewins, P.; Nester, C.M.; Pendergraft, W.F.; Magnuson, T.R.; Jennette, J.C.; et al. Epigenetic Basis for Aberrant Upregulation of Autoantigen Genes in Humans with ANCA Vasculitis. J. Clin. Investig. 2010, 120, 3209–3219. [Google Scholar] [CrossRef]
  72. Yang, J.; Ge, H.; Poulton, C.J.; Hogan, S.L.; Hu, Y.; Jones, B.E.; Henderson, C.D.; McInnis, E.A.; Pendergraft, W.F.; Jennette, J.C.; et al. Histone Modification Signature at Myeloperoxidase and Proteinase 3 in Patients with Anti-Neutrophil Cytoplasmic Autoantibody-Associated Vasculitis. Clin. Epigenetics 2016, 8, 85. [Google Scholar] [CrossRef] [PubMed]
  73. Wong, C.-K.; Dong, J.; Lam, C.W.-K. Molecular Mechanisms Regulating the Synergism between IL-32γ and NOD for the Activation of Eosinophils. J. Leukoc. Biol. 2013, 95, 631–642. [Google Scholar] [CrossRef] [PubMed]
  74. Olsen, R.L.; Little, C. Purification and Some Properties of Myeloperoxidase and Eosinophil Peroxidase from Human Blood. Biochem. J. 1983, 209, 781–787. [Google Scholar] [CrossRef]
  75. Shao, B.; Oda, M.N.; Oram, J.F.; Heinecke, J.W. Myeloperoxidase: An Oxidative Pathway for Generating Dysfunctional High-Density Lipoprotein. Chem. Res. Toxicol. 2010, 23, 447–454. [Google Scholar] [CrossRef]
  76. Sugawara, S. Immune Functions of Proteinase 3. Crit. Rev. Immunol. 2005, 25, 343–360. [Google Scholar] [CrossRef]
  77. Schreiber, A.; Luft, F.C.; Kettritz, R. Membrane Proteinase 3 Expression and ANCA-Induced Neutrophil Activation. Kidney Int. 2004, 65, 2172–2183. [Google Scholar] [CrossRef]
  78. van der Geld, Y.M.; Limburg, P.C.; Kallenberg, C.G. Proteinase 3, Wegener’s Autoantigen: From Gene to Antigen. J. Leukoc. Biol. 2001, 69, 177–190. [Google Scholar] [CrossRef] [PubMed]
  79. Silvescu, C.I.; Sackstein, R. G-CSF Induces Membrane Expression of a Myeloperoxidase Glycovariant That Operates as an E-Selectin Ligand on Human Myeloid Cells. Proc. Natl. Acad. Sci. USA 2014, 111, 10696–10701. [Google Scholar] [CrossRef]
  80. Tan, D.S.; Gan, P.Y.; O’Sullivan, K.M.; Hammett, M.V.; Summers, S.A.; Ooi, J.D.; Lundgren, B.A.; Boyd, R.L.; Scott, H.S.; Kitching, A.R.; et al. Thymic deletion and regulatory T cells prevent antimyeloperoxidase GN. J. Am. Soc. Nephrol. 2013, 24, 573–585. [Google Scholar] [CrossRef]
  81. Gan, P.Y.; Holdsworth, S.R.; Kitching, A.R.; Ooi, J.D. Myeloperoxidase (MPO)-specific CD4+ T cells contribute to MPO-anti-neutrophil cytoplasmic antibody (ANCA) associated glomerulonephritis. Cell. Immunol. 2013, 282, 21–27. [Google Scholar] [CrossRef]
  82. Primo, V.C.; Marusic, S.; Franklin, C.C.; Goldmann, W.H.; Achaval, C.G.; Smith, R.N.; Arnaout, M.A.; Nikolic, B. Anti-PR3 immune responses induce segmental and necrotizing glomerulonephritis. Clin. Exp. Immunol. 2010, 159, 327–337. [Google Scholar] [CrossRef]
  83. Sharma, R.K.; Yoosuf, N.; Afonso, M.; Scheffschick, A.; Avik, A.; Bartoletti, A.; Horuluoglu, B.; Diaz Boada, J.S.; Boddul, S.K.; Jonasdottir, A.D.; et al. Identification of proteinase 3 autoreactive CD4+T cells and their T-cell receptor repertoires in antineutrophil cytoplasmic antibody-associated vasculitis. Kidney Int. 2023, 103, 973–985. [Google Scholar] [CrossRef] [PubMed]
  84. Porges, A.J.; Redecha, P.B.; Kimberly, W.T.; Csernok, E.; Gross, W.L.; Kimberly, R.P. Anti-Neutrophil Cytoplasmic Antibodies Engage and Activate Human Neutrophils via Fc Gamma RIIa. J. Immunol. 1994, 153, 1271–1280. [Google Scholar] [CrossRef] [PubMed]
  85. Williams, J.M.; Ben-Smith, A.; Hewins, P.; Dove, S.K.; Hughes, P.; McEwan, R.; Wakelam, M.J.O.; Savage, C.O.S. Activation of the Gi Heterotrimeric G Protein by ANCA IgG F(Ab′)2 Fragments Is Necessary but Not Sufficient to Stimulate the Recruitment of Those Downstream Mediators Used by Intact ANCA IgG. J. Am. Soc. Nephrol. 2003, 14, 661–669. [Google Scholar] [CrossRef]
  86. Maverakis, E.; Kim, K.; Shimoda, M.; Gershwin, M.E.; Patel, F.; Wilken, R.; Raychaudhuri, S.; Ruhaak, L.R.; Lebrilla, C.B. Glycans in the Immune System and The Altered Glycan Theory of Autoimmunity: A Critical Review. J. Autoimmun. 2015, 57, 1–13. [Google Scholar] [CrossRef] [PubMed]
  87. Falk, R.J.; Terrell, R.S.; Charles, L.A.; Jennette, J.C. Anti-Neutrophil Cytoplasmic Autoantibodies Induce Neutrophils to Degranulate and Produce Oxygen Radicals in Vitro. Proc. Natl. Acad. Sci. USA 1990, 87, 4115–4119. [Google Scholar] [CrossRef]
  88. Thomas, C.J.; Schroder, K. Pattern recognition receptor function in neutrophils. Trends Immunol. 2013, 34, 317–328. [Google Scholar] [CrossRef]
  89. Rodrigues, D.A.S.; Prestes, E.B.; Gama, A.M.S.; Silva, L.S.; Pinheiro, A.A.S.; Ribeiro, J.M.C.; Campos, R.M.P.; Pimentel-Coelho, P.M.; De Souza, H.S.; Dicko, A.; et al. CXCR4 and MIF are required for neutrophil extracellular trap release triggered by Plasmodium-infected erythrocytes. PLoS Pathog. 2020, 16, e1008230. [Google Scholar] [CrossRef]
  90. Kumar, S.V.; Kulkarni, O.P.; Mulay, S.R.; Darisipudi, M.N.; Romoli, S.; Thomasova, D.; Scherbaum, C.R.; Hohenstein, B.; Hugo, C.; Müller, S.; et al. Neutrophil Extracellular Trap-Related Extracellular Histones Cause Vascular Necrosis in Severe GN. J. Am. Soc. Nephrol. 2015, 26, 2399–2413. [Google Scholar] [CrossRef]
  91. Schreiber, A.; Rousselle, A.; Becker, J.U.; von Mässenhausen, A.; Linkermann, A.; Kettritz, R. Necroptosis controls NET generation and mediates complement activation, endothelial damage, and autoimmune vasculitis. Proc. Natl. Acad. Sci. USA 2017, 114, E9618–E9625. [Google Scholar] [CrossRef]
  92. Sangaletti, S.; Tripodo, C.; Chiodoni, C.; Guarnotta, C.; Cappetti, B.; Casalini, P.; Piconese, S.; Parenza, M.; Guiducci, C.; Vitali, C.; et al. Neutrophil Extracellular Traps Mediate Transfer of Cytoplasmic Neutrophil Antigens to Myeloid Dendritic Cells toward ANCA Induction and Associated Autoimmunity. Blood 2012, 120, 3007–3018. [Google Scholar] [CrossRef] [PubMed]
  93. Kessenbrock, K.; Krumbholz, M.; Schönermarck, U.; Back, W.; Gross, W.L.; Werb, Z.; Gröne, H.-J.; Brinkmann, V.; Jenne, D.E. Netting Neutrophils in Autoimmune Small-Vessel Vasculitis. Nat. Med. 2009, 15, 623–625. [Google Scholar] [CrossRef] [PubMed]
  94. Matsuda, Y.; Hamayasu, H.; Seki, A.; Nonaka, K.; Wang, T.; Matsumoto, T.; Hamano, Y.; Sumikura, H.; Kumasaka, T.; Murayama, S.; et al. Presence of Citrullinated Histone H3-Positive Neutrophils in Microscopic Polyangiitis from the Early Phase: An Autopsy Proven Case. Pathol. Int. 2016, 66, 466–471. [Google Scholar] [CrossRef] [PubMed]
  95. Takeuchi, H.; Kawasaki, T.; Shigematsu, K.; Kawamura, K.; Oka, N. Neutrophil extracellular traps in neuropathy with anti-neutrophil cytoplasmic autoantibody-associated microscopic polyangiitis. Clin. Rheumatol. 2017, 36, 913–917. [Google Scholar] [CrossRef]
  96. Meng, W.; Paunel-Görgülü, A.; Flohé, S.; Witte, I.; Schädel-Höpfner, M.; Windolf, J.; Lögters, T.T. Deoxyribonuclease is a potential counter regulator of aberrant neutrophil extracellular traps formation after major trauma. Mediat. Inflamm. 2012, 2012, 149560. [Google Scholar] [CrossRef]
  97. Söderberg, D.; Kurz, T.; Motamedi, A.; Hellmark, T.; Eriksson, P.; Segelmark, M. Increased levels of neutrophil extracellular trap remnants in the circulation of patients with small vessel vasculitis, but an inverse correlation to anti-neutrophil cytoplasmic antibodies during remission. Rheumatology 2015, 54, 2085–2094. [Google Scholar] [CrossRef]
  98. Nakazawa, D.; Shida, H.; Tomaru, U.; Yoshida, M.; Nishio, S.; Atsumi, T.; Ishizu, A. Enhanced formation and disordered regulation of NETs in myeloperoxidase-ANCA-associated microscopic polyangiitis. J. Am. Soc. Nephrol. 2014, 25, 990–997. [Google Scholar] [CrossRef] [PubMed]
  99. Argyropoulou, O.D.; Goules, A.V.; Boutzios, G.; Tsirogianni, A.; Sfontouris, C.; Manoussakis, M.N.; Vlachoyiannopoulos, P.G.; Tzioufas, A.G.; Kapsogeorgou, E.K. Occurrence and Antigenic Specificity of Perinuclear Anti-Neutrophil Cytoplasmic Antibodies (P-ANCA) in Systemic Autoimmune Diseases. Cells 2021, 10, 2128. [Google Scholar] [CrossRef]
  100. Novikov, P.; Smitienko, I.; Bulanov, N.; Zykova, A.; Moiseev, S. Testing for antineutrophil cytoplasmic antibodies (ANCAs) in patients with systemic vasculitides and other diseases. Ann. Rheum. Dis. 2017, 76, e23. [Google Scholar] [CrossRef]
  101. Kagiyama, N.; Takayanagi, N.; Kanauchi, T.; Ishiguro, T.; Yanagisawa, T.; Sugita, Y. Antineutrophil cytoplasmic antibody-positive conversion and microscopic polyangiitis development in patients with idiopathic pulmonary fibrosis. BMJ Open Respir. Res. 2015, 2, e000058. [Google Scholar] [CrossRef]
  102. Johansson, Å.C.; Ohlsson, S.; Pettersson, Å.; Bengtsson, A.A.; Selga, D.; Hansson, M.; Hellmark, T. Impaired Phagocytosis and Reactive Oxygen Species Production in Phagocytes Is Associated with Systemic Vasculitis. Arthritis Res. Ther. 2016, 18, 92. [Google Scholar] [CrossRef]
  103. Mishra, R.; Behera, L.M.; Rana, S. Binding of raloxifene to human complement fragment 5a (hC5a): A perspective on cytokine storm and COVID19. J. Biomol. Struct. Dyn. 2022, 40, 982–994. [Google Scholar] [CrossRef] [PubMed]
  104. Schreiber, A.; Xiao, H.; Jennette, J.C.; Schneider, W.; Luft, F.C.; Kettritz, R. C5a receptor mediates neutrophil activation and ANCA-induced glomerulonephritis. J. Am. Soc. Nephrol. 2009, 20, 289–298. [Google Scholar] [CrossRef] [PubMed]
  105. Rzeniewicz, K.; Newe, A.; Rey Gallardo, A.; Davies, J.; Holt, M.R.; Patel, A.; Charras, G.T.; Stramer, B.; Molenaar, C.; Tedder, T.F.; et al. L-selectin shedding is activated specifically within transmigrating pseudopods of monocytes to regulate cell polarity in vitro. Proc. Natl. Acad. Sci. USA 2015, 112, E1461–E1470. [Google Scholar] [CrossRef]
  106. Wlodarczyk, A.; Løbner, M.; Cédile, O.; Owens, T. Comparison of microglia and infiltrating CD11c⁺ cells as antigen presenting cells for T cell proliferation and cytokine response. J. Neuroinflammation 2014, 11, 57. [Google Scholar] [CrossRef]
  107. Hattar, K.; Bickenbach, A.; Csernok, E.; Rosseau, S.; Grandel, U.; Seeger, W.; Grimminger, F.; Sibelius, U. Wegener’s Granulomatosis: Antiproteinase 3 Antibodies Induce Monocyte Cytokine and Prostanoid Release—Role of Autocrine Cell Activation. J. Leukoc. Biol. 2002, 71, 996–1004. [Google Scholar] [CrossRef] [PubMed]
  108. Weidner, S.; Neupert, W.; Goppelt-Struebe, M.; Rupprecht, H.D. Antineutrophil Cytoplasmic Antibodies Induce Human Monocytes to Produce Oxygen Radicals in Vitro. Arthritis Rheum. 2001, 44, 1698–1706. [Google Scholar] [CrossRef]
  109. Tarzi, R.M.; Liu, J.; Schneiter, S.; Hill, N.R.; Page, T.H.; Cook, H.T.; Pusey, C.D.; Woollard, K.J. CD14 Expression Is Increased on Monocytes in Patients with Anti-Neutrophil Cytoplasm Antibody (ANCA)-Associated Vasculitis and Correlates with the Expression of ANCA Autoantigens. Clin. Exp. Immunol. 2015, 181, 65–75. [Google Scholar] [CrossRef]
  110. Matsumoto, T.; Matsui, T.; Hirano, F.; Tohma, S.; Mori, M. Disease Activity, Treatment and Long-Term Prognosis of Adult Juvenile Idiopathic Arthritis Patients Compared with Rheumatoid Arthritis Patients. Mod. Rheumatol. 2020, 30, 78–84. [Google Scholar] [CrossRef]
  111. Kobold, A.C.M.; Kallenberg, C.G.M.; Tervaert, J.W.C. Monocyte Activation in Patients with Wegener’s Granulomatosis. Ann. Rheum. Dis. 1999, 58, 237–245. [Google Scholar] [CrossRef]
  112. Haller, H.; Eichhorn, J.; Pieper, K.; Göbel, U.; Luft, F.C. Circulating Leukocyte Integrin Expression in Wegener’s Granulomatosis. J. Am. Soc. Nephrol. 1996, 7, 40–48. [Google Scholar] [CrossRef]
  113. Emmi, G.; Bettiol, A.; Gelain, E.; Bajema, I.M.; Berti, A.; Burns, S.; Cid, M.C.; Cohen Tervaert, J.W.; Cottin, V.; Durante, E.; et al. Evidence-Based Guideline for the Diagnosis and Management of Eosinophilic Granulomatosis with Polyangiitis. Nat. Rev. Rheumatol. 2023, 19, 378–393. [Google Scholar] [CrossRef] [PubMed]
  114. Watanabe, R.; Hashimoto, M. Eosinophilic Granulomatosis with Polyangiitis: Latest Findings and Updated Treatment Recommendations. J. Clin. Med. 2023, 12, 5996. [Google Scholar] [CrossRef]
  115. Kotas, M.E.; Dion, J.; Van Dyken, S.; Ricardo-Gonzalez, R.R.; Danel, C.J.; Taillé, C.; Mouthon, L.; Locksley, R.M.; Terrier, B. A Role for IL-33–Activated ILC2s in Eosinophilic Vasculitis. JCI Insight 2021, 6, e143366. [Google Scholar] [CrossRef] [PubMed]
  116. Fulkerson, P.C.; Schollaert, K.L.; Bouffi, C.; Rothenberg, M.E. IL-5 Triggers a Cooperative Cytokine Network That Promotes Eosinophil Precursor Maturation. J. Immunol. 2014, 193, 4043–4052. [Google Scholar] [CrossRef] [PubMed]
  117. Wechsler, M.E.; Akuthota, P.; Jayne, D.; Khoury, P.; Klion, A.; Langford, C.A.; Merkel, P.A.; Moosig, F.; Specks, U.; Cid, M.C.; et al. Mepolizumab or Placebo for Eosinophilic Granulomatosis with Polyangiitis. N. Engl. J. Med. 2017, 376, 1921–1932. [Google Scholar] [CrossRef]
  118. Fukuchi, M.; Kamide, Y.; Ueki, S.; Miyabe, Y.; Konno, Y.; Oka, N.; Takeuchi, H.; Koyota, S.; Hirokawa, M.; Yamada, T.; et al. Eosinophil ETosis–Mediated Release of Galectin-10 in Eosinophilic Granulomatosis with Polyangiitis. Arthritis Rheumatol. 2021, 73, 1683–1693. [Google Scholar] [CrossRef]
  119. Rodríguez-Alcázar, J.F.; Ataide, M.A.; Engels, G.; Schmitt-Mabmunyo, C.; Garbi, N.; Kastenmüller, W.; Latz, E.; Franklin, B.S. Charcot–Leyden Crystals Activate the NLRP3 Inflammasome and Cause IL-1β Inflammation in Human Macrophages. J. Immunol. 2019, 202, 550–558. [Google Scholar] [CrossRef]
  120. Hilhorst, M.; Shirai, T.; Berry, G.; Goronzy, J.J.; Weyand, C.M. T Cell-acrophage Interactions and Granuloma Formation in Vasculitis. Front. Immunol. 2014, 5, 432. [Google Scholar] [CrossRef]
  121. Su, B.; Mao, X.; Yin, B.; Chen, C.; Zhang, M.; Cui, T.; Hao, Y. TIM-3 Regulates the NETs-Mediated Dendritic Cell Activation in Myeloperoxidase-ANCA-Associated Vasculitis. Clin. Exp. Rheumatol. 2021, 39, 13–20. [Google Scholar] [CrossRef]
  122. Uno, K.; Muso, E.; Ito-Ihara, T.; Endo, T.; Yasuda, Y.; Yagi, K.; Suzuki, K. Impaired HVJ-Stimulated Interferon Producing Capacity in MPO-ANCA-Associated Vasculitis with Rapidly Progressive Glomerulonephritis Lead to Susceptibility to Infection. Cytokine 2020, 136, 155221. [Google Scholar] [CrossRef] [PubMed]
  123. Odobasic, D.; Holdsworth, S.R. Emerging Cellular Therapies for Anti-Myeloperoxidase Vasculitis and Other Autoimmune Diseases. Front. Immunol. 2021, 12, 642127. [Google Scholar] [CrossRef]
  124. Braudeau, C.; Néel, A.; Amouriaux, K.; Martin, J.C.; Rimbert, M.; Besançon, A.; Giraudet, S.; Terrien, C.; Aliaga, M.; Salabert-Le Guen, N.; et al. Dysregulated Responsiveness of Circulating Dendritic Cells to Toll-Like Receptors in ANCA-Associated Vasculitis. Front. Immunol. 2017, 8, 102. [Google Scholar] [CrossRef]
  125. Wilde, B.; Van Paassen, P.; Damoiseaux, J.; Heerings-Rewinkel, P.; Van Rie, H.; Witzke, O.; Tervaert, J.W.C. Dendritic Cells in Renal Biopsies of Patients with ANCA-Associated Vasculitis. Nephrol. Dial. Transplant. 2009, 24, 2151–2156. [Google Scholar] [CrossRef] [PubMed]
  126. Tsurikisawa, N.; Saito, H.; Oshikata, C.; Tsuburai, T.; Ishiyama, M.; Mitomi, H.; Akiyama, K. An Increase of CD83+ Dendritic Cells Ex Vivo Correlates with Increased Regulatory T Cells in Patients with Active Eosinophilic Granulomatosis and Polyangiitis. BMC Immunol. 2014, 15, 32. [Google Scholar] [CrossRef]
  127. Fijolek, J.; Radzikowska, E. Eosinophilic granulomatosis with polyangiitis—Advances in pathogenesis, diagnosis, and treatment. Front. Med. 2023, 10, 1145257. [Google Scholar] [CrossRef] [PubMed]
  128. Hoshino, A.; Nagao, T.; Nagi-Miura, N.; Ohno, N.; Yasuhara, M.; Yamamoto, K.; Nakayama, T.; Suzuki, K. MPO-ANCA induces IL-17 production by activated neutrophils in vitro via classical complement pathway-dependent manner. J. Autoimmun. 2008, 31, 79–89. [Google Scholar] [CrossRef]
  129. Mulder, A.H.; Stegeman, C.A.; Kallenberg, C.G. Activation of granulocytes by anti-neutrophil cytoplasmic antibodies (ANCA) in Wegener’s granulomatosis: A predominant role for the IgG3 subclass of ANCA. Clin. Exp. Immunol. 1995, 101, 227–232. [Google Scholar] [CrossRef]
  130. Nozaki, Y. New Insights into Novel Therapeutic Targets in ANCA-Associated Vasculitis. Front. Immunol. 2021, 12, 631055. [Google Scholar] [CrossRef]
  131. Martinez Valenzuela, L.; Bordignon Draibe, J.; Fulladosa Oliveras, X.; Bestard Matamoros, O.; Cruzado Garrit, J.M.; Torras Ambrós, J. T-Lymphocyte in ANCA-Associated Vasculitis: What Do We Know? A Pathophysiological and Therapeutic Approach. Clin. Kidney J. 2019, 12, 503–511. [Google Scholar] [CrossRef]
  132. Balding, C.E.J.; Howie, A.J.; Drake-Lee, A.B.; Savage, C.O.S. Th2 Dominance in Nasal Mucosa in Patients with Wegener’s Granulomatosis. Clin. Exp. Immunol. 2001, 125, 332–339. [Google Scholar] [CrossRef] [PubMed]
  133. Abdulahad, W.H.; Van Der Geld, Y.M.; Stegeman, C.A.; Kallenberg, C.G.M. Persistent Expansion of CD4+ Effector Memory T Cells in Wegener’s Granulomatosis. Kidney Int. 2006, 70, 938–947. [Google Scholar] [CrossRef] [PubMed]
  134. Szczeklik, W.; Jakieła, B.; Wawrzycka-Adamczyk, K.; Sanak, M.; Hubalewska-Mazgaj, M.; Padjas, A.; Surmiak, M.; Szczeklik, K.; Sznajd, J.; Musiał, J. Skewing toward Treg and Th2 Responses Is a Characteristic Feature of Sustained Remission in ANCA-positive Granulomatosis with Polyangiitis. Eur. J. Immunol. 2017, 47, 724–733. [Google Scholar] [CrossRef] [PubMed]
  135. Wilde, B.; Thewissen, M.; Damoiseaux, J.; Van Paassen, P.; Witzke, O.; Tervaert, J.W. T Cells in ANCA-Associated Vasculitis: What Can We Learn from Lesional versus Circulating T Cells? Arthritis Res. Ther. 2010, 12, 204. [Google Scholar] [CrossRef]
  136. Marinaki, S.; Kälsch, A.-I.; Grimminger, P.; Breedijk, A.; Birck, R.; Schmitt, W.H.; Weiss, C.; Van Der Woude, F.J.; Yard, B.A. Persistent T-Cell Activation and Clinical Correlations in Patients with ANCA-Associated Systemic Vasculitis. Nephrol. Dial. Transplant. 2006, 21, 1825–1832. [Google Scholar] [CrossRef]
  137. Marinaki, S.; Neumann, I.; Kälsch, A.-I.; Grimminger, P.; Breedijk, A.; Birck, R.; Schmitt, W.; Waldherr, R.; Yard, B.A.; Van Der Woude, F.J. Abnormalities of CD4+ T Cell Subpopulations in ANCA-Associated Vasculitis. Clin. Exp. Immunol. 2005, 140, 181–191. [Google Scholar] [CrossRef]
  138. Moosig, F.; Csernok, E.; Wang, G.; Gross, W.L. Costimulatory Molecules in Wegener’s Granulomatosis (WG): Lack of Expression of CD28 and Preferential up-Regulation of Its Ligands B7-1 (CD80) and B7-2 (CD86) on T Cells. Clin. Exp. Immunol. 2001, 114, 113–118. [Google Scholar] [CrossRef]
  139. Komocsi, A.; Lamprecht, P.; Csernok, E.; Mueller, A.; Holl-Ulrich, K.; Seitzer, U.; Moosig, F.; Schnabel, A.; Gross, W.L. Peripheral Blood and Granuloma CD4+CD28− T Cells Are a Major Source of Interferon-γ and Tumor Necrosis Factor-α in Wegener’s Granulomatosis. Am. J. Pathol. 2002, 160, 1717–1724. [Google Scholar] [CrossRef]
  140. Capraru, D.; Müller, A.; Csernok, E.; Gross, W.L.; Holl-Ulrich, K.; Northfield, J.; Klenerman, P.; Herlyn, K.; Holle, J.; Gottschlich, S.; et al. Expansion of Circulating NKG2D+ Effector Memory T-Cells and Expression of NKG2D-Ligand MIC in Granulomaous Lesions in Wegener’s Granulomatosis. Clin. Immunol. 2008, 127, 144–150. [Google Scholar] [CrossRef]
  141. Żabińska, M.; Kościelska-Kasprzak, K.; Krajewska, J.; Bartoszek, D.; Augustyniak-Bartosik, H.; Krajewska, M. Immune Cells Profiling in ANCA-Associated Vasculitis Patients—Relation to Disease Activity. Cells 2021, 10, 1773. [Google Scholar] [CrossRef]
  142. Evans, H.G.; Gullick, N.J.; Kelly, S.; Pitzalis, C.; Lord, G.M.; Kirkham, B.W.; Taams, L.S. In Vivo Activated Monocytes from the Site of Inflammation in Humans Specifically Promote Th17 Responses. Proc. Natl. Acad. Sci. USA 2009, 106, 6232–6237. [Google Scholar] [CrossRef] [PubMed]
  143. Saito, H.; Tsurikisawa, N.; Tsuburai, T.; Oshikata, C.; Akiyama, K. Cytokine Production Profile of CD4+ T Cells from Patients with Active Churg-Strauss Syndrome Tends Toward Th17. Int. Arch. Allergy Immunol. 2009, 149, 61–65. [Google Scholar] [CrossRef]
  144. Nogueira, E.; Hamour, S.; Sawant, D.; Henderson, S.; Mansfield, N.; Chavele, K.-M.; Pusey, C.D.; Salama, A.D. Serum IL-17 and IL-23 Levels and Autoantigen-Specific Th17 Cells Are Elevated in Patients with ANCA-Associated Vasculitis. Nephrol. Dial. Transplant. 2010, 25, 2209–2217. [Google Scholar] [CrossRef] [PubMed]
  145. Fuchs, S.; Scheffschick, A.; Gunnarsson, I.; Brauner, H. Natural Killer Cells in Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis—A Review of the Literature. Front. Immunol. 2022, 12, 796640. [Google Scholar] [CrossRef]
  146. Tognarelli, S.; Gayet, J.; Lambert, M.; Dupuy, S.; Karras, A.; Cohen, P.; Guillevin, L.; De Menthon, M.; Caillat-Zucman, S. Tissue-Specific Microvascular Endothelial Cells Show Distinct Capacity to Activate NK Cells: Implications for the Pathophysiology of Granulomatosis with Polyangiitis. J. Immunol. 2014, 192, 3399–3408. [Google Scholar] [CrossRef] [PubMed]
  147. Oleinika, K.; Mauri, C.; Salama, A.D. Effector and Regulatory B Cells in Immune-Mediated Kidney Disease. Nat. Rev. Nephrol. 2019, 15, 11–26. [Google Scholar] [CrossRef]
  148. Rincón-Arévalo, H.; Sanchez-Parra, C.C.; Castaño, D.; Yassin, L.; Vásquez, G. Regulatory B Cells and Mechanisms. Int. Rev. Immunol. 2015, 35, 156–176. [Google Scholar] [CrossRef]
  149. Lepse, N.; Abdulahad, W.H.; Kallenberg, C.G.M.; Heeringa, P. Immune Regulatory Mechanisms in ANCA-Associated Vasculitides. Autoimmun. Rev. 2011, 11, 77–83. [Google Scholar] [CrossRef]
  150. Jones, R.B.; Cohen Tervaert, J.W.; Hauser, T.; Luqmani, R.; Morgan, M.D.; Peh, C.A.; Savage, C.O.; Segelmark, M.; Tesar, V.; Van Paassen, P.; et al. Rituximab versus Cyclophosphamide in ANCA-Associated Renal Vasculitis. N. Engl. J. Med. 2010, 363, 211–220. [Google Scholar] [CrossRef]
  151. Stone, J.H.; Merkel, P.A.; Spiera, R.; Seo, P.; Langford, C.A.; Hoffman, G.S.; Kallenberg, C.G.M.; St. Clair, E.W.; Turkiewicz, A.; Tchao, N.K.; et al. Rituximab versus Cyclophosphamide for ANCA-Associated Vasculitis. N. Engl. J. Med. 2010, 363, 221–232. [Google Scholar] [CrossRef]
  152. Alberici, F.; Smith, R.M.; Jones, R.B.; Roberts, D.M.; Willcocks, L.C.; Chaudhry, A.; Smith, K.G.C.; Jayne, D.R.W. Long-Term Follow-up of Patients Who Received Repeat-Dose Rituximab as Maintenance Therapy for ANCA-Associated Vasculitis. Rheumatology 2015, 54, 1153–1160. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The course of AAV development.
Figure 1. The course of AAV development.
Ijms 26 06065 g001
Table 1. ANCA types.
Table 1. ANCA types.
p-ANCAc-ANCA
AutoantigensMPO, cathepsin G, elastase, lactoferrin, and lysozymeproteinase 3
Staining patternperinuclearcytoplasmic
AAVMPA, EGPAGPA
Table 2. Comparison of GPA, MPA, and EGPA.
Table 2. Comparison of GPA, MPA, and EGPA.
GPAMPAEGPA
Clinical presentationRespiratory tract involvement, and ocular symptoms (uveitis, ocular ulcers, episcleritis, and scleritis)Glomerulonephritis, pulmonary capillaritis, skin, nerves, and gastrointestinal tract involvementRespiratory tract involvement (asthma as a typical early feature), and neurological symptoms
GranulomasPresentNot presentPresent
The main type of ANCAPR3-ANCAMPO-ANCAMPO-ANCA
Percentage of ANCA(−) patients10%10%50%
Main effector cellsNeutrophilsNeutrophilsNeutrophils in ANCA(+), and eosinophils in ANCA(−)
T-cell polarizationTh1 (localized disease), and Th2 (generalized disease)Th1Th1, Th17 in ANCA(+), and Th2 in ANCA(−)
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

Lisowska, K.A.; Wardowska, A. Immunopathology of Antineutrophil Cytoplasmic Antibody (ANCA)-Associated Vasculitis. Int. J. Mol. Sci. 2025, 26, 6065. https://doi.org/10.3390/ijms26136065

AMA Style

Lisowska KA, Wardowska A. Immunopathology of Antineutrophil Cytoplasmic Antibody (ANCA)-Associated Vasculitis. International Journal of Molecular Sciences. 2025; 26(13):6065. https://doi.org/10.3390/ijms26136065

Chicago/Turabian Style

Lisowska, Katarzyna Aleksandra, and Anna Wardowska. 2025. "Immunopathology of Antineutrophil Cytoplasmic Antibody (ANCA)-Associated Vasculitis" International Journal of Molecular Sciences 26, no. 13: 6065. https://doi.org/10.3390/ijms26136065

APA Style

Lisowska, K. A., & Wardowska, A. (2025). Immunopathology of Antineutrophil Cytoplasmic Antibody (ANCA)-Associated Vasculitis. International Journal of Molecular Sciences, 26(13), 6065. https://doi.org/10.3390/ijms26136065

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