Skip to main content

An immunogenetic perspective of ANCA-associated vasculitides

Abstract

Background

Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) are a group of small vessel vasculitides characterized by necrotizan vasculitis and inflammation. The phenotypes of AAV include microscopic polyangiitis (MPA), granulomatosis and polyangiitis (GPA), and eosinophilic granulomatosis and polyangiitis (EGPA). The pathogenesis of AAV is multifactorial, and it is suggested that both genetic and environmental factors can influence these disorders.

Main body

Several candidate gene studies and genome-wide association studies (GWAS) have been conducted to investigate the genetic associations with AAV in recent years. Numerous genes have been related to the pathogenesis of AAV, including the innate, adaptive immune system and coagulation systems.

Conclusion

This review summarizes the immunological mechanisms involved in the etiopathogenesis of AAV and recent advances in susceptibility genes.

Background

Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) are primary systemic disorders that cause necrotizing inflammation of the small blood vessels such as arterioles, capillaries, and venules. The presence of anti-neutrophil cytoplasmic antibodies (ANCA) is a characteristic feature of ANCA-associated vasculitides [1]. This group includes granulomatosis with polyangiitis (GPA, formerly Wegener granulomatosis), microscopic polyangiitis (MPA), and eosinophilic GPA (EGPA, formerly Churg-Strauss syndrome) [2]. Although the prevalence of AAV varies by geographic region, GPA is more common in Europe, and MPA is more common in Asia [3]. MPA is more common in Southern European countries than in Northern European countries [4]. The pathogenesis of AAV is still unknown. Various genetic, immunological, and micro- and macroenvironmental factors have been suggested to play a role in the development of AAV [5]. Infective triggers that facilitate neutrophil activation and increase migration of cytoplasmic PR3 and MPO antigens to the neutrophil surface may lead to disease onset [6, 7]. In this review, we will focus on the potential effects of polymorphisms in many genes involved in the immune response, which may play a role in the immunogenetic basis of AAV.

Main text

Human leukocyte antigen (HLA) genes

The MHC, also known as the human leukocyte antigen (HLA) region, encodes several molecules that play key roles in the immune system. HLA genes, including both class I (A, B, C) and class II (DR, DQ, and DP), have been associated with susceptibility to AAV [8]. Katz et al. showed an association between HLA-B*08 and GPA [9]. One study demonstrated that the HLA-A*01-B*08-DRB1*03 haplotype was more frequent in AAV patients compared to those with GPA [10]. The relationship between HLA class II genes and AAV shows divergent results across ethnic groups and geographic regions. Previous studies demonstrated that HLA-DPB1 was a major contributor to AAV genetic risk (particularly HLA-DPB1*0401 in GPA) [11, 12]. The alleles of HLA-DRB1*13 were found to be a protective factor for GPA in a Dutch cohort [13]. On the other hand, the alleles of HLA-DRB1*04 have been found to be associated with GPA in Dutch patients [10]. The HLA-DBP1*03:01 was found to be less common in GPA patients with + ANCA [14]. The G allele of HLA-DBP1 (rs3117242) has been shown to be a risk factor for GPA in Caucasian patients [11]. The HLA-DRB1*15 was found to be linked with PR3-ANCA+ disease in a population of African-Americans [15]. In Japanese subjects, the HLA-DRB1*09:01 was found to be associated with both MPA and MPO-ANCA [16]. The rs5000634 single nucleotide polymorphism (SNP) of HLA-DQB1 has also been associated with MPA in Caucasian patients [11]. In another study, HLA-DRB1*08 alleles were found more frequently in EGPA patients compared to the control group [10]. Also, the HLA-DRB4 alleles were found to be related to symptoms of vasculitis in German patients [17]. In a recent study, the DRB1*03 and DQB1*02 alleles were associated with confirmed in proteinase 3 (PR3)-AAV patients, whereas the DRB1*10, DRB1*14, and DQB1*05 were found to be protective alleles in AAV [18].

ANCA target proteins

Proteinase 3 (PR3) rs62132295 was found to be associated with PR3-ANCA+ [19]. In a German population, the − 564 A/G SNP of the PR3 gene was associated with GPA [20]. The GG genotype of myeloperoxidase (MPO) 463 G/A was found to be associated with MPO-ANCA+ in female patients [21]. A recent meta-analysis showed no association between SNPs of the MPO gene and AAV [22]. Serpin family A member 1, encoding α1-antitrypsin (SERPINA1) protease inhibitor Z (PiZ), was found to be linked with AAV [23]. The S and Z alleles of SERPINA1 were related to GPA patients in a North American cohort [24]. However, this significant association was not found in a study with Chinese patients [25].

Co-stimulatory molecules and signaling regulators

Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) gene plays a major role in T cell inhibition. The SNPs of CTLA-4 CT60 and CTLA-4 + 49 have been related to AAV susceptibility in European patients [26]. Another study demonstrated that the CTLA-4-318 SNP was associated with GPA susceptibility [27]. The GG genotype of CTLA-4 + 49A/G was found to be correlated with AAV (PR3-AAV and GPA) in an Indian cohort [18]. The A allele of rs3087243 CTLA has been found to be a protective factor against AAV [28]. The association of programmed cell death 1 (PDCD1) SNPs with AAV alone has not been proven. The combination of CTLA-4 + 49 AA and PDCD-1.5 T SNP was found to be less frequent in patients with AAV compared to controls [27]. Cluster of differentiation 226 (CD226) is a receptor expressed by T cells, NK cells, and some B cells and plays a role in the cytolytic function of T cells [29]. The minor allele (Ser307) of the CD226 Gly307Ser was linked to GPA [30]. The protein tyrosine phosphatase non-receptor type 22 (PTPN22) gene encodes a protein tyrosine phosphatase that induces type I interferon. The R620W SNP of PTPN22 was found to be related to GPA ANCA+ in different studies [31, 32]. Moreover, the R620W polymorphism of PTPN22 has also been related to MPA susceptibility in a British population [28].

Cytokines and cytokine-related genes

Tumor necrosis factor-alfa (TNF- α) is a tissue damage mediator produced by T helper 17 (Th17) and macrophages in response to immune system activation. The TNF-α-238 SNP was found to be associated with GPA in German patients [33]. However, a meta-analysis showed that the TNF-α-308 SNP was not linked with AAV in Europeans [26]. The AA genotype of IL-10 (-1082) was found to be associated in female patients with MPA [34]. A CA repeat polymorphism of the IL-10 gene was associated with GPA [35]. The AA genotype of IL-10 (-1082) was found to be associated with GPA [34]. Wieczorek et al. observed that the IL-10.2 SNP was linked with ANCA-negative EGPA patients [36]. Interferon gamma (IFN-γ) is a mediator that induces pro-inflammatory IL-12 secretion and inhibits anti-inflammatory IL-10 production. IFNγ (+874 T/T) and (+874 A/A) polymorphisms were found to be related to an overall increased risk of AAV [33]. IL2RA is the gene that encodes the alpha chain of the high-affinity IL-2 receptor, which is primarily expressed by T cells, activated B cells, activated monocytes, and NK cells [37]. In the study of Carr et al., it was demonstrated that the IL2RA rs41295061 polymorphism has been associated with AAV in British patients [38]. The leptin/ghrelin system is two mediators with opposing effects in the regulation of the immune system. The 656Lys allele has been shown to be associated with high EGPA and low GPA risk [39]. Interferon regulatory factor 5 (IRF5) is a gene that stimulates the expression of type I interferon, IL-6, IL-12, and TNF-α and is expressed by B cells and monocytes. The rs2004640-G/Exon6-ins/rs2070197-T/rs10954213-G haplotype has been found to be a protective factor in GPA patients [40]. The T allele of rs35705950 oligomeric mucus/gel-forming (MUC5B) was found to be related with AAV susceptibility [41]. ETS proto-oncogene 1 is a transcription factor that affects the expression of cytokine and chemokine genes and plays a role in various immune responses. A study with a Japanese population showed that ETS1 polymorphism was found to be associated with GPA [42]. In a study conducted by Kawasaki et al., the telomerase reverse transcriptase (TERT) rs2736100A and Desmoplakin rs2076295G were associated with MPA and MPO-AAV [43]. Haplotypes 1 and 4 of glucocorticoid receptor and 11β-hydroxysteroid dehydrogenase type 1 gene were associated with clinically relevant inflammatory and metabolic outcomes in ANCA-associated vasculitis [44]. The A/A genotype at position -2518 in the monocyte chemoattractant protein-1 (MCP-1) was associated with a poor prognosis in Swedish patients with AAV [45].

Fc receptors

Receptors for IgG on leucocytes (FcγR) bind to the Fc-part of IgG and serve as a link between the humoral and cellular parts of the immune system [46]. FcγR is divided into three main classes: FcγRI (CD64), FcγRII (CD 32), and FcγRIII (CD16). FcγRI facilitates antigen presentation to T cells, and FcγRIIa induces degranulation and phagocytosis. FcγRIIb mediates downregulation of antibody responses [47]. The FcγR IIIa-V/V158 SNP was found to be related to GPA predisposition. In addition, patients with both R/R131 homozygous for FcγR IIa and F/F158 homozygous for FcγR IIIa were more prone to disease relapse than the other groups [48]. The FcγR IIIb homozygous allele NA1 was related to MPO-ANCA+ in Caucasian patients with AAV [49]. The FcγR IIIb polymorphism was not associated with GPA, but the frequency of this SNP was found higher in patients with renal involvement compared to those without [50]. The A allele of Fcα receptor (FCAR) rs16986050 was associated with a higher susceptibility to GPA. However, the G allele was shown to be more frequent in patients with renal involvement than in those without [50].

Other receptors

CD18 is a β2 integrin chain that contributes to chemotaxis, phagocytosis, and homotypic adhesion. The C44T, AvaII, and T-1G SNPs of the CD18 gene have been related with MPO-ANCA+, but not with PR3-ANCA+ [51]. Killer cell immunoglobulin-like receptors (KIRs) are important receptors that activate or inhibit NK cells by recognizing class I major tissue compatibility complex (MHC) molecules [52]. The frequency of HLA-Bw4 and KIR3DL1 was shown to be higher in MPA patients compared with controls [53]. Leukocyte immunoglobulin-like receptors (LILRs) are mainly expressed in myelomonocytic and B cells [54]. In a Japanese population, rs2241524 in the A allele of LILRA2 was found to be more common in MPA patients compared to controls [55].

Other proteins

The alternative pathway of complement activation increases chemotaxis by activating neutrophils, and as a result, the development of AAV damage is facilitated [56]. The C3F allele was found to be related to PR3-ANCA+ in Swedish patients [57]. Although the C4A3 allele was found to be associated with susceptibility to AAV, it was not correlated with any clinical findings [57]. Defensins are antimicrobial peptidic components involved in a variety of immunomodulatory activities, including the recruitment of adaptive and innate immune cells [58]. Human neutrophil peptides (α-defensin) and human β-defensin 2 (DEFB4) were demonstrated to be related to GPA susceptibility [59]. GPA patients have been shown to have a higher DEFB4 gene copy number than controls [60]. COL11A2 gene encodes the α2(XI) chain of type XI collagen, it is expressed in the inner ear and the nucleus pulposus intervertebral discs. Lyons et al. showed that the rs3130233 and rs3117016 polymorphisms of COL11A2 were found to be related to AAV [11].

Somatic cell mutations

Somatic UBA1, an X-chromosome gene encoding ubiquitin-like modifier-activating enzyme 1, gene mutations are related to vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic (VEXAS) syndrome. The VEXAS syndrome was identified in patients with cytopenias, dysplastic bone marrow, vasculitis, and pulmonary inflammation [61]. The majority of patients have somatic missense mutations in UBA1 that affect p.Met41Leu [62].

Conclusions

The AAV is a systemic small vessel vasculitis characterized by the presence of autoantibodies that recognize neutrophil cytoplasmic antigens. The mechanisms by which neutrophil expression of these species cause autoimmunity are not yet known. Although a combination of genetic predisposition and environmental factors has been suggested to cause the disease in patients with AAV, convincing data to support this is still inconclusive. In recent years, genetic factors that predispose to ANCA development have been better understood through genome-wide association studies and candidate gene studies. The results of genome-wide association studies of AAV demonstrated that HLA DBP1 (rs3117242), proteinase 3 (rs62132295), and SERPINA1 (rs715156) SNPs were related to PR3-ANCA+. Furthermore, these SNPs were found to be associated with PR3-ANCA+ in patients with MPA. Other genetic polymorphisms, such as CTLA4, IRF5, MUC5B, PTPN22, ETS1, and TERT, were also likely to be contributory to AAV. This article provides a review of the relevant literature to determine the genetic basis of AAV. We believe that elucidating the immunogenetics of AAV will lead to the identification of biomarkers to be used in disease progression and possibly the discovery of new therapeutic targets.

Availability of data and materials

Not applicable.

Abbreviations

AAV:

Anti-neutrophil cytoplasmic antibodies-associated vasculitides

ANCA:

Anti-neutrophil cytoplasmic antibody

CD:

Cluster of differentiation

COL11A2:

Collagen type XI alpha 2 chain

CTL4:

Cytotoxic T-lymphocyte associated protein 4

EGPA:

Eosinophilic granulomatosis with polyangiitis

ETS1:

ETS proto-oncogene 1, transcription factor

FCAR:

Fcα receptor

GPA:

Granulomatosis with polyangiitis

GWAS:

Genome-wide association study

HLA:

Human leukocyte antigen

IFN:

Interferon

IFN-γ:

Interferon gamma

IL:

Interleukin

IRF5:

Interferon regulatory factor 5

KIRs:

Killer cell immunoglobulin-like receptors

LILRs:

Leukocyte immunoglobulin-like receptors

MCP-1:

Monocyte chemoattractant protein-1

MHC:

Major histocompatibility complex

MPA:

Microscopic polyangiitis

MPO:

Myeloperoxidase

MUC5B:

Mucin 5B, oligomeric mucus/gel-forming

PDCD1:

Programmed cell death 1

PiZ:

Protease inhibitor Z

PR3:

Proteinase 3

PTPN22:

Protein tyrosine phosphatase non-receptor type 22

SERPINA1:

Serpin family A member 1

TERT:

Telomerase reverse transcriptase

Th17:

T helper 17

TNF:

Tumor necrosis factor

UBA1:

Ubiquitin-like modifier-activating enzyme 1

VEXAS:

Vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic

References

  1. Mahr A, Katsahian S, Varet H, Guillevin L, Hagen EC, Höglund P et al (2013) Revisiting the classification of clinical phenotypes of anti-neutrophil cytoplasmic antibody-associated vasculitis: a cluster analysis. Ann Rheum Dis 72(6):1003–1010

    Article  PubMed  Google Scholar 

  2. Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F et al (2013) 2012 revised international Chapel Hill consensus conference nomenclature of Vasculitides. Arthritis Rheum 65(1):1–11

    Article  CAS  PubMed  Google Scholar 

  3. Kobayashi S, Fujimoto S (2013) Epidemiology of vasculitides: differences between Japan, Europe and North America. Clin Exp Nephrol 17(5):611–614

    Article  PubMed  PubMed Central  Google Scholar 

  4. Watts RA, Gonzalez-Gay MA, Lane SE, Garcia-Porrua C, Bentham G, Scott DG (2001) Geoepidemiology of systemic vasculitis: comparison of the incidence in two regions of Europe. Ann Rheum Dis 60(2):170–172

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Brogan P, Eleftheriou D (2018) Vasculitis update: pathogenesis and biomarkers. Pediatr Nephrol 33(2):187–198

    Article  PubMed  Google Scholar 

  6. Chen M, Kallenberg CG (2010) ANCA-associated vasculitides--advances in pathogenesis and treatment. Nat Rev Rheumatol 6(11):653–664

    Article  CAS  PubMed  Google Scholar 

  7. Salama AD (2018) Genetics and pathogenesis of small-vessel vasculitis. Best Pract Res Clin Rheumatol 32(1):21–30

    Article  PubMed  Google Scholar 

  8. Carmona FD, López-Mejías R, Márquez A, Martín J, González-Gay MA (2019) Genetic basis of vasculitides with neurologic involvement. Neurol Clin 37(2):219–234

    Article  PubMed  Google Scholar 

  9. Katz P, Alling DW, Haynes BF, Fauci AS (1979) Association of Wegener’s granulomatosis with HLA-B8. Clin Immunol Immunopathol 14(2):268–270

    Article  CAS  PubMed  Google Scholar 

  10. Stassen PM, Cohen-Tervaert JW, Lems SP, Hepkema BG, Kallenberg CG, Stegeman CA (2009) HLA-DR4, DR13(6) and the ancestral haplotype A1B8DR3 are associated with ANCA-associated vasculitis and Wegener’s granulomatosis. Rheumatology (Oxford) 48(6):622–625

    Article  CAS  Google Scholar 

  11. Lyons PA, Rayner TF, Trivedi S, Holle JU, Watts RA, Jayne DR et al (2012) Genetically distinct subsets within ANCA-associated vasculitis. N Engl J Med 367(3):214–223

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Merkel PA, Xie G, Monach PA, Ji X, Ciavatta DJ, Byun J et al (2017) Identification of functional and expression polymorphisms associated with risk for antineutrophil cytoplasmic autoantibody-associated vasculitis. Arthritis Rheum 69(5):1054–1066

    Article  CAS  Google Scholar 

  13. Hagen EC, Stegeman CA, D’Amaro J, Schreuder GM, Lems SP, Tervaert JW et al (1995) Decreased frequency of HLA-DR13DR6 in Wegener’s granulomatosis. Kidney Int 48(3):801–805

    Article  CAS  PubMed  Google Scholar 

  14. Jagiello P, Gencik M, Arning L, Wieczorek S, Kunstmann E, Csernok E et al (2004) New genomic region for Wegener’s granulomatosis as revealed by an extended association screen with 202 apoptosis-related genes. Hum Genet 114(5):468–477

    Article  CAS  PubMed  Google Scholar 

  15. Cao Y, Schmitz JL, Yang J, Hogan SL, Bunch D, Hu Y et al (2011) DRB1*15 allele is a risk factor for PR3-ANCA disease in African Americans. J Am Soc Nephrol 22(6):1161–1167

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Tsuchiya N (2013) Genetics of ANCA-associated vasculitis in Japan: a role for HLA-DRB1*09:01 haplotype. Clin Exp Nephrol 17(5):628–630

    Article  CAS  PubMed  Google Scholar 

  17. Wieczorek S, Hellmich B, Gross WL, Epplen JT (2008) 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 58(1):329–330

    Article  CAS  PubMed  Google Scholar 

  18. Singh J, Sharma A, Rani L, Kaur N, Anand S, Saikia B et al (2020) Distinct HLA and non-HLA associations in different subtypes of ANCA-associated vasculitides in North India. Int J Rheum Dis 23(7):958–965

    Article  CAS  PubMed  Google Scholar 

  19. Gencik M, Meller S, Borgmann S, Fricke H (2000) Proteinase 3 gene polymorphisms and Wegener’s granulomatosis. Kidney Int 58(6):2473–2477

    Article  CAS  PubMed  Google Scholar 

  20. Abdgawad M, Hellmark T, Gunnarsson L, Westman KW, Segelmark M (2006) Increased neutrophil membrane expression and plasma level of proteinase 3 in systemic vasculitis are not a consequence of the - 564 a/G promotor polymorphism. Clin Exp Immunol 145(1):63–70

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Reynolds WF, Stegeman CA, Tervaert JW (2002) 463 G/a myeloperoxidase promoter polymorphism is associated with clinical manifestations and the course of disease in MPO-ANCA-associated vasculitis. Clin Immunol 103(2):154–160

    Article  CAS  PubMed  Google Scholar 

  22. Rajp A, Adu D, Savage CO (2007) Meta-analysis of myeloperoxidase G-463/a polymorphism in anti-neutrophil cytoplasmic autoantibody-positive vasculitis. Clin Exp Immunol 149(2):251–256

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Callea F, Gregorini G, Sinico A, Consalez GG, Bossolasco M, Salvidio G et al (1997) α1-antitrypsin (AAT) deficiency and ANCA-positive systemic vasculitis: genetic and clinical implications. Eur J Clin Investig 27(8):696–702

    Article  CAS  Google Scholar 

  24. Mahr AD, Edberg JC, Stone JH, Hoffman GS, St Clair EW, Specks U et al (2010) Alpha1-antitrypsin deficiency-related alleles Z and S and the risk of Wegener’s granulomatosis. Arthritis Rheum 62(12):3760–3767

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lee SS, Lawton JW, Ko KH (2001) Alpha1 antitrypsin phenotypic variability is not associated with ANCA in southern Chinese. Ann Rheum Dis 60(7):725–726

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lee YH, Choi SJ, Ji JD, Song GG (2012) CTLA-4 and TNF-α promoter-308 A/G polymorphisms and ANCA-associated vasculitis susceptibility: a meta-analysis. Mol Biol Rep 39(1):319–326

    Article  CAS  PubMed  Google Scholar 

  27. Slot MC, Sokolowska MG, Savelkouls KG, Janssen RG, Damoiseaux JG, Tervaert JW (2008) Immunoregulatory gene polymorphisms are associated with ANCA-related vasculitis. Clin Immunol 128(1):39–45

    Article  CAS  PubMed  Google Scholar 

  28. Carr EJ, Niederer HA, Williams J, Harper L, Watts RA, Lyons PA et al (2009) Confirmation of the genetic association of CTLA4 and PTPN22 with ANCA-associated vasculitis. BMC Med Genet 10:121

    Article  PubMed  PubMed Central  Google Scholar 

  29. Shibuya A, Campbell D, Hannum C, Yssel H, Franz-Bacon K, McClanahan T et al (1996) DNAM-1, a novel adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity. 4(6):573–581

    Article  CAS  PubMed  Google Scholar 

  30. Wieczorek S, Hoffjan S, Chan A, Rey L, Harper L, Fricke H et al (2009) Novel association of the CD226 (DNAM-1) Gly307Ser polymorphism in Wegener’s granulomatosis and confirmation for multiple sclerosis in German patients. Genes Immun 10(6):591–595

    Article  CAS  PubMed  Google Scholar 

  31. Martorana D, Maritati F, Malerba G, Bonatti F, Alberici F, Oliva E et al (2012) PTPN22 R620W polymorphism in the ANCA-associated vasculitides. Rheumatology (Oxford) 51(5):805–812

    Article  CAS  Google Scholar 

  32. Jagiello P, Aries P, Arning L, Wagenleiter SE, Csernok E, Hellmich B et al (2005) The PTPN22 620W allele is a risk factor for Wegener’s granulomatosis. Arthritis Rheum 52(12):4039–4043

    Article  CAS  PubMed  Google Scholar 

  33. Spriewald BM, Witzke O, Wassmuth R, Wenzel RR, Arnold ML, Philipp T et al (2005) Distinct tumour necrosis factor alpha, interferon gamma, interleukin 10, and cytotoxic T cell antigen 4 gene polymorphisms in disease occurrence and end stage renal disease in Wegener’s granulomatosis. Ann Rheum Dis 64(3):457–461

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bártfai Z, Gaede KI, Russell KA, Muraközy G, Müller-Quernheim J, Specks U (2003) Different gender-associated genotype risks of Wegener’s granulomatosis and microscopic polyangiitis. Clin Immunol 109(3):330–337

    Article  PubMed  Google Scholar 

  35. Zhou Y, Giscombe R, Huang D, Lefvert AK (2002) Novel genetic association of Wegener’s granulomatosis with the interleukin 10 gene. J Rheumatol 29(2):317–320

    CAS  PubMed  Google Scholar 

  36. Wieczorek S, Hellmich B, Arning L, Moosig F, Lamprecht P, Gross WL et al (2008) Functionally relevant variations of the interleukin-10 gene associated with antineutrophil cytoplasmic antibody-negative Churg-Strauss syndrome, but not with Wegener’s granulomatosis. Arthritis Rheum 58(6):1839–1848

    Article  CAS  PubMed  Google Scholar 

  37. Goldstein JD, Pérol L, Zaragoza B, Baeyens A, Marodon G, Piaggio E (2013) Role of cytokines in thymus- versus peripherally derived-regulatory T cell differentiation and function. Front Immunol 4:155

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Carr EJ, Clatworthy MR, Lowe CE, Todd JA, Wong A, Vyse TJ et al (2009) Contrasting genetic association of IL2RA with SLE and ANCA-associated vasculitis. BMC Med Genet 10:22

    Article  PubMed  PubMed Central  Google Scholar 

  39. Wieczorek S, Holle JU, Bremer JP, Wibisono D, Moosig F, Fricke H et al (2010) Contrasting association of a non-synonymous leptin receptor gene polymorphism with Wegener’s granulomatosis and Churg-Strauss syndrome. Rheumatology (Oxford) 49(5):907–914

    Article  CAS  Google Scholar 

  40. Wieczorek S, Holle JU, Müller S, Fricke H, Gross WL, Epplen JT (2010) A functionally relevant IRF5 haplotype is associated with reduced risk to Wegener’s granulomatosis. J Mol Med (Berl) 88(4):413–421

    Article  Google Scholar 

  41. Namba N, Kawasaki A, Sada KE, Hirano F, Kobayashi S, Yamada H et al (2019) Association of MUC5B promoter polymorphism with interstitial lung disease in myeloperoxidase-antineutrophil cytoplasmic antibody-associated vasculitis. Ann Rheum Dis 78(8):1144–1146

    Article  PubMed  Google Scholar 

  42. Kawasaki A, Yamashita K, Hirano F, Sada KE, Tsukui D, Kondo Y et al (2018) Association of ETS1 polymorphism with granulomatosis with polyangiitis and proteinase 3-anti-neutrophil cytoplasmic antibody positive vasculitis in a Japanese population. J Hum Genet 63(1):55–62

    Article  CAS  PubMed  Google Scholar 

  43. Kawasaki A, Namba N, Sada KE, Hirano F, Kobayashi S, Nagasaka K et al (2020) Association of TERT and DSP variants with microscopic polyangiitis and myeloperoxidase-ANCA positive vasculitis in a Japanese population: a genetic association study. Arthritis Res Ther 22(1):246

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hessels AC, Tuin J, Sanders JSF, Huitema MG, van Rossum EFC, Koper JW et al (2019) Clinical outcome in anti-neutrophil cytoplasmic antibody-associated vasculitis and gene variants of 11β-hydroxysteroid dehydrogenase type 1 and the glucocorticoid receptor. Rheumatology (Oxford) 58(3):447–454

    Article  CAS  Google Scholar 

  45. Jönsson N, Erlandsson E, Gunnarsson L, Pettersson Å, Ohlsson S (2018) Monocyte chemoattractant protein-1 in antineutrophil cytoplasmic autoantibody-associated vasculitis: biomarker potential and association with polymorphisms in the MCP-1 and the CC chemokine receptor-2 gene. Mediat Inflamm 2018:6861257

    Article  Google Scholar 

  46. Castro-Dopico T, Clatworthy MR (2019) IgG and Fcγ receptors in intestinal immunity and inflammation. Front Immunol 10:805

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wang Y, Jönsson F (2019) Expression, role, and regulation of neutrophil Fcγ receptors. Front Immunol 10:1958

    Article  PubMed  PubMed Central  Google Scholar 

  48. Dijstelbloem HM, Scheepers RH, Oost WW, Stegeman CA, van der Pol WL, Sluiter WJ et al (1999) Fcgamma receptor polymorphisms in Wegener’s granulomatosis: risk factors for disease relapse. Arthritis Rheum 42(9):1823–1827

    Article  CAS  PubMed  Google Scholar 

  49. Tse WY, Abadeh S, Jefferis R, Savage CO, Adu D (2000) Neutrophil FcgammaRIIIb allelic polymorphism in anti-neutrophil cytoplasmic antibody (ANCA)-positive systemic vasculitis. Clin Exp Immunol 119(3):574–577

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kelley JM, Monach PA, Ji C, Zhou Y, Wu J, Tanaka S et al (2011) IgA and IgG antineutrophil cytoplasmic antibody engagement of fc receptor genetic variants influences granulomatosis with polyangiitis. Proc Natl Acad Sci U S A 108(51):20736–20741

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gencik M, Meller S, Borgmann S, Sitter T, Menezes Saecker AM, Fricke H et al (2000) The association of CD18 alleles with anti-myeloperoxidase subtypes of ANCA-associated systemic vasculitides. Clin Immunol 94(1):9–12

    Article  CAS  PubMed  Google Scholar 

  52. Shen C, Ge Z, Dong C, Wang C, Shao J, Cai W et al (2021) Genetic variants in KIR/HLA-C genes are associated with the susceptibility to HCV infection in a high-risk Chinese population. Front Immunol 12:632353

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Miyashita R, Tsuchiya N, Yabe T, Kobayashi S, Hashimoto H, Ozaki S et al (2006) Association of killer cell immunoglobulin-like receptor genotypes with microscopic polyangiitis. Arthritis Rheum 54(3):992–997

    Article  CAS  PubMed  Google Scholar 

  54. Anderson KJ, Allen RL (2009) Regulation of T-cell immunity by leucocyte immunoglobulin-like receptors: innate immune receptors for self on antigen-presenting cells. Immunology. 127(1):8–17

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Mamegano K, Kuroki K, Miyashita R, Kusaoi M, Kobayashi S, Matsuta K et al (2008) Association of LILRA2 (ILT1, LIR7) splice site polymorphism with systemic lupus erythematosus and microscopic polyangiitis. Genes Immun 9(3):214–223

    Article  CAS  PubMed  Google Scholar 

  56. Jennette JC, Falk RJ, Hu P, Xiao H (2013) Pathogenesis of antineutrophil cytoplasmic autoantibody-associated small-vessel vasculitis. Annu Rev Pathol 8:139–160

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Persson U, Truedsson L, Westman KW, Segelmark M (1999) C3 and C4 allotypes in anti-neutrophil cytoplasmic autoantibody (ANCA)-positive vasculitis. Clin Exp Immunol 116(2):379–382

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Selsted ME, Ouellette AJ (2005) Mammalian defensins in the antimicrobial immune response. Nat Immunol 6(6):551–557

    Article  CAS  PubMed  Google Scholar 

  59. Vordenbäumen S, Timm D, Bleck E, Richter J, Fischer-Betz R, Chehab G et al (2011) Altered serum levels of human neutrophil peptides (HNP) and human beta-defensin 2 (hBD2) in Wegener’s granulomatosis. Rheumatol Int 31(9):1251–1254

    Article  PubMed  Google Scholar 

  60. Zhou XJ, Cheng FJ, Lv JC, Luo H, Yu F, Chen M et al (2012) Higher DEFB4 genomic copy number in SLE and ANCA-associated small vasculitis. Rheumatology (Oxford) 51(6):992–995

    Article  CAS  Google Scholar 

  61. Koster MJ et al (2021) Clinical heterogeneity of the VEXAS syndrome: a case series. Mayo Clin Proc 96(10):2653–2659

    Article  PubMed  Google Scholar 

  62. Grayson PC, Patel BA, Young NS (2021) VEXAS syndrome. Blood. 137(26):3591–3594

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: A.K and M.K. Writing original draft preparation: A.K. Writing, review, and editing: A.K and M.K. Visualization: M.K. Supervision: A.K and M.K. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to A. Kocaaga.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kocaaga, A., Kocaaga, M. An immunogenetic perspective of ANCA-associated vasculitides. Egypt Rheumatol Rehabil 49, 14 (2022). https://doi.org/10.1186/s43166-022-00114-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43166-022-00114-4

Keywords