Hypotheses on the Etiology of Antineutrophil Cytoplasmic Autoantibody–Associated Vasculitis: The Cause Is Hidden, but the Result Is Known : Clinical Journal of the American Society of Nephrology

Journal Logo

In-Depth Reviews

Hypotheses on the Etiology of Antineutrophil Cytoplasmic Autoantibody–Associated Vasculitis

The Cause Is Hidden, but the Result Is Known

de Lind van Wijngaarden, Robert A.F.; van Rijn, Leendert; Hagen, E. Christiaan; Watts, Richard A.; Gregorini, Gina; Tervaert, Jan Willem Cohen; Mahr, Alfred D.; Niles, John L.; de Heer, Emile; Bruijn, Jan A.; Bajema, Ingeborg M.

Author Information
Clinical Journal of the American Society of Nephrology 3(1):p 237-252, January 2008. | DOI: 10.2215/CJN.03550807
  • Free

Abstract

Antineutrophil cytoplasmic autoantibody (ANCA)-associated necrotizing vasculitis was probably first described in 1866 by Kussmaul and Maier (1) as “polyarteritis nodosa.” It was not until the early 1930s when the first case of what was later named Wegener's granulomatosis (WG) was described (2). The disease was named after Friedrich Wegener, who described it as an entity in 1939 (3). In 1985, antibodies associated with the disease were detected and later became known as ANCA (4). WG is a systemic autoimmune disease that can cause damage in various organs. Later, microscopic polyangiitis (MPA) was distinguished as a separate ANCA-associated vasculitis. The disease—or its immunosuppressive treatment—can cause high levels of morbidity and death, especially in patients with renal involvement. Animal experiments have shown that ANCA directed against myeloperoxidase (MPO) can cause vasculitis that resembles human vasculitic disease (5,6); however, the cause of ANCA production remains unresolved.

Theories have been developed to explain how ANCA could interact with neutrophils, along with monocytes and most probably T lymphocytes, to form the lesions that are characteristic of ANCA-associated vasculitis, such as fibrinoid necrosis and granulomas (7). A recent theory of interest has been postulated by Pendergraft et al. (8), stating that an antibody against the complementary peptide of proteinase-3 (PR3) in an idiotypic/anti-idiotypic network is essential to the development of ANCA and to the development of clinical vasculitis, but why and how these ANCA are produced in the first place remains unanswered. Nonetheless, many hypotheses have been developed about the initiating factor for ANCA production. Many factors, either directly or indirectly, have been considered important in the development of ANCA: Silica exposure (9); genetic predisposition (10); bacterial infection by Staphylococcus aureus (11); viral infection by, for instance, parvovirus B19 (12); and thyroid drugs (13) all have been correlated with and held to contribute to the incidence of ANCA-associated vasculitis. Some of these hypotheses are still the focus of ongoing research, whereas others have been put aside. Most of the proposed mechanisms did not disappear from the spotlight because they were proved wrong but because they could not be proved right.

In this review, a number of the most influential hypotheses about the causes of ANCA-associated vasculitis are described. We summarize research activities aimed at proving the possible mechanisms of these causes in relation to the disease. Moreover, this article focuses on the feasibility of the hypotheses and current views on their relevance to our understanding of ANCA production.

Epidemiology: Incidences with Respect to Geography and Seasons

To put the spectrum of ANCA-associated vasculitides into perspective, we first outline the epidemiologic background. Studies describing differences in incidence between the various subtypes of systemic vasculitis must be carefully interpreted with regard to the classification criteria used. Accurate and reliable classification of systemic vasculitis into WG and MPA remains difficult and controversial. The recent development of an algorithm using both the American College of Rheumatology criteria and the Chapel Hill consensus conference definitions for epidemiologic purposes has improved the situation (14).

The annual incidence of the systemic vasculitides as a group is similar among regions of Norway, the United Kingdom, Germany, Spain, and Kuwait, ranging from 11 to 47 patients per million (1518). From north to south, there seems to be a decreasing incidence of WG, complemented by an increasing incidence of MPA, as shown in Figure 1. Remarkably, a recent study from New Zealand found a much higher incidence of WG than of MPA, suggesting a reciprocal gradient for the southern hemisphere (19). The incidence of Churg-Strauss syndrome, a disease also within the spectrum of ANCA-associated vasculitis, was highest in the United Kingdom, compared with Norway and Spain; in the United Kingdom, incidences of WG and MPA were similar (16). Annual incidences for different geographic areas in the northern hemisphere are depicted in Table 1. These topographic incidence differences might indicate a difference in pathogenesis between WG and MPA (15).

In all areas and all disease categories, the incidence was greater in men than in women (16,18,20), except in Germany, where incidences were similar (17). The peak incidence of vasculitis was between 65 and 74 yr of age in the United Kingdom, Spain, Norway, and China, but in Sweden and the United States, these peak age ranges seemed to be somewhat lower (16,2023).

There are few epidemiologic studies from outside Europe or North America, and data on the occurrence of vasculitis in nonwhite populations is sparse. A French study showed a risk two times greater for patients of European ancestry (24). In China, there is a striking preponderance of MPO-ANCA–associated vasculitis, and PR3-associated disease is relatively rare (22,25). A recent retrospective study from Japan has shown an incidence of renal vasculitis similar to that observed in the United Kingdom, but strikingly all of the patients from Japan were classified as having MPA and were MPO-ANCA positive. No patients with WG or PR3-ANCA were observed, whereas in the United Kingdom, there were roughly equal incidences of WG and MPA (26,27). Furthermore, it is generally believed that systemic vasculitis is rare in the Afro-Caribbean populations of the United Kingdom and North America.

Findings that point to an environmental factor inducing ANCA-associated vasculitis are supported by reports on seasonal variation, although this phenomenon is still subject to discussion. Some reports found no seasonal variability (15,21,28,29). Studies from Europe, the United States, and China that did find seasonal trends usually reported a higher incidence of vasculitis and especially WG in winter and a lower incidence in summer (20,22,3035). Some recent reports from Italy and France, however, have identified higher incidences of WG and MPA in summer (35,36). In Australia, anti-PR3 was mostly found in April, May, and June, the fall season in the southern hemisphere (32). This apparent seasonal variability might be explained by seasonal differences in incidence of infections such as influenza or the correlation between influenza vaccinations and predominantly MPA (37,38), which might be explained by a direct effect of the vaccine or by a more general immunologic activation (39). The major confounding factor in the analyses concerning seasonal variation is the precise definition of disease onset: First symptom or clinical diagnosis.

Another remarkable finding was reported in two detailed studies from Norway and Sweden that found 3- to 5-yr trends of peak incidences, with peaks in 1985, 1989 to 1990, and 1994 (20,21) (Figure 2). It was speculated that this trend was due to environmental factors, such as infection (20,21); however, a British report found neither cyclic fluctuation of systemic vasculitis over 15 yr nor an association with peaks of influenza or other infections (34). Some evidence suggests an increase in overall incidence of ANCA-associated vasculitides (21,30,40,41), but this was most likely due to increased recognition after introduction of ANCA testing (30,40,41).

Although geographic differences in incidence of the different vasculitides are striking, they provide only clues to the cause of the diseases. The seasonal differences in incidence might indicate an infectious agent or another environmental factor that is mainly present during or before the season of high incidence. Geographic differences may suggest that the initiating factor in vasculitis has a different temporal distribution or extent in various countries; however, the different genetic backgrounds may be as important, if not more so, in determining the response to triggering or initiating factors.

Silica

Background

Silica is probably the most extensively studied environmental factor hypothesized to play a causative role in the pathogenesis of ANCA-associated glomerulonephritis, predominantly in MPA. After oxygen, silicon (Si) is the most prominent element of the earth's crust (9). Silicates (SiO4) occur in glass and cement, and silicic acid (H4SiO4) is one of the main constituents of soil water, soil itself, and grasses (42). When interpreting studies on the relationship between ANCA-associated vasculitis and silica exposure, the route of silica exposure and silica source may be relevant. In earlier studies of the relationship between silica exposure and disease, only exposure to mineral silica was evaluated. Since the 1980s, biologic forms of silica, such as sand, grass, grain, wood, cotton, wool, quartz, flint, and “coal,” have been considered indicative of silica exposure. Jobs that have high exposure to silica dust and that have been associated with the occurrence of ANCA-associated vasculitis are farming, mill and textile work, sandblasting, lumber work, and drilling (43). Exposure to mineral silica is frequent in jobs such as mining and quarrying and construction work that involves cement, stone, brick, or concrete and also in pottery or china manufacturing (9,43,44).

Silica and Vasculitis

Already by the early 1950s, silica exposure was described as being associated with renal insufficiency (45). In the 1980s, several case reports of rapidly progressive glomerulonephritis in patients who were previously exposed to silica appeared (4650). When ANCA testing became available, the first reports on silica exposure in ANCA-positive patients appeared in the early 1990s (51,52). In patients with pulmonary silicosis and renal failure, a renal biopsy revealed a pauci-immune necrotizing crescentic glomerulonephritis in a number of studies (5257). ANCA positivity was later confirmed in these patients (52,5761), who received a diagnosis of ANCA-associated glomerulonephritis (58,6264). Silica-induced ANCA-positive disease is often associated with a perinuclear (P-ANCA) staining pattern under indirect immunofluorescence and with antibodies directed against MPO (52,65). Although silica-induced disease has been reported in patients with C-ANCA and anti–PR3-ANCA, as well (52,66), the clinical picture for these patients usually justifies a diagnosis of microscopic polyangiitis but very rarely of WG.

Although it has been found that silicosis is a risk factor for developing ANCA, as determined by indirect immunofluorescence, the presence of ANCA need not necessarily be accompanied by clinical vasculitis (6769). In fact, the titers of anti-PR3 and anti-MPO, as determined by ELISA, are usually negative or very low. The explanation for this could be that in 50% of patients with silicosis, antinuclear antibodies are present, allowing an interpretation of P-ANCA. Moreover, many of these patients have a high percentage of rheumatoid factor and high Ig levels (9).

Silica exposure is also associated with other systemic diseases, in particular Sjo[Combining Diaeresis]gren's syndrome, systemic sclerosis, and systemic lupus erythematosus (70), although for the last, the association is disputed (43). Next to silica, asbestos exposure was reported to be associated with ANCA positivity (71), but this finding was contradicted in case-control studies (72,73); however, patients who are exposed to asbestos can be exposed to silica in the same occupations (74).

Epidemiologic Studies

To find epidemiologic evidence for a relationship between silica exposure and renal failure, several case-control studies have been performed (44,52,75,76). The results showed that among patients with ANCA-associated rapidly progressive glomerulonephritis or WG, 22 to 46% were previously exposed to silica (43,44,52,72,73). Silica exposure in each study was significantly more frequent in patients compared with control subjects. Most of the studies defined silica exposure in terms of duration rather than intensity. Recently, this choice was justified by the finding that duration was more important than intensity in the onset of ANCA-associated vasculitis (77), although other reports contradicted this finding (78,79). Of note is the difference between silica exposure and silicosis. At time of diagnosis with vasculitic disease, some patients exhibit a picture of pulmonary silicosis, whereas others do not. This difference may arise from the limitations of imaging techniques. Moreover, predisposition to vasculitis does not require the presence of severe pulmonary lesions (9,74).

Risk Factors

The finding that farming and livestock exposure are risk factors for primary systemic vasculitides (73,79) supports the causative role of silica in their pathogenesis. Other environmental risk factors are exposure to fumes or materials from construction; pesticides (29); exposure to hydrocarbons such as paint, solvent, cleaning agents, and diesel (80); and air pollution after an earthquake (81). The association between silica and ANCA-associated vasculitis is not always obvious, however. In a survey (23) among 701 patients with ANCA-associated vasculitis, no association with environmental exposure, occupation, or hobby was found. Among those who were exposed to silica, there was no increased frequency of ANCA compared with control subjects (67). The different outcomes of these studies probably arise from differences in methods of establishing silica exposure.

Hypotheses

Although silica exposure has been shown to evoke an immune response (8285) and inflammatory reactions (86), its role in the cause of ANCA-associated glomerulonephritis is not well understood. Several hypotheses have been described. It has been suggested that at the site of pulmonary lesions, which can be caused by silica exposure, cytokines released by activated macrophages can attract polymorphonuclear cells. These cells can express the ANCA antigens PR3 and MPO and are taken up by pulmonary macrophages (8789). Another hypothesis is that alveolar macrophages that are exposed to silica, especially quartz, release a large amount of lysosomal enzymes, such as PR3 and MPO, and reactive oxygen species (9092). Release of these compounds is even greater when the macrophage or monocyte enters apoptosis (93). Silica can cause apoptosis of monocytes and macrophages and probably also of neutrophils by rupturing their phagolysosomal membrane (93,94). Binding of ANCA to their antigens on apoptotic cells amplifies the release of lysosomal enzymes and reactive oxygen species (95). Furthermore, silica may decrease total lymphocyte counts, which might explain the observed lymphopenia reported in some studies (96). In addition, in vitro silica inactivates α1-antitrypsin (97), the natural inhibitor of PR3. Although not all associations between silica exposure and vasculitis are clear-cut, these findings may suggest a causative role of silica in ANCA-associated glomerulonephritis and vasculitis.

One explanation for the geographic differences in vasculitis incidence is the climate. If silica or farming is related to the development of (predominantly MPO-associated) vasculitis, then it would be expected that in wet weather, silica particles would be scattered to a lesser extent than in dry weather. This difference could explain the relatively higher prevalence of vasculitis with MPO-ANCA in warmer countries, illustrated by a high incidence of patients with ANCA directed against MPO in Kuwait (18). The high incidence of MPO-ANCA–associated vasculitis may be explained by the amount of sand present in the air in this region. The role that farming plays in a society might also explain differences in mainly MPO-ANCA–associated vasculitis. This weather-related hypothesis is supported by the higher incidence of vasculitis with MPO-ANCA in Spain combined with the fact that 6% of the population engages in agricultural employment, as opposed to 2% in the United Kingdom (98).

These climate-based considerations could be applied only to exposure to silica dust (e.g., farming, air pollution), not to people who are exposed to occupational mineral silica. Geographic differences could arise from the almost exclusive link between silica exposure and P-ANCA/anti–MPO-positive MPA that is prevalent in southern Europe and Japan.

Staphylococcus aureus

Background

Microbial infections have been associated with initiation and relapse of WG (99,100), and there are several case reports of patients with WG preceded by infections (87,101,102). In particular, S. aureus is often cultured in these cases (103,104). Furthermore, several patients developed ANCA-associated systemic vasculitis during subacute bacterial endocarditis (105,106). Of note, in all cocaine users with cocaine-induced midline destructive lesions, S. aureus nasal colonization was present, and some of these patients were ANCA positive (107). This observation emphasizes the need for awareness regarding chronic bacterial infections in patients with ANCA-associated vasculitis, because these patients can be treated with antibiotics (105,108,109). Treatment with trimethoprim-sulfamethoxazole has been described as successful in patients with WG, even as monotherapy (103,104), helping them achieve remission (110119) or prevent relapses (113). Although this effect could be attributed to the immunosuppressant activity of trimethoprim-sulfamethoxazole, its effects could also be ascribed to its anti-staphylococcal properties. In line with these characteristics, hypotheses have been proposed that infections, particularly in the upper or lower respiratory tract, may play a role in the pathogenesis of WG (120).

The most extensively studied bacterial association with ANCA-associated vasculitis involves S. aureus found in the nasal cavity, which is often affected in WG. A study of 57 patients with WG identified the chronic nasal presence of S. aureus as an independent risk factor for relapse (121), suggesting a role for S. aureus in the cause of ANCA-associated vasculitis. More recently, the presence of nasal S. aureus expressing the superantigen staphylococcal-toxic-shock-syndrome-toxin 1 was identified as a risk factor for relapse in WG (122). CD4+ T cells from the peripheral blood of patients with WG showed reactivity to S. aureus, and a substantial number also recognized PR3, suggesting a role for staphylococci-specific CD4+ T cells in triggering the immune response (123). In an animal model, however, after immunization with S. aureus, no significant T cell proliferation in response to S. aureus could be observed (124).

Other bacterial infections, such as Stenotrophomonas (Pseudomonas) maltophilia, Pseudomonas aeruginosa, and Haemophilus influenzae, have also been associated with crescentic glomerulonephritis (102,125). In these cases, vasculitis was noticed after chronic bronchial suppuration, which is hypothesized to be responsible for MPO-ANCA formation and subsequently causing vasculitis (126128).

Hypotheses

Different hypotheses have been postulated in extensive reviews on the etiology behind infections that lead to vasculitis (11,119,129131), most of which could act in proximity with each other. Bacterial toxins, such as those produced by S. aureus, may function as superantigens that may unrestrictedly stimulate B and T cells, resulting in ANCA production (130,132135); however, part of this hypothesis was more or less debunked by the fact that no relationship could be demonstrated between T cell expansion and S. aureus or its superantigens (136). Otherwise, a staphylococcal acid phosphatase may be nephritogenic (137,138) and, bound to endothelial cells, can act as a planted antigen and be recognized by sera of patients with WG (138). Molecular mimicry could also explain the pathogenesis (139); granzyme B, a serine protease with strong similarity to PR3, is induced by S. aureus enterotoxin A (140,141). Lawyer et al. (140) postulated that the S. aureus genome can also directly encode serine proteases with antigenic cross-reactivity to the C-ANCA autoantigen. Molecular mimicry of the complementary peptide to PR3 could also explain the onset of the pathogenic mechanism (8).

Infections cause a rise in proinflammatory cytokines (e.g., TNF-α, IL-1β, IFN-γ), leading to the production of PR3 and bringing it to the cell surface, thereby exposing it to ANCA (119,142). Although this mechanism cannot be responsible for ANCA induction, it can play a role in relapse of disease (143). Finally, infections promote interaction between B and T cells, supposedly by molecular mimicry, mediated in an HLA-II–dependent manner and resulting in differentiation of ANCA-producing plasma cells (119,139,144). In summary, infections can induce autoimmune responses by antigenic mimicry and by enhancing immunogenicity of host antigens as a result of triggering of the innate immune system (145).

As observed, many mechanisms have been proposed for S. aureus to explain its role in the cause of ANCA-associated vasculitis. Despite numerous indications for an association between ANCA-associated vasculitis and infections in general—and S. aureus infection in particular—no attempt has been successful in fully explaining their cause-and-effect relationship, and their connection remains poorly understood.

Epidemiology

With regard to incidence, both the 4- to 5-yr cyclic pattern and the seasonal fluctuations are hypothesized to be associated with infections of bacterial or viral origin (20,21). Underlining this hypothesis, the peak incidences of S. aureus infection have been reported in the wet season in Australia (146). This relationship may possibly explain the higher incidence of vasculitis directly after the wet season, in winter, but also the higher incidence of vasculitis with ANCA directed to PR3 in countries with more rainy days, such as those in Northern Europe. Another explanation for a higher incidence of ANCA-associated vasculitis in winter is the higher incidence of respiratory illness, such as that arising from influenza infection, when S. aureus residing in the nose can cross the damaged nasal epithelium.

Viral Infections

There is not much supportive evidence for the hypothesis that ANCA-associated vasculitides are caused or triggered by a virus. Among the few viruses that are suspected to play a role, parvovirus B19 likely represents the most compelling candidate (12). B19 has been associated with a variety of autoimmune diseases (147), and several publications have reported the co-occurrence of acute B19 infection and WG (12,148,149). Moreover, acute B19 infection may trigger production of C- or P-ANCA and PR3- or MPO-ANCA, and these autoantibodies disappear, at least in some instances, once the infection has subsided (147,150,151); however, none of these cases of ANCA-positive B19 infection presented with clinical signs suggestive of systemic vasculitis (147,150,151), and larger serologic (152,153) or molecular investigations (153,154) have not supported the relationship between B19 infection and ANCA-associated vasculitis. Taken together, B19 might constitute a possible but not a predominant cause or trigger of ANCA-associated vasculitis.

Hepatitis B virus infection has been closely linked to polyarteritis nodosa (155), but the data to support that this or other hepatotropic viruses might be involved in ANCA-associated vasculitis are not convincing. The presence of anti–hepatitis B antibodies has been occasionally observed in WG (156), including a study that detected hepatitis B antigen in three (20%) of 15 patients with WG as compared with 3% of the healthy population (156). This finding might point to a pathogenic role of hepatitis B virus, but it could also arise from an increased susceptibility to hepatitis B infection as a consequence of immunosuppressive therapy (156). Conversely, studies have suggested that viral hepatitis might induce the production of ANCA. One study found two P-/MPO-ANCA–positive individuals among 22 individuals who were infected with hepatitis B virus (157). Similar reports have emerged regarding the presence of ANCA in patients with hepatitis C infection (158,159), including one study that found ANCA in as many as 56% of patients with a predominant anti-PR3 specificity (158). Although the high prevalence of ANCA in the setting of hepatitis is intriguing, evidence for a causal association between these viruses and ANCA-associated vasculitis remains poor.

A number of other viral illnesses, including Epstein-Barr virus (151), arbovirus (160), HIV (161165), and influenza virus (166), may give rise to ANCA of various types, but these observations do not provide strong support to implicate those viruses in ANCA-associated vasculitis. Results of a study examining bronchoalveolar lavage fluids and lung biopsies of patients WG and active lower airway disease failed to detect viral agents (167).

Genetics

One hypothesis that has been broadly accepted is that genetic factors predispose to the induction of ANCA-associated vasculitis. Several reports have described familial cases of ANCA-associated vasculitis (168173), but such cases are unusual. Numerous studies have been conducted on the genetic susceptibility of both development and relapse of the disease (reviewed in reference [10]). Efforts target gaining insight into the genes involved in immune responses in patients and into factors that determine the expression of target antigens, PR3 and MPO, in and on cells. Polymorphisms of the main inhibitor of PR3, α-1-AT, and of HLA genes have been described as well (174182).

An overview of genetic alterations and their possible clinical implications is given in Table 2. Polymorphisms of HLA genes, predominantly of HLA-DR, have been held responsible for a longer duration of the immune response, in both WG and MPA. In patients with WG and MPA, Hagen et al. (175) found a decreased frequency of HLA-DR13DR6, whereas others (156,183) discovered that the frequency of HLA-DQw7 and DR1 was increased and that of HLA-DR3 was decreased.

ANCA can activate neutrophils by binding to their Fcγ receptors (FcγR). The co-occurrence of the homozygous polymorphisms FcγRIIa-H/H131 and FcγRIIIa-V/V158 has been identified as a risk factor for development of WG (184). These polymorphisms result in a decrease of Fc receptor–mediated clearance, promoting a chronic nasal presence of S. aureus. Moreover, these polymorphisms could bind more IgG1 and IgG3, suggesting a stronger interaction with ANCA (184). As for complement, the C4A3 and C3F were increased in ANCA-positive vasculitis, whereas the C4B allele seemed to be increased in PR3-ANCA–positive patients, possibly modulating the immune response and influencing antibody production (185).

Other polymorphisms in relation to immune responses were also studied. CD18 gene polymorphisms were found to be associated with MPO-ANCA–positive vasculitis (186), indicating a facilitated degranulation and respiratory burst by an increase in adhesion of polymorphonuclear neutrophils to endothelial cells (187). Furthermore, regarding genes that encode inhibitory molecules of T cell activation, a microsatellite of CTLA-4 has been associated with WG (188190) and may account for increased T cell activation (190192).

In WG and MPA, especially in women, a polymorphism of IL-10, was more frequent compared with healthy control subjects (193). Differences in frequencies of polymorphisms in other genes encoding for proinflammatory cytokines, such as TNF-α, IL-1, IL-1β, IL-2, IL-5Rα, and IL-6, could not be found (187,188,191), although others found increased polymorphisms of the TNF-α, the IFN-γ, and the TGF-β1 genes (194196). IL-1 and IL-1R antagonist genotypes (197) and polymorphisms of the IFN-γ and CTLA-4 genes were associated with ESRD in PR3-ANCA–positive vasculitis (194).

A homozygous phenotype of deficient α1-AT and a heterozygous deficient/normal phenotype were associated with PR3-ANCA–positive vasculitis (174) and may contribute to disease induction. Moreover, heterozygote α1-AT deficiency—as compared with homozygote nondeficiency—in PR3-ANCA–associated vasculitis increases the risk for dissemination of the vasculitic process and the risk for fatal outcome (198).

As for the PR3 gene itself, the promoter region seemed to be overexpressed in WG (199). Polymorphisms in the promoter region of the MPO gene have been associated with an increased risk for development of MPO-ANCA–associated vasculitis but also with increased relapse and earlier age of diagnosis (200). Moreover, an increase in PR3 and MPO mRNA expression in circulating leukocytes was associated with ANCA-associated glomerulonephritis (201). PR3 membrane expression was significantly increased in patients with ANCA-associated vasculitis as compared with healthy subjects, suggesting a genetic susceptibility (202,203). More evidence for genetically controlled PR3 membrane expression came from a study that found a strong correlation between the percentage of PR3-positive polymorphonuclear neutrophils in identical twins (204). Anti-PR3 antibody was shown to induce activation of neutrophils with high PR3 membrane expression in a dosage-dependent manner, whereas high PR3 membrane expression seemed to be associated with relapse, possibly mimicking the initial activation step of induction of PR3-ANCA–positive vasculitis (205,206).

Pharmacologic Induction

Numerous articles have appeared on ANCA positivity arising from administration of propylthiouracil (PTU) for anti-thyroid treatment in patients with Graves disease, hyperthyroidism, and thyrotoxicosis (13,207209). The majority of these patients had ANCA directed against MPO, although ANCA that were simultaneously directed against other antigens such as human leukocyte elastase, lactoferrin, and even PR3 were also described (210,211). Even cases of classical WG induced by PTU have been reported (212,213); however, a number of patients showed no clinical signs of vasculitis, despite persistent positive PR3-ANCA titers (211). In 4.1 to 64.0% of patients who were treated with PTU, MPO-ANCA levels could be detected but were detectable in only 0 to 3.4% of patients who were treated with methimazole and in 0 to 5.9% of untreated patients (207,211,212,214217). Although in some cases, the development of anti–MPO-ANCA seems to be related to PTU treatment, only a minority of patients with thyroid disease, PTU treatment, and anti-MPO antibodies developed clinical vasculitis (211,216,217). Usually, with cessation of PTU treatment, MPO levels decreased significantly and sometimes even vanished (207,217). One case report even described a change in ANCA type in a patient with WG from PR3-ANCA to MPO-ANCA after PTU therapy and a switch back upon cessation of the therapy (218). In most patients with vasculitis, these symptoms disappeared when PTU therapy was stopped (218), although additional immunosuppressive therapy may still be needed; therefore, PTU may be regarded as a factor inducing ANCA and (transient) vasculitis in a specific group of patients under specific circumstances.

Hypotheses for how PTU can induce vasculitis or modulate the immune system remain controversial. PTU therapy has been shown to reduce intrathyroidal CD4+ lymphocytes, whereas CD8+ T cells were increased (219). Whereas some reports (220,221) claimed that PTU induced polyclonal activation of B cells, thereby causing vasculitis, in other studies (222,223), no difference in early T and B cell activation after PTU treatment was demonstrated. MPO is necessary to transform PTU into its reactive product, which induces neutrophil-dependent cytotoxicity (224). In animal models, an induction of T cell sensitization (225) and a decrease in T cell proliferation appeared in PTU-treated mice, but no differences in B cell response were apparent (226). Thus, the link between PTU and vasculitis remains unclear.

The occurrence of ANCA-associated vasculitis and glomerulonephritis has also been associated with hydralazine treatment (227229), administered for hypertension. Again, MPO is the most important antigen that ANCA target in these patients (230,231). Some case reports suggested that, next to PTU and hydralazine, a variety of other medications, such as penicillamine, minocycline, allopurinol, sulfasalazine, levamisole, and thioridazine, were associated with ANCA-associated vasculitis (232236); however, in larger studies, these hypotheses could not be confirmed (237). The patient's condition usually improved with withdrawal of the drug (232234).

It is possible that drug allergies are responsible for the association between these drug treatments and the development of ANCA-associated vasculitis (79). Allergies in general have a relatively high prevalence among patients with vasculitis and their families (238). Theories have been postulated about the role for a Th2 (atopic) cytokine environment, and these ideas find support in the association of allergies with vasculitis; however, drug allergies can be evoked by antibiotics and therefore be a surrogate marker for previous infection (79), which has been correlated with the occurrence of vasculitis, as described.

Conclusions

Evidence accumulates supporting a pathogenetic role for ANCA in ANCA-associated vasculitides, and many factors of various origins have been assigned a part in its cause (Figure 3). Environmental factors, such as silica, bacterial or viral infectious agents, medication, and genetic susceptibility, all have been described as being involved in either creating the environment for inducing ANCA production or inducing ANCA themselves. Although the typically advanced age at disease onset suggests an environmental cause rather than genetic factors, genetic differences have been identified between patients and control subjects. Differences in incidence by season and by geographic region could point to an environmental agent, such as S. aureus, as triggering predominantly PR3-ANCA–associated disease or to silica as triggering predominantly MPO-ANCA–associated disease; however, these factors cannot explain the etiologic role of medication. For each environmental factor, exposure does not in all cases result in ANCA-associated vasculitis, and these factors are not necessarily associated with all patients with vasculitis. Although it is important to identify etiologic and risk factors for disease, not much is known about how these factors contribute to or are necessary for disease onset. The induction of ANCA-associated vasculitis seems multifactorial, with an interplay of environmental factors and genetic predisposition creating the environment for development of disease.

Disclosures

None.

F1-37
Figure 1:
Map of Europe, the Middle East, and North Africa in which an impression of the geographic distribution of incidence numbers of antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis is given. CSS, Churg-Strauss syndrome; MPA, microscopic polyangiitis; WG, Wegener's granulomatosis.
F2-37
Figure 2:
Incidence of WG in northern Norway between 1984 and 1998 (solid line) (21) and incidence of ANCA-associated vasculitis with renal involvement in O[Combining Diaeresis]rebro, Sweden, between 1975 and 1995 (dashed line) (20). Of note are the concordant peaks in incidences in 1985, 1989 to 1990, and 1994.
F3-37
Figure 3:
In people with a genetic predisposition, the interplay of various environmental factors can eventually lead to ANCA-associated vasculitis.
T1-37
Table 1:
Annual incidence of primary systemic vasculitis in different regions in Europe and the Middle Easta
T2-37
Table 2:
Genetic alterations and their possible clinical implications in patients with ANCA-associated vasculitisa

Published online ahead of print. Publication date available at www.cjasn.org.

a Publius Ovidius Naso, Metamorphoses; IV: 287

References

1. Kussmaul A, Maier R: About a not previously described peculiar arterial disease (Periarteris nodosa), that coincides with Morbus Brightii and rapidly progressive muscular palsy [in German]. Deutches Arch Klin Med1 :484– 518,1866
2. Klinger H: Borderline variants of periarteritis nodosa [in German]. Frankfurt Z Pathol42 :455– 480,1932
3. Wegener F: About a peculiar rhinogenic granulomatosis with marked involvement of the arterial system and kidneys [in German]. Beitr Pathol Anat102 :36– 68,1939
4. van der Woude FJ, Rasmussen N, Lobatto S, Wiik A, Permin H, van Es LA, van der GM, van der Hem GK: The TH: Autoantibodies against neutrophils and monocytes: Tool for diagnosis and marker of disease activity in Wegener's granulomatosis. Lancet1 :425– 429,1985
5. Xiao H, Heeringa P, Hu P, Liu Z, Zhao M, Aratani Y, Maeda N, Falk RJ, Jennette JC: Antineutrophil cytoplasmic autoantibodies specific for myeloperoxidase cause glomerulonephritis and vasculitis in mice. J Clin Invest110 :955– 963,2002
6. Little MA, Smyth CL, Yadav R, Ambrose L, Cook HT, Nourshargh S, Pusey CD: Antineutrophil cytoplasm antibodies directed against myeloperoxidase augment leukocyte-microvascular interactions in vivo. Blood106 :2050– 2058,2005
7. Day CJ, Hewins P, Savage CO: New developments in the pathogenesis of ANCA-associated vasculitis. Clin Exp Rheumatol21 :S35– S48,2003
8. Pendergraft WF III, Preston GA, Shah RR, Tropsha A, Carter CW, Jr., Jennette JC, Falk RJ: Autoimmunity is triggered by cPR-3(105–201), a protein complementary to human autoantigen proteinase-3. Nat Med10 :72– 79,2004
9. Gregorini G, Tira P, Frizza J, D'Haese PC, Elseviers MM, Nuyts G, Maiorca R, De Broe ME: ANCA-associated diseases and silica exposure. Clin Rev Allergy Immunol15 :21– 40,1997
10. Kallenberg CG, Rarok A, Stegeman CA: Genetics of ANCA-associated vasculitides. Cleve Clin J Med69[Suppl 2] :SII61– SII63,2002
11. Popa ER, Stegeman CA, Kallenberg CG, Tervaert JW: Staphylococcus aureus and Wegener's granulomatosis. Arthritis Res4 :77– 79,2002
12. Finkel TH, Torok TJ, Ferguson PJ, Durigon EL, Zaki SR, Leung DY, Harbeck RJ, Gelfand EW, Saulsbury FT, Hollister JR: Chronic parvovirus B19 infection and systemic necrotising vasculitis: Opportunistic infection or aetiological agent? Lancet343 :1255– 1258,1994
13. Dolman KM, Gans RO, Vervaat TJ, Zevenbergen G, Maingay D, Nikkels RE, Donker AJ, dem Borne AE, Goldschmeding R: Vasculitis and antineutrophil cytoplasmic autoantibodies associated with propylthiouracil therapy. Lancet342 :651– 652,1993
14. Watts R, Lane S, Hanslik T, Hauser T, Hellmich B, Koldingsnes W, Mahr A, Segelmark M, Cohen-Tervaert JW, Scott D: Development and validation of a consensus methodology for the classification of the ANCA-associated vasculitides and polyarteritis nodosa for epidemiological studies. Ann Rheum Dis66 :222– 227,2007
15. Watts RA, Gonzalez-Gay MA, Lane SE, Garcia-Porrua C, Bentham G, Scott DG: Geoepidemiology of systemic vasculitis: Comparison of the incidence in two regions of Europe. Ann Rheum Dis60 :170– 172,2001
16. Watts RA, Lane SE, Scott DG, Koldingsnes W, Nossent H, Gonzalez-Gay MA, Garcia-Porrua C, Bentham GA: Epidemiology of vasculitis in Europe. Ann Rheum Dis60 :1156– 1157,2001
17. Reinhold-Keller E, Herlyn K, Wagner-Bastmeyer R, Gutfleisch J, Peter HH, Raspe HH, Gross WL: No difference in the incidences of vasculitides between north and south Germany: First results of the German vasculitis register. Rheumatology (Oxford)41 :540– 549,2002
18. el Reshaid K, Kapoor MM, el Reshaid W, Madda JP, Varro J: The spectrum of renal disease associated with microscopic polyangiitis and classic polyarteritis nodosa in Kuwait. Nephrol Dial Transplant12 :1874– 1882,1997
19. Gibson A, Stamp LK, Chapman PT, O'Donnell JL: The epidemiology of Wegener's granulomatosis and microscopic polyangiitis in a Southern hemisphere region. Rheumatology (Oxford)45 :624– 628,2006
20. Tidman M, Olander R, Svalander C, Danielsson D: Patients hospitalized because of small vessel vasculitides with renal involvement in the period 1975–95: Organ involvement, anti-neutrophil cytoplasmic antibodies patterns, seasonal attack rates and fluctuation of annual frequencies. J Intern Med244 :133– 141,1998
21. Koldingsnes W, Nossent H: Epidemiology of Wegener's granulomatosis in northern Norway. Arthritis Rheum43 :2481– 2487,2000
22. Xin G, Zhao MH, Wang HY: Detection rate and antigenic specificities of antineutrophil cytoplasmic antibodies in chinese patients with clinically suspected vasculitis. Clin Diagn Lab Immunol11 :559– 562,2004
23. Abdou NI, Kullman GJ, Hoffman GS, Sharp GC, Specks U, McDonald T, Garrity J, Goeken JA, Allen NB: Wegener's granulomatosis: Survey of 701 patients in North America—Changes in outcome in the 1990s. J Rheumatol29 :309– 316,2002
24. Mahr A, Guillevin L, Poissonnet M, Ayme S: Prevalences of polyarteritis nodosa, microscopic polyangiitis, Wegener's granulomatosis, and Churg-Strauss syndrome in a French urban multiethnic population in 2000: A capture-recapture estimate. Arthritis Rheum51 :92– 99,2004
25. Li PK, Leung JC, Lai FM, Wang A, Lui SF, Leung CB, Lai KN: Use of antineutrophil cytoplasmic autoantibodies in diagnosing vasculitis in a Chinese patient population. Am J Nephrol14 :99– 105,1994
26. Fujimoto S, Uezono S, Hisanaga S, Fukudome K, Kobayashi S, Suzuki K, Hashimoto H, Nakao H, Nunoi H: Incidence of ANCA-associated primary renal vasculitis in the Miyazaki Prefecture: The first population-based, retrospective, epidemiologic survey in Japan. Clin J Am Soc Nephrol1 :1016– 1022,2006
27. Watts RA, Scott DGI, Fujimoto S, Kobayashi S, Suzuki K, Shigeto S, Hashimoto H, Jayne DRW: Epidemiology of renal ANCA-associated vasculitis in the UK and Japan. Clin Exp Rheumatol25 :s95 ,2007
28. Cotch MF, Hoffman GS, Yerg DE, Kaufman GI, Targonski P, Kaslow RA: The epidemiology of Wegener's granulomatosis: Estimates of the five-year period prevalence, annual mortality, and geographic disease distribution from population-based data sources. Arthritis Rheum39 :87– 92,1996
29. Duna GF, Cotch MF, Galperin C, Hoffman DB, Hoffman GS: Wegener's granulomatosis: Role of environmental exposures. Clin Exp Rheumatol16 :669– 674,1998
30. Carruthers DM, Watts RA, Symmons DP, Scott DG: Wegener's granulomatosis: Increased incidence or increased recognition? Br J Rheumatol35 :142– 145,1996
31. Raynauld JP, Bloch DA, Fries JF: Seasonal variation in the onset of Wegener's granulomatosis, polyarteritis nodosa and giant cell arteritis. J Rheumatol20 :1524– 1526,1993
    32. Jennings JG, Chang L, Savige JA: Anti-proteinase 3 antibodies, their characterization and disease associations. Clin Exp Immunol95 :251– 256,1994
    33. Jennette JC, Falk RJ: Antineutrophil cytoplasmic autoantibodies and associated diseases: A review. Am J Kidney Dis15 :517– 529,1990
      34. Watts RA, Lane SE, Bentham G, Scott DG: Seasonal and periodic variation in primary systemic vasculitis (PSV). Cleve Clin J Med69 :SII167 ,2002
      35. Pavone L, Grasselli C, Chierici E, Maggiore U, Garini G, Ronda N, Manganelli P, Pesci A, Rioda WT, Tumiati B, Pavesi G, Vaglio A, Buzio C: Outcome and prognostic factors during the course of primary small-vessel vasculitides. J Rheumatol33 :1299– 1306,2006
      36. Mahr A, Artigues N, Coste J, Aouba A, Pagnoux C, Guillevin L: Seasonal variations in onset of Wegener's granulomatosis: Increased in summer? J Rheumatol33 :1615– 1622,2006
      37. Blumberg S, Bienfang D, Kantrowitz FG: A possible association between influenza vaccination and small-vessel vasculitis. Arch Intern Med140 :847– 848,1980
      38. Kelsall JT, Chalmers A, Sherlock CH, Tron VA, Kelsall AC: Microscopic polyangiitis after influenza vaccination. J Rheumatol24 :1198– 1202,1997
      39. Uji M, Matsushita H, Iwata S: Microscopic polyangiitis after influenza vaccination. Intern Med44 :892– 896,2005
      40. Lane SE, Scott DG, Heaton A, Watts RA: Primary renal vasculitis in Norfolk: Increasing incidence or increasing recognition? Nephrol Dial Transplant15 :23– 27,2000
      41. Andrews M, Edmunds M, Campbell A, Walls J, Feehally J: Systemic vasculitis in the 1980s: Is there an increasing incidence of Wegener's granulomatosis and microscopic polyarteritis? J R Coll Physicians Lond24 :284– 288,1990
      42. Epstein E: The anomaly of silicon in plant biology. Proc Natl Acad Sci U S A91 :11– 17,1994
      43. Hogan SL, Satterly KK, Dooley MA, Nachman PH, Jennette JC, Falk RJ: Silica exposure in anti-neutrophil cytoplasmic autoantibody-associated glomerulonephritis and lupus nephritis. J Am Soc Nephrol12 :134– 142,2001
      44. Nuyts GD, Van Vlem E, De Vos A, Daelemans RA, Rorive G, Elseviers MM, Schurgers M, Segaert M, D'Haese PC, De Broe ME: Wegener granulomatosis is associated to exposure to silicon compounds: A case-control study. Nephrol Dial Transplant10 :1162– 1165,1995
      45. Saita G, Zavaglia O: Renal function in silicotics. Med Lav42 :41– 48,1951
      46. Slavin RE, Swedo JL, Brandes D, Gonzalez-Vitale JC, Osornio-Vargas A: Extrapulmonary silicosis: A clinical, morphologic, and ultrastructural study. Hum Pathol16 :393– 412,1985
      47. Perez Perez AJ, Sobrado J, Cigarran S, Courel M, Gonzalez L, Fernandez R, Perez VJ: Renal vasculitis, diffuse pulmonary hemorrhage and silicosis: Analysis of 2 cases. Med Clin (Barc)87 :858– 860,1986
        48. Arnalich F, Lahoz C, Picazo ML, Monereo A, Arribas JR, Martinez AJ, Vazquez JJ: Polyarteritis nodosa and necrotizing glomerulonephritis associated with long-standing silicosis. Nephron51 :544– 547,1989
          49. Dracon M, Noel C, Wallaert B, Dequiedt P, Lelievre G, Tacquet A: Rapidly progressive glomerulonephritis in pneumoconiotic coal miners. Nephrologie11 :61– 65,1990
            50. Sanchez M, Devinuesa SG, Luno J, Barrio V, Lafuente J, Niembro E, Valderrabano F: Rapidly progressive glomerulonephritis and diffuse pulmonary hemorrhage not mediated by anti basement-membrane antibodies. Nefrologia5 :157– 160,1985
            51. Talaszka A, Boulanger E, Le Monies H: Silicosis, anti-myeloperoxidase antibodies and glomerular nephropathy. Nephrologie13 :234 ,1992
            52. Gregorini G, Ferioli A, Donato F, Tira P, Morassi L, Tardanico R, Lancini L, Maiorca R: Association between silica exposure and necrotizing crescentic glomerulonephritis with p-ANCA and anti-MPO antibodies: A hospital-based case-control study. Adv Exp Med Biol336 :435– 440,1993
            53. Bonnin A, Mousson C, Justrabo E, Tanter Y, Chalopin JM, Rifle G: Silicosis associated with crescentic IgA mesangial nephropathy. Nephron47 :229– 230,1987
              54. Sherson D, Jorgensen F: Rapidly progressive crescenteric glomerulonephritis in a sandblaster with silicosis. Br J Ind Med46 :675– 676,1989
                55. Rispal P, Wen L, De Precigout V, Aparicio M: Silicon nephropathy in a dental prosthetist [in French]. Presse Med20 :176 ,1991
                  56. Pouthier D, Duhoux P, Van Damme B: Pulmonary silicosis and glomerular nephropathy: Apropos of 1 case. Nephrologie12 :8– 11,1991
                    57. Neyer U, Woss E, Neuweiler J: Wegener's granulomatosis associated with silicosis. Nephrol Dial Transplant9 :559– 561,1994
                    58. Tervaert JW, Goldschmeding R, Elema JD, van der GM, Huitema MG, van der Hem GK, The TH, dem Borne AE, Kallenberg CG: Autoantibodies against myeloid lysosomal enzymes in crescentic glomerulonephritis. Kidney Int37 :799– 806,1990
                    59. Niles JL, Pan GL, Collins AB, Shannon T, Skates S, Fienberg R, Arnaout MA, Mccluskey RT: Antigen-specific radioimmunoassays for anti-neutrophil cytoplasmic antibodies in the diagnosis of rapidly progressive glomerulonephritis. J Am Soc Nephrol2 :27– 36,1991
                      60. Bachmeyer C, Grateau G, Gomez V, Choukroun G, Noel LH, Choudat D, Sereni D: Periarteritis nodosa in a dental prosthetist. Presse Med23 :446 ,1994
                        61. Chevailler A, Carrere F, Renier G, Hurez D, Subra JF, Reboul P, Riberi P, Masson C: Silicon nephropathy and myeloperoxidase antibodies. Ann Rheum Dis53 :781– 782,1994
                        62. Kallenberg CG, Mulder AH, Tervaert JW: Antineutrophil cytoplasmic antibodies: A still-growing class of autoantibodies in inflammatory disorders. Am J Med93 :675– 682,1992
                        63. Jennette JC, Falk RJ, Andrassy K, Bacon PA, Churg J, Gross WL, Hagen EC, Hoffman GS, Hunder GG, Kallenberg CG: Nomenclature of systemic vasculitides: Proposal of an international consensus conference. Arthritis Rheum37 :187– 192,1994
                          64. Niles JL, Bottinger EP, Saurina GR, Kelly KJ, Pan G, Collins AB, Mccluskey RT: The syndrome of lung hemorrhage and nephritis is usually an ANCA-associated condition. Arch Intern Med156 :440– 445,1996
                          65. Nakajima H, Miyazaki M, Imai N, Yokokawa T, Yamamoto S: A case of silicosis with MPO-ANCA-associated glomerulonephritis and alveolar hemorrhage. Nippon Jinzo Gakkai Shi43 :351– 356,2001
                          66. Siebels M, Schulz V, Andrassy K: Silicosis and systemic diseases. Immun Infekt21[Suppl 1] :53– 54,1993
                          67. Bartunkova J, Pelclova D, Kolarova I, Fenclova Z, Lebedova J, Sediva A, Tesar V: Exposure to silica and ANCA-associated vasculitis. Cleve Clin J Med69[Suppl 2] :SII168 ,2002
                          68. Wichmann I, Sanchez-Roman J, Morales J, Castillo MJ, Ocana C, Nunez-Roldan A: Antimyeloperoxidase antibodies in individuals with occupational exposure to silica. Ann Rheum Dis55 :205– 207,1996
                            69. Bartunkova J, Pelclova D, Fenclova Z, Sediva A, Lebedova J, Tesar V, Hladikova M, Klusackova P: Exposure to silica and risk of ANCA-associated vasculitis. Am J Ind Med49 :569– 576,2006
                            70. Sanchez-Roman J, Wichmann I, Salaberri J, Varela JM, Nunez-Roldan A: Multiple clinical and biological autoimmune manifestations in 50 workers after occupational exposure to silica. Ann Rheum Dis52 :534– 538,1993
                            71. Pelclova D, Bartunkova J, Fenclova Z, Lebedova J, Hladikova M, Benakova H: Asbestos exposure and antineutrophil cytoplasmic Antibody (ANCA) positivity. Arch Environ Health58 :662– 668,2003
                            72. Beaudreuil S, Lasfargues G, Laueriere L, Ghoul ZE, Fourquet F, Longuet C, Halimi JM, Nivet H, Buchler M: Occupational exposure in ANCA-positive patients: A case-control study. Kidney Int67 :1961– 1966,2005
                            73. Stratta P, Messuerotti A, Canavese C, Coen M, Luccoli L, Bussolati B, Giorda L, Malavenda P, Cacciabue M, Bugiani M, Bo M, Ventura M, Camussi G, Fubini B: The role of metals in autoimmune vasculitis: Epidemiological and pathogenic study. Sci Total Environ270 :179– 190,2001
                            74. De Vuyst P, Camus P: The past and present of pneumoconioses. Curr Opin Pulm Med6 :151– 156,2000
                            75. Steenland NK, Thun MJ, Ferguson CW, Port FK: Occupational and other exposures associated with male end-stage renal disease: A case/control study. Am J Public Health80 :153– 157,1990
                            76. Nuyts GD, Van Vlem E, Thys J, De Leersnijder D, D'Haese PC, Elseviers MM, De Broe ME: New occupational risk factors for chronic renal failure. Lancet346 :7– 11,1995
                            77. Hogan SL, Cooper GS, Nylander French LA, Parks CG, Savitz DA, Chin H, Jennette CE, Jennette JC, Falk RJ: Duration of silica exposure and development of ANCA-associated small vessel vasculitis (ANCA-SVV) with glomerular involvement: A case-control study. J Am Soc Nephrol15 :SA-PO182 ,2004
                            78. Watts RA, Lane SE, Bentham G, Innes NJ, Scott DG: Are environmental factors important in systemic vasculitis? Cleve Clin J Med69 :SII166– SII167,2002
                            79. Lane SE, Watts RA, Bentham G, Innes NJ, Scott DG: Are environmental factors important in primary systemic vasculitis? A case-control study. Arthritis Rheum48 :814– 823,2003
                            80. Pai P, Bone JM, Bell GM: Hydrocarbon exposure and glomerulonephritis due to systemic vasculitis. Nephrol Dial Transplant13 :1321– 1323,1998
                            81. Yashiro M, Muso E, Itoh-Ihara T, Oyama A, Hashimoto K, Kawamura T, Ono T, Sasayama S: Significantly high regional morbidity of MPO-ANCA-related angitis and/or nephritis with respiratory tract involvement after the 1995 great earthquake in Kobe (Japan). Am J Kidney Dis35 :889– 895,2000
                            82. McHugh NJ, Whyte J, Harvey G, Haustein UF: Anti-topoisomerase I antibodies in silica-associated systemic sclerosis: A model for autoimmunity. Arthritis Rheum37 :1198– 1205,1994
                            83. Watanabe S, Shirakami A, Takeichi T, Ohara T, Saito S: Alterations in lymphocyte subsets and serum immunoglobulin levels in patients with silicosis. J Clin Lab Immunol23 :45– 51,1987
                              84. Youinou P, Ferec C, Cledes J, Zabbe C, Philippon P, Dewitte JD, Guillerm D, Clavier J: Immunological effect of silica dust analyzed by monoclonal antibodies. J Clin Lab Immunol16 :207– 210,1985
                                85. Pernis B, Paronetto F: Adjuvant effect of silica (tridymite) on antibody production. Proc Soc Exp Biol Med110 :390– 392,1962
                                86. Kallenberg CG: Overlapping syndromes, undifferentiated connective tissue disease, and other fibrosing conditions. Curr Opin Rheumatol7 :568– 573,1995
                                87. Sitara D, Hoffbrand BI: Chronic bronchial suppuration and antineutrophil cytoplasmic antibody (ANCA) positive systemic vasculitis. Postgrad Med J66 :669– 671,1990
                                88. Finnegan MJ, Hinchcliffe J, Russell-Jones D, Neill S, Sheffield E, Jayne D, Wise A, Hodson ME: Vasculitis complicating cystic fibrosis. Q J Med72 :609– 621,1989
                                  89. Pinching AJ, Lockwood CM, Pussell BA, Rees AJ, Sweny P, Evans DJ, Bowley N, Peters DK: Wegener's granulomatosis: Observations on 18 patients with severe renal disease. Q J Med52 :435– 460,1983
                                  90. Allison AC, Harington JS, Birbeck M: An examination of the cytotoxic effects of silica on macrophages. J Exp Med124 :141– 154,1966
                                  91. Sarih M, Souvannavong V, Brown SC, Adam A: Silica induces apoptosis in macrophages and the release of interleukin-1 alpha and interleukin-1 beta. J Leukoc Biol54 :407– 413,1993
                                    92. Vallyathan V, Mega JF, Shi X, Dalal NS: Enhanced generation of free radicals from phagocytes induced by mineral dusts. Am J Respir Cell Mol Biol6 :404– 413,1992
                                    93. Gilligan HM, Bredy B, Brady HR, Hebert MJ, Slayter HS, Xu Y, Rauch J, Shia MA, Koh JS, Levine JS: Antineutrophil cytoplasmic autoantibodies interact with primary granule constituents on the surface of apoptotic neutrophils in the absence of neutrophil priming. J Exp Med184 :2231– 2241,1996
                                    94. Iyer R, Hamilton RF, Li L, Holian A: Silica-induced apoptosis mediated via scavenger receptor in human alveolar macrophages. Toxicol Appl Pharmacol141 :84– 92,1996
                                    95. Tervaert JW, Stegeman CA, Kallenberg CG: Silicon exposure and vasculitis. Curr Opin Rheumatol10 :12– 17,1998
                                    96. Subra JF, Renier G, Reboul P, Tollis F, Boivinet R, Schwartz P, Chevailler A: Lymphopenia in occupational pulmonary silicosis with or without autoimmune disease. Clin Exp Immunol126 :540– 544,2001
                                    97. Zay K, Devine D, Churg A: Quartz inactivates alpha 1-antiproteinase: Possible role in mineral dust-induced emphysema. J Appl Physiol78 :53– 58,1995
                                    98. Food and Agriculture Organization of the United Nations, The Statistics Division. Compendium of Food and Agri-culture Indicators 2006. http://www.fao.org/es/ess/compendium_2006/pdf/SPA_ESS_E.pdf and http://www.fao.org/es/ess/compendium_2006/pdf/UKM_ESS_E.pdf.
                                    99. Fauci AS, Haynes BF, Katz P, Wolff SM: Wegener's granulomatosis: Prospective clinical and therapeutic experience with 85 patients for 21 years. Ann Intern Med98 :76– 85,1983
                                    100. Pinching AJ, Rees AJ, Pussell BA, Lockwood CM, Mitchison RS, Peters DK: Relapses in Wegener's granulomatosis: The role of infection. BMJ281 :836– 838,1980
                                    101. van Putten JW, van Haren EH, Lammers JW: Association between Wegener's granulomatosis and Staphylococcus aureus infection? Eur Respir J9 :1955– 1957,1996
                                    102. Park J, Banno S, Sugiura Y, Yoshikawa K, Naniwa T, Wakita K, Hayami Y, Sato S, Ueda R: Microscopic polyangiitis associated with diffuse panbronchiolitis. Intern Med43 :331– 335,2004
                                    103. Boudewyns A, Verbelen J, Koekelkoren E, Van Offel J, Van de HP: Wegener's granulomatosis triggered by infection? Acta Otorhinolaryngol Belg55 :57– 63,2001
                                    104. Ohtake T, Kobayashi S, Honjou Y, Shirai T, Takayanagi S, Tohyama K, Tokura Y, Kimura M: Generalized Wegener's granulomatosis responding to sulfamethoxazole-trimethoprim monotherapy. Intern Med40 :666– 670,2001
                                    105. Subra JF, Michelet C, Laporte J, Carrere F, Reboul P, Cartier F, Saint-Andre JP, Chevailler A: The presence of cytoplasmic antineutrophil cytoplasmic antibodies (C-ANCA) in the course of subacute bacterial endocarditis with glomerular involvement, coincidence or association? Clin Nephrol49 :15– 18,1998
                                    106. Hellmich B, Ehren M, Lindstaedt M, Meyer M, Pfohl M, Schatz H: Anti-MPO-ANCA-positive microscopic polyangiitis following subacute bacterial endocarditis. Clin Rheumatol20 :441– 443,2001
                                    107. Trimarchi M, Gregorini G, Facchetti F, Morassi ML, Manfredini C, Maroldi R, Nicolai P, Russell KA, McDonald TJ, Specks U: Cocaine-induced midline destructive lesions: Clinical, radiographic, histopathologic, and serologic features and their differentiation from Wegener granulomatosis. Medicine (Baltimore)80 :391– 404,2001
                                    108. Choi HK, Lamprecht P, Niles JL, Gross WL, Merkel PA: Subacute bacterial endocarditis with positive cytoplasmic antineutrophil cytoplasmic antibodies and anti-proteinase 3 antibodies. Arthritis Rheum43 :226– 231,2000
                                    109. Mandell BF, Calabrese LH: Infections and systemic vasculitis. Curr Opin Rheumatol10 :51– 57,1998
                                    110. DeRemee RA, McDonald TJ, Weiland LH: Wegener's granulomatosis: Observations on treatment with antimicrobial agents. Mayo Clin Proc60 :27– 32,1985
                                    111. DeRemee RA: The treatment of Wegener's granulomatosis with trimethoprim/sulfamethoxazole: Illusion or vision? Arthritis Rheum31 :1068– 1074,1988
                                      112. Leavitt RY, Hoffman GS, Fauci AS: The role of trimethoprim/sulfamethoxazole in the treatment of Wegener's granulomatosis. Arthritis Rheum31 :1073– 1074,1988
                                        113. Stegeman CA, Tervaert JW, De Jong PE, Kallenberg CG: Trimethoprim-sulfamethoxazole (co-trimoxazole) for the prevention of relapses of Wegener's granulomatosis. Dutch Co-Trimoxazole Wegener Study Group. N Engl J Med335 :16– 20,1996
                                        114. Israel HL: Sulfamethoxazole-trimethoprim therapy for Wegener's granulomatosis. Arch Intern Med148 :2293– 2295,1988
                                          115. West BC, Todd JR, King JW: Wegener granulomatosis and trimethoprim-sulfamethoxazole: Complete remission after a twenty-year course. Ann Intern Med106 :840– 842,1987
                                            116. Axelson JA, Clark RH, Ancerewicz S: Wegener granulomatosis and trimethoprim-sulfamethoxazole. Ann Intern Med107 :600 ,1987
                                              117. Yuasa K, Tokitsu M, Goto H, Kato H, Shimada K: Wegener's granulomatosis: Diagnosis by transbronchial lung biopsy, evaluation by gallium scintigraphy and treatment with sulfamethoxazole/trimethoprim. Am J Med84 :371– 372,1988
                                                118. Valeriano-Marcet J, Spiera H: Treatment of Wegener's granulomatosis with sulfamethoxazole-trimethoprim. Arch Intern Med151 :1649– 1652,1991
                                                  119. George J, Levy Y, Kallenberg CG, Shoenfeld Y: Infections and Wegener's granulomatosis: A cause and effect relationship? QJM90 :367– 373,1997
                                                  120. Roberts DE, Curd JG: Sulfonamides as antiinflammatory agents in the treatment of Wegener's granulomatosis. Arthritis Rheum33 :1590– 1593,1990
                                                  121. Stegeman CA, Tervaert JW, Sluiter WJ, Manson WL, De Jong PE, Kallenberg CG: Association of chronic nasal carriage of Staphylococcus aureus and higher relapse rates in Wegener granulomatosis. Ann Intern Med120 :12– 17,1994
                                                  122. Popa ER, Stegeman CA, Abdulahad WH, van der MB, Arends J, Manson WM, Bos NA, Kallenberg CG, Cohen Tervaert JW: Staphylococcal toxic-shock-syndrome-toxin-1 as a risk factor for disease relapse in Wegener's granulomatosis. Rheumatology (Oxford)46 :1029– 1033,2007
                                                  123. Mayet WJ, Marker-Hermann E, Schlaak J, Meyer zum Buschenfelde KH: Irregular cytokine pattern of CD4+ T lymphocytes in response to Staphylococcus aureus in patients with Wegener's granulomatosis. Scand J Immunol49 :585– 594,1999
                                                  124. Savige J, Nassis L, Cooper T, Paspaliaris B, Martinello P, MacGregor D: Antineutrophil cytoplasmic antibody (ANCA)-associated systemic vasculitis after immunisation with bacterial proteins. Clin Exp Rheumatol20 :783– 789,2002
                                                  125. Tanaka E, Tada K, Amitani R, Kuze F: Systemic hypersensitivity vasculitis associated with bronchiectasis. Chest102 :647– 649,1992
                                                  126. Saku N, Sugiyama Y, Kitamura S, Fujii T, Saitoh K: Diffuse panbronchiolitis with P-ANCA-positive arteritis and necrotizing glomerulitis. Nihon Kyobu Shikkan Gakkai Zasshi34 :434– 438,1996
                                                  127. Bruce IN, McAteer JA, Gardiner PV, McFarland RJ, Sloan JM, Bell AL: Chronic suppurative lung disease with associated vasculitis. Postgrad Med J71 :24– 27,1995
                                                    128. Miyamoto D, Ichinose Y, Kikawada M, Kusumoto H, Yanagisawa N, Kanai E, Minemura K, Yonemaru M, Toyama K: Diffuse panbronchiolitis with myeloperoxidase-specific antineutrophil cytoplasmic antibody-related vasculitis. Nihon Kokyuki Gakkai Zasshi36 :453– 458,1998
                                                    129. Kallenberg CG, Rarok A, Stegeman CA, Limburg PC: New insights into the pathogenesis of antineutrophil cytoplasmic autoantibody-associated vasculitis. Autoimmun Rev1 :61– 66,2002
                                                    130. Capizzi SA, Specks U: Does infection play a role in the pathogenesis of pulmonary vasculitis? Semin Respir Infect18 :17– 22,2003
                                                    131. Popa ER, Tervaert JW: The relation between Staphylococcus aureus and Wegener's granulomatosis: Current knowledge and future directions. Intern Med42 :771– 780,2003
                                                    132. Fleischer B, Schrezenmeier H: T cell stimulation by staphylococcal enterotoxins: Clonally variable response and requirement for major histocompatibility complex class II molecules on accessory or target cells. J Exp Med167 :1697– 1707,1988
                                                    133. Hamidou M, Belizna C: Superantigens and vasculitis [in French]. Ann Med Interne (Paris)154 :96– 100,2003
                                                      134. Tervaert JW, Popa ER, Bos NA: The role of superantigens in vasculitis. Curr Opin Rheumatol11 :24– 33,1999
                                                        135. Voswinkel J, Muller A, Lamprecht P: Is PR3-ANCA formation initiated in Wegener's granulomatosis lesions? Granulomas as potential lymphoid tissue maintaining autoantibody production. Ann N Y Acad Sci1051 :12– 19,2005
                                                        136. Popa ER, Stegeman CA, Bos NA, Kallenberg CG, Tervaert JW: Staphylococcal superantigens and T cell expansions in Wegener's granulomatosis. Clin Exp Immunol132 :496– 504,2003
                                                        137. Yousif Y, Okada K, Batsford S, Vogt A: Induction of glomerulonephritis in rats with staphylococcal phosphatase: New aspects in post-infectious ICGN. Kidney Int50 :290– 297,1996
                                                        138. Brons RH, Bakker HI, Van Wijk RT, Van Dijk NW, Muller Kobold AC, Limburg PC, Manson WL, Kallenberg CG, Tervaert JW: Staphylococcal acid phosphatase binds to endothelial cells via charge interaction: A pathogenic role in Wegener's granulomatosis? Clin Exp Immunol119 :566– 573,2000
                                                        139. Rasmussen N, Petersen J: Cellular immune responses and pathogenesis in c-ANCA positive vasculitides. J Autoimmun6 :227– 236,1993
                                                        140. Lawyer C, Henkle J, Bakir H: Nasal carriage of staphylococcal infection in Wegener granulomatosis. Ann Intern Med121 :74– 75,1994
                                                        141. Caputo A, Fahey D, Lloyd C, Vozab R, McCairns E, Rowe PB: Structure and differential mechanisms of regulation of expression of a serine esterase gene in activated human T lymphocytes. J Biol Chem263 :6363– 6369,1988
                                                        142. Mayet WJ, Schwarting A, Meyer zum Buschenfelde KH: Cytotoxic effects of antibodies to proteinase 3 (C-ANCA) on human endothelial cells. Clin Exp Immunol97 :458– 465,1994
                                                        143. Huugen D, Xiao H, van Esch A, Falk RJ, Peutz-Kootstra CJ, Buurman WA, Tervaert JW, Jennette JC, Heeringa P: Aggravation of anti-myeloperoxidase antibody-induced glomerulonephritis by bacterial lipopolysaccharide: Role of tumor necrosis factor-alpha. Am J Pathol167 :47– 58,2005
                                                        144. Brouwer E, Stegeman CA, Huitema MG, Limburg PC, Kallenberg CG: T cell reactivity to proteinase 3 and myeloperoxidase in patients with Wegener's granulomatosis (WG). Clin Exp Immunol98 :448– 453,1994
                                                        145. Wraith DC, Goldman M, Lambert PH: Vaccination and autoimmune disease: What is the evidence? Lancet362 :1659– 1666,2003
                                                        146. Skull SA, Krause V, Coombs G, Pearman JW, Roberts LA: Investigation of a cluster of Staphylococcus aureus invasive infection in the top end of the Northern Territory. Aust N Z J Med29 :66– 72,1999
                                                        147. Lehmann HW, Von Landenberg P, Modrow S: Parvovirus B19 infection and autoimmune disease. Autoimmun Rev2 :218– 223,2003
                                                        148. Nikkari S, Mertsola J, Korvenranta H, Vainionpaa R, Toivanen P: Wegener's granulomatosis and parvovirus B19 infection. Arthritis Rheum37 :1707– 1708,1994
                                                        149. Corman LC, Staud R: Association of Wegener's granulomatosis with parvovirus B19 infection: Comment on the concise communication by Nikkari et al. Arthritis Rheum38 :1174– 1175,1995
                                                        150. Chou TN, Hsu TC, Chen RM, Lin LI, Tsay GJ: Parvovirus B19 infection associated with the production of anti-neutrophil cytoplasmic antibody (ANCA) and anticardiolipin antibody (aCL). Lupus9 :551– 554,2000
                                                        151. Hermann J, Demel U, Stunzner D, Daghofer E, Tilz G, Graninger W: Clinical interpretation of antineutrophil cytoplasmic antibodies: Parvovirus B19 infection as a pitfall. Ann Rheum Dis64 :641– 643,2005
                                                        152. Nikkari S, Vainionpaa R, Toivanen P, Gross WL, Mistry N, Csernok E, Szpirt W, Baslund B, Wiik A: Association of Wegener granulomatosis with parvovirus b19 infection: Comment—Reply. Arthritis Rheum38 :1175 ,1995
                                                        153. Eden A, Mahr A, Servant A, Radjef N, Amard S, Mouthon L, Garbarg-Chenon A, Guillevin L: Lack of association between B19 or V9 erythrovirus infection and ANCA-positive vasculitides: A case-control study. Rheumatology (Oxford)42 :660– 664,2003
                                                        154. Sachetto Z, Costa SC, Andrade PD, Conde RA, Amstalden EM, Samara AM, Fernandes SR: No evidence of parvovirus B19 in tissue samples from patients with polyarteritis nodosa and microscopic polyangiitis as assessed by the polymerase chain reaction. Ann Rheum Dis65 :418– 420,2006
                                                        155. Trepo C, Guillevin L: Polyarteritis nodosa and extrahepatic manifestations of HBV infection: The case against autoimmune intervention in pathogenesis. J Autoimmun16 :269– 274,2001
                                                        156. Boki KA, Dafni U, Karpouzas GA, Papasteriades C, Drosos AA, Moutsopoulos HM: Necrotizing vasculitis in Greece: Clinical, immunological and immunogenetic aspects—A study of 66 patients. Br J Rheumatol36 :1059– 1066,1997
                                                        157. Hauschild S, Csernok E, Schmitt WH, Gross WL: Antineutrophil cytoplasmic antibodies in systemic polyarteritis nodosa with and without hepatitis B virus infection and Churg-Strauss syndrome: 62 patients. J Rheumatol21 :1173– 1174,1994
                                                        158. Wu YY, Hsu TC, Chen TY, Liu TC, Liu GY, Lee YJ, Tsay GJ: Proteinase 3 and dihydrolipoamide dehydrogenase (E3) are major autoantigens in hepatitis C virus (HCV) infection. Clin Exp Immunol128 :347– 352,2002
                                                        159. Lamprecht P, Gutzeit O, Csernok E, Gause A, Longombardo G, Zignego AL, Gross WL, Ferri C: Prevalence of ANCA in mixed cryoglobulinemia and chronic hepatitis C virus infection. Clin Exp Rheumatol21 :S89– S94,2003
                                                        160. Davies DJ, Moran JE, Niall JF, Ryan GB: Segmental necrotising glomerulonephritis with antineutrophil antibody: Possible arbovirus aetiology? BMJ (Clin Res Ed)285 :606 ,1982
                                                        161. Savige JA, Chang L, Crowe SM: Anti-neutrophil cytoplasm antibodies in HIV infection. Adv Exp Med Biol336 :349– 352,1993
                                                        162. Sakkas L, Kistis C, Akritidis N: Antineutrophil cytoplasmic autoantibodies in human immunodeficiency virus infection. Am J Kidney Dis24 :731 ,1994
                                                          163. Cornely OA, Hauschild S, Weise C, Csernok E, Gross WL, Salzberger B, Fatkenheuer G, Diehl V, Schrappe M: Seroprevalence and disease association of antineutrophil cytoplasmic autoantibodies and antigens in HIV infection. Infection27 :92– 96,1999
                                                            164. Bonnet F, Pineau JJ, Taupin JL, Feyler A, Bonarek M, de Witte S, Bernard N, Lacoste D, Morlat P, Beylot J: Prevalence of cryoglobulinemia and serological markers of autoimmunity in human immunodeficiency virus infected individuals: A cross-sectional study of 97 patients. J Rheumatol30 :2005– 2010,2003
                                                              165. Schultz H, Csernok E, Herlyn K, Reichel PH, Moosig F, Cornely OA, Fagerhol MK, Gross WL: ANCA against bactericidal/permeability-increasing protein, azurocidin, calprotectin and defensins in rheumatic and infectious diseases: Prevalence and clinical associations. Clin Exp Rheumatol21 :S117– S120,2003
                                                              166. Staud R, Ramos LG: Influenza A-associated bronchiolitis obliterans organizing pneumonia mimicking Wegener's granulomatosis. Rheumatol Int20 :125– 128,2001
                                                              167. Hoffman GS, Sechler JM, Gallin JI, Shelhamer JH, Suffredini A, Ognibene FP, Baltaro RJ, Fleisher TA, Leavitt RY, Travis WD: Bronchoalveolar lavage analysis in Wegener's granulomatosis: A method to study disease pathogenesis. Am Rev Respir Dis143 :401– 407,1991
                                                              168. Muniain MA, Moreno JC, Gonzalez CR: Wegener's granulomatosis in two sisters. Ann Rheum Dis45 :417– 421,1986
                                                              169. Hay EM, Beaman M, Ralston AJ, Ackrill P, Bernstein RM, Holt PJ: Wegener's granulomatosis occurring in siblings. Br J Rheumatol30 :144– 145,1991
                                                                170. Franssen CF, ter Maaten JC, Hoorntje SJ: Brother and sister with myeloperoxidase associated autoimmune disease. Ann Rheum Dis53 :213 ,1994
                                                                  171. Nowack R, Lehmann H, Flores-Suarez LF, Nanhou A, van der Woude FJ: Familial occurrence of systemic vasculitis and rapidly progressive glomerulonephritis. Am J Kidney Dis34 :364– 373,1999
                                                                    172. Hull CM, Couser WG, Knostman JD: A familial case of P-ANCA glomerulonephritis presenting in a father and daughter. Am J Kidney Dis35 :E23 ,2000
                                                                      173. Brener Z, Cohen L, Goldberg SJ, Kaufman AM: ANCA-associated vasculitis in Greek siblings with chronic exposure to silica. Am J Kidney Dis38 :E28 ,2001
                                                                      174. Esnault VL, Testa A, Audrain M, Roge C, Hamidou M, Barrier JH, Sesboue R, Martin JP, Lesavre P: Alpha 1-antitrypsin genetic polymorphism in ANCA-positive systemic vasculitis. Kidney Int43 :1329– 1332,1993
                                                                      175. Hagen EC, Stegeman CA, D'Amaro J, Schreuder GM, Lems SP, Tervaert JW, de Jong GM, Hene RJ, Kallenberg CG, Daha MR: Decreased frequency of HLA-DR13DR6 in Wegener's granulomatosis. Kidney Int48 :801– 805,1995
                                                                      176. Gencik M, Borgmann S, Zahn R, Albert E, Sitter T, Epplen JT, Fricke H: Immunogenetic risk factors for anti-neutrophil cytoplasmic antibody (ANCA)-associated systemic vasculitis. Clin Exp Immunol117 :412– 417,1999
                                                                        177. Papiha SS, Murty GE, Ad'Hia A, Mains BT, Venning M: Association of Wegener's granulomatosis with HLA antigens and other genetic markers. Ann Rheum Dis51 :246– 248,1992
                                                                          178. Katz P, Alling DW, Haynes BF, Fauci AS: Association of Wegener's granulomatosis with HLA-B8. Clin Immunol Immunopathol14 :268– 270,1979
                                                                            179. Cotch MF, Fauci AS, Hoffman GS: HLA typing in patients with Wegener granulomatosis. Ann Intern Med122 :635 ,1995
                                                                              180. Elkon KB, Sutherland DC, Rees AJ, Hughes GR, Batchelor JR: HLA antigen frequencies in systemic vasculitis: Increase in HLA-DR2 in Wegener's granulomatosis. Arthritis Rheum26 :102– 105,1983
                                                                                181. Jagiello P, Gencik M, Arning L, Wieczorek S, Kunstmann E, Csernok E, Gross WL, Epplen JT: New genomic region for Wegener's granulomatosis as revealed by an extended association screen with 202 apoptosis-related genes. Hum Genet114 :468– 477,2004
                                                                                  182. Tsuchiya N, Kobayashi S, Hashimoto H, Ozaki S, Tokunaga K: Association of HLA-DRB1*0901-DQB1*0303 haplotype with microscopic polyangiitis in Japanese. Genes Immun7 :81– 84,2006
                                                                                  183. Spencer SJ, Burns A, Gaskin G, Pusey CD, Rees AJ: HLA class II specificities in vasculitis with antibodies to neutrophil cytoplasmic antigens. Kidney Int41 :1059– 1063,1992
                                                                                  184. Dijstelbloem HM, Scheepers RH, Oost WW, Stegeman CA, van der Pol WL, Sluiter WJ, Kallenberg CG, van de Winkel JG, Tervaert JW: Fcgamma receptor polymorphisms in Wegener's granulomatosis: Risk factors for disease relapse. Arthritis Rheum42 :1823– 1827,1999
                                                                                  185. Persson U, Truedsson L, Westman KW, Segelmark M: C3 and C4 allotypes in anti-neutrophil cytoplasmic autoantibody (ANCA)-positive vasculitis. Clin Exp Immunol116 :379– 382,1999
                                                                                  186. Gencik M, Meller S, Borgmann S, Sitter T, Menezes Saecker AM, Fricke H, Epplen JT: The association of CD18 alleles with anti-myeloperoxidase subtypes of ANCA-associated systemic vasculitides. Clin Immunol94 :9– 12,2000
                                                                                  187. Meller S, Jagiello P, Borgmann S, Fricke H, Epplen JT, Gencik M: Novel SNPs in the CD18 gene validate the association with MPO-ANCA+ vasculitis. Genes Immun2 :269– 272,2001
                                                                                  188. Huang D, Giscombe R, Zhou Y, Lefvert AK: Polymorphisms in CTLA-4 but not tumor necrosis factor-alpha or interleukin 1beta genes are associated with Wegener's granulomatosis. J Rheumatol27 :397– 401,2000
                                                                                  189. Segelmark M, Persson U, Westman KWA, Sturfelt G, Truedsson L: Polymorphisms of candidate genes in ANCA positive vasculitis. Cleve Clin J Med69[Suppl 2] ,SII155– SII156,2002
                                                                                    190. Cohen Tervaert JW, Slot MC, Sokolowska M, Savelkouls K, Janssen R, Damoiseaux J: Immunoregulatory gene polymorphisms in ANCA-associated vasculitis. Kidney Blood Press Res28 :176 ,2005
                                                                                    191. Zhou Y, Huang D, Paris PL, Sauter CS, Prock KA, Hoffman GS: An analysis of CTLA-4 and proinflammatory cytokine genes in Wegener's granulomatosis. Arthritis Rheum50 :2645– 2650,2004
                                                                                    192. Giscombe R, Wang X, Huang D, Lefvert AK: Coding sequence 1 and promoter single nucleotide polymorphisms in the CTLA-4 gene in Wegener's granulomatosis. J Rheumatol29 :950– 953,2002
                                                                                    193. Bartfai Z, Gaede KI, Russell KA, Murakozy G, Muller-Quernheim J, Specks U: Different gender-associated genotype risks of Wegener's granulomatosis and microscopic polyangiitis. Clin Immunol109 :330– 337,2003
                                                                                    194. Spriewald BM, Witzke O, Wassmuth R, Wenzel RR, Arnold ML, Philipp T, Kalden JR: 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 Dis64 :457– 461,2005
                                                                                    195. Murakozy G, Gaede KI, Ruprecht B, Gutzeit O, Schurmann M, Schnabel A, Schlaak M, Gross WL, Muller-Quernheim J: Gene polymorphisms of immunoregulatory cytokines and angiotensin-converting enzyme in Wegener's granulomatosis. J Mol Med79 :665– 670,2001
                                                                                      196. Csernok E, Szymkowiak CH, Mistry N, Daha MR, Gross WL, Kekow J: Transforming growth factor-beta (TGF-beta) expression and interaction with proteinase 3 (PR3) in anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis. Clin Exp Immunol105 :104– 111,1996
                                                                                      197. Borgmann S, Endisch G, Hacker UT, Song BS, Fricke H: Proinflammatory genotype of interleukin-1 and interleukin-1 receptor antagonist is associated with ESRD in proteinase 3-ANCA vasculitis patients. Am J Kidney Dis41 :933– 942,2003
                                                                                      198. Segelmark M, Elzouki AN, Wieslander J, Eriksson S: The PiZ gene of alpha 1-antitrypsin as a determinant of outcome in PR3-ANCA-positive vasculitis. Kidney Int48 :844– 850,1995
                                                                                      199. Gencik M, Meller S, Borgmann S, Fricke H: Proteinase 3 gene polymorphisms and Wegener's granulomatosis. Kidney Int58 :2473– 2477,2000
                                                                                      200. Reynolds WF, Stegeman CA, Tervaert JW: −463 G/A myeloperoxidase promoter polymorphism is associated with clinical manifestations and the course of disease in MPO-ANCA-associated vasculitis. Clin Immunol103 :154– 160,2002
                                                                                      201. Alcorta D, Preston G, Munger W, Sullivan P, Yang JJ, Waga I, Jennette JC, Falk R: Microarray studies of gene expression in circulating leukocytes in kidney diseases. Exp Nephrol10 :139– 149,2002
                                                                                      202. Witko-Sarsat V, Lesavre P, Lopez S, Bessou G, Hieblot C, Prum B, Noel LH, Guillevin L, Ravaud P, Sermet-Gaudelus I, Timsit J, Grunfeld JP, Halbwachs-Mecarelli L: A large subset of neutrophils expressing membrane proteinase 3 is a risk factor for vasculitis and rheumatoid arthritis. J Am Soc Nephrol10 :1224– 1233,1999
                                                                                      203. Muller Kobold AC, Kallenberg CG, Tervaert JW: Leucocyte membrane expression of proteinase 3 correlates with disease activity in patients with Wegener's granulomatosis. Br J Rheumatol37 :901– 907,1998
                                                                                      204. Schreiber A, Busjahn A, Luft FC, Kettritz R: Membrane expression of proteinase 3 is genetically determined. J Am Soc Nephrol14 :68– 75,2003
                                                                                      205. van Rossum AP, Rarok AA, Huitema MG, Fassina G, Limburg PC, Kallenberg CG: Constitutive membrane expression of proteinase 3 (PR3) and neutrophil activation by anti-PR3 antibodies. J Leukoc Biol76 :1162– 1170,2004
                                                                                      206. Rarok AA, Stegeman CA, Limburg PC, Kallenberg CG: Neutrophil membrane expression of proteinase 3 (PR3) is related to relapse in PR3-ANCA-associated vasculitis. J Am Soc Nephrol13 :2232– 2238,2002
                                                                                      207. Gunton JE, Stiel J, Clifton-Bligh P, Wilmshurst E, McElduff A: Prevalence of positive anti-neutrophil cytoplasmic antibody (ANCA) in patients receiving anti-thyroid medication. Eur J Endocrinol142 :587 ,2000
                                                                                      208. D'Cruz D, Chesser AM, Lightowler C, Comer M, Hurst MJ, Baker LR, Raine AE: Antineutrophil cytoplasmic antibody-positive crescentic glomerulonephritis associated with anti-thyroid drug treatment. Br J Rheumatol34 :1090– 1091,1995
                                                                                        209. Yuasa S, Hashimoto M, Yura T, Sumikura T, Takahashi N, Shoji T, Uchida K, Fujioka H, Kihara M, Matsuo H: Antineutrophil cytoplasmic antibodies (ANCA)-associated crescentic glomerulonephritis and propylthiouracil therapy. Nephron73 :701– 703,1996
                                                                                        210. Gao Y, Chen M, Ye H, Guo XH, Zhao MH, Wang HY: The target antigens of antineutrophil cytoplasmic antibodies (ANCA) induced by propylthiouracil. Int Immunopharmacol7 :55– 60,2007
                                                                                        211. Slot MC, Links TP, Stegeman CA, Tervaert JW: Occurrence of antineutrophil cytoplasmic antibodies and associated vasculitis in patients with hyperthyroidism treated with antithyroid drugs: A long-term followup study. Arthritis Rheum53 :108– 113,2005
                                                                                        212. Gao Y, Zhao MH, Guo XH, Xin G, Gao Y, Wang HY: The prevalence and target antigens of antithyroid drugs induced antineutrophil cytoplasmic antibodies (ANCA) in Chinese patients with hyperthyroidism. Endocr Res30 :205– 213,2004
                                                                                        213. Pillinger M, Staud R: Wegener's granulomatosis in a patient receiving propylthiouracil for Graves’ disease. Semin Arthritis Rheum28 :124– 129,1998
                                                                                        214. Sato H, Hattori M, Fujieda M, Sugihara S, Inomata H, Hoshi M, Miyamoto S: High prevalence of antineutrophil cytoplasmic antibody positivity in childhood onset Graves’ disease treated with propylthiouracil. J Clin Endocrinol Metab85 :4270– 4273,2000
                                                                                        215. Sera N, Ashizawa K, Ando T, Abe Y, Ide A, Usa T, Tominaga T, Ejima E, Yokoyama N, Eguchi K: Treatment with propylthiouracil is associated with appearance of antineutrophil cytoplasmic antibodies in some patients with Graves’ disease. Thyroid10 :595– 599,2000
                                                                                          216. Noh JY, Asari T, Hamada N, Makino F, Ishikawa N, Abe Y, Ito K, Ito K: Frequency of appearance of myeloperoxidase-antineutrophil cytoplasmic antibody (MPO-ANCA) in Graves’ disease patients treated with propylthiouracil and the relationship between MPO-ANCA and clinical manifestations. Clin Endocrinol (Oxf)54 :651– 654,2001
                                                                                          217. Wada N, Mukai M, Kohno M, Notoya A, Ito T, Yoshioka N: Prevalence of serum anti-myeloperoxidase antineutrophil cytoplasmic antibodies (MPO-ANCA) in patients with Graves' disease treated with propylthiouracil and thiamazole. Endocr J49 :329– 334,2002
                                                                                          218. Choi HK, Merkel PA, Tervaert JW, Black RM, Mccluskey RT, Niles JL: Alternating antineutrophil cytoplasmic antibody specificity: Drug-induced vasculitis in a patient with Wegener's granulomatosis. Arthritis Rheum42 :384– 388,1999
                                                                                          219. Walfish PG, Tseng KH: Intrathyroidal activated (Ia+) T-lymphocyte CD+ subsets and B cells in Graves’ hyperthyroidism respond rapidly to propylthiouracil therapy: Demonstration using fine needle aspirates and two-colour laser flow cytometry. Autoimmunity13 :35– 41,1992
                                                                                          220. Guo XH, Zhao MH, Gao Y, Wang SF, Gao Y: Antineutrophil cytoplasmic antibody associated vasculitis induced by antithyroid agents. Zhonghua Yi Xue Za Zhi83 :932– 935,2003
                                                                                          221. Xu X, Zhao M, Zhang Y, Guo X, Wang H: Clinicopathological characteristics of propylthiouracil-induced antineutrophil cytoplasmic antibodies-positive vasculitis and their target antigens: A report of 4 cases and literature review. Zhonghua Nei Ke Za Zhi41 :404– 407,2002
                                                                                          222. Elias AN, Goodman MM, Rohan MK: Serum ICAM-1 concentrations in patients with psoriasis treated with antithyroid thioureylenes. Clin Exp Dermatol18 :526– 529,1993
                                                                                          223. Elias AN, Goodman MM, Rohan MK: Effect of propylthiouracil and methimazole on serum levels of interleukin-2 receptors in patients with psoriasis. Int J Dermatol32 :537– 540,1993
                                                                                          224. Jiang X, Khursigara G, Rubin RL: Transformation of lupus-inducing drugs to cytotoxic products by activated neutrophils. Science266 :810– 813,1994
                                                                                          225. von Schmiedeberg S, Hanten U, Goebel C, Schuppe HC, Uetrecht J, Gleichmann E: T cells ignore the parent drug propylthiouracil but are sensitized to a reactive metabolite generated in vivo. Clin Immunol Immunopathol80 :162– 170,1996
                                                                                          226. Silberman DM, Wald M, Genaro AM: Effects of chronic mild stress on lymphocyte proliferative response: Participation of serum thyroid hormones and corticosterone. Int Immunopharmacol2 :487– 497,2002
                                                                                          227. Nassberger L, Sjoholm AG, Thysell H: Antimyeloperoxidase antibodies in patients with extracapillary glomerulonephritis. Nephron56 :152– 156,1990
                                                                                          228. Short AK, Lockwood CM: Antigen specificity in hydralazine associated ANCA positive systemic vasculitis. QJM88 :775– 783,1995
                                                                                            229. Choi HK, Merkel PA, Walker AM, Niles JL: Drug-associated antineutrophil cytoplasmic antibody-positive vasculitis: Prevalence among patients with high titers of antimyeloperoxidase antibodies. Arthritis Rheum43 :405– 413,2000
                                                                                            230. Almroth G, Enestrom S, Hed J, Samuelsson I, Sjostrom P: Autoantibodies to leucocyte antigens in hydralazine-associated nephritis. J Intern Med231 :37– 42,1992
                                                                                            231. Cambridge G, Wallace H, Bernstein RM, Leaker B: Autoantibodies to myeloperoxidase in idiopathic and drug-induced systemic lupus erythematosus and vasculitis. Br J Rheumatol33 :109– 114,1994
                                                                                            232. Greenfield JR, McGrath M, Kossard S, Charlesworth JA, Campbell LV: ANCA-positive vasculitis induced by thioridazine: Confirmed by rechallenge. Br J Dermatol147 :1265– 1267,2002
                                                                                            233. Powell J, Grech H, Holder J: A boy with cutaneous necrosis occurring during treatment with levamisole. Clin Exp Dermatol27 :32– 33,2002
                                                                                              234. Merkel PA: Drugs associated with vasculitis. Curr Opin Rheumatol10 :45– 50,1998
                                                                                              235. Jones BF, Major GA: Crescentic glomerulonephritis in a patient taking penicillamine associated with antineutrophil cytoplasmic antibody. Clin Nephrol38 :293 ,1992
                                                                                                236. Noel LH: Antineutrophil cytoplasm antibodies (ANCA): Description and immunopathological role [in French]. Ann Med Interne (Paris)151 :178– 183,2000
                                                                                                237. Choi HK, Slot MC, Pan G, Weissbach CA, Niles JL, Merkel PA: Evaluation of antineutrophil cytoplasmic antibody seroconversion induced by minocycline, sulfasalazine, or penicillamine. Arthritis Rheum43 :2488– 2492,2000
                                                                                                238. Cuadrado MJ, D'Cruz D, Lloyd M, Mujic F, Khamashta MA, Hughes GR: Allergic disorders in systemic vasculitis: A case-controlled study. Br J Rheumatol33 :749– 753,1994
                                                                                                Copyright © 2008 The Authors. Published by Wolters Kluwer Health, Inc. All rights reserved.