Quick viewing(Text Mode)

(MPO) and Proteinase 3 (PR3) in Patients with Systemic Vasculitis

(MPO) and Proteinase 3 (PR3) in Patients with Systemic Vasculitis

Clin Exp Immunol 1996; 103:253–258

T cell responses to myeloperoxidase (MPO) and (PR3) in patients with systemic

M. E. GRIFFITH, A. COULTHART & C. D. PUSEY Renal Unit, Department of Medicine, Royal Postgraduate Medical School, Hammersmith Hospital, London, UK

(Accepted for publication 26 October 1995)

SUMMARY T cell-mediated immune responses are likely to be important in the pathogenesis of systemic vasculitis. However, identifying the T cells involved has proved difficult, and there are conflicting reports regarding T cell proliferation in response to different autoantigens. Perinuclear (P) and cytoplasmic (C) anti- cytoplasmic (ANCA) are closely associated with systemic vasculitis, and are generally specific for MPO or PR3, respectively. We studied the proliferative responses to MPO and PR3 of peripheral blood mononuclear cells from patients with P- ANCA or C-ANCA specific for these antigens by ELISA. These responses were compared with those of normal controls, and of disease controls with P- or C-ANCA not specific for MPO or PR3.

The patient group as a whole showed significant T cell proliferation in response to the autoantigens

X

compared with controls (P ˆ 0 005). Cells from nine of 13 P-ANCA-positive, anti-MPO-positive

X patients proliferated in response to MPO, compared with five of 16 controls (P ˆ 0 04). Cells from

five of eight C-ANCA-positive, anti-PR3-positive patients proliferated in response to PR3, compared

X with two of 11 controls (P ˆ 0 05). These experiments demonstrate that patients with P-ANCA or C- ANCA possess T cells which respond to MPO or PR3, respectively. As in other autoimmune diseases, responses to both antigens were also seen in a proportion of healthy controls. Further analysis of these responses will be important in understanding the pathogenesis of systemic vasculitis and in designing specific immunotherapy.

Keywords T cells systemic vasculitis proteinase 3 myeloperoxidase anti-neutrophil cytoplasmic antibodies

INTRODUCTION WG, although they are also found in MP [2]. The major target antigen of C-ANCA is PR3 [4,5], a neutrophil (MP) and Wegener’s granulomatosis with bacteriocidal properties, which is identical to myeloblastin, (WG) are systemic vasculitides characterized by pauci-immune a leucocyte growth factor [6]. focal segmental and necrotizing glomerulonephritis, together The presence and titre of ANCA have been shown to with involvement of a variety of other organs. There is correlate with disease extent and activity, and a rise in titre increasing evidence that an autoimmune process underlies generally predicts relapse [7]. In vitro, ANCA are able to induce the development of systemic vasculitis. Two types of anti- neutrophil degranulation [8], and may both enhance neutrophil neutrophil cytoplasmic (ANCA) are seen in these cytotoxicity and also have direct cytotoxic effects on endo- disorders, perinuclear (P)-ANCA and cytoplasmic (C)-ANCA, thelial cells [9,10]. T cell involvement in the disease process as defined by their appearance on indirect immunofluorescence is likely, not only in providing help for autoantibody production, of normal human [1]. P-ANCA are found in a wider but also by direct cytotoxic mechanisms. Pathological lesions spectrum of disorders than C-ANCA, and have a greater variety

in WG are characterized by granuloma formation in the of target autoantigens [2]. When P-ANCA are present in MP, the ‡

lungs, upper respiratory tract and elsewhere, and both CD4 target autoantigen is usually MPO [3], a neutrophil granule ‡ and CD8 T cells have been identified in the glomeruli and enzyme involved in host defence. C-ANCA predominate in interstitium in pauci-immune rapidly progressive glomerulone- phritis (RPGN) [11]. Attempts to identify the T cells involved Correspondence: Dr M. E. Griffith, Renal Unit, Royal Postgraduate have been hindered by difficulties in obtaining sufficiently Medical School, Hammersmith Hospital, Du Cane Road, London W12 pure antigen preparations, and previous studies have given

0NN, UK. conflicting results [12–15]. 5 1996 Blackwell Science 253 254 M. E. Griffith, A. Coulthart & C. D. Pusey

Table 1. Diagnoses of patients with anti-neutrophil cytoplasmic antibodies

(ANCA) specific for MPO or PR3

‡

P-ANCA‡ C-ANCA

‡ Diagnosis MPO‡ PR3

Microscopic polyangiitis 7 2 Wegener’s granulomatosis 0 6 Idiopathic rapidly progressive nephritis 3 0 Goodpasture’s disease 3 0

In this study, we examined responses of peripheral blood mononuclear cells (PBMC) to purified preparations of MPO or Fig. 1. A. SDS–PAGE of the PR3-containing fraction from a high PR3 in patients with P-ANCA or C-ANCA specific for these performance liquid chromatography (HPLC) separation, indicating its antigens purity. B. Western blot with anti-PR3 serum from a patient with Wegener’s granulomatosis (WG), demonstrating antigenicity.

PATIENTS AND METHODS Patients Preparation of antigens Blood or buffy coat (collected during plasma exchange) was Purified MPO was purchased from Calbiochem (Nottingham), obtained from 21 patients, 13 of whom had P-ANCA specific for UK) and its purity checked by SDS–PAGE. PR3 was purified MPO, and eight of whom had C-ANCA specific for using the method described by Gaskin et al. [18]. Briefly, whole PR3. Eighteen of these patients had systemic vasculitis, with blood was mixed with 3% dextran and the erythrocytes allowed diagnoses made according to criteria defined by the Chapel to sediment. The supernatant was resuspended on Lymphoprep Hill consensus conference (Table 1) [16]. Three patients with (Nycomed) and the lymphocytes were separated by density P-ANCA also had Goodpasture’s disease, with anti-glomerular gradient centrifugation. The resulting pellet was washed with basement membrane (anti-GBM) antibodies as detected by distilled water to remove any remaining erythrocytes by

hypotonic lysis, and the neutrophils then suspended in relaxa- ELISA specific for 3(IV)NCI [17]. Seven other vasculitis patients were selected as controls (Table 2), four with P- tion buffer before nitrogen bomb cavitation on ice. The alpha ANCA and three with C-ANCA which were not specific granules were then separated on a Percoll (Sigma Aldrich and for MPO or PR3. Cells from 12 normal controls were tested Co., Poole, UK) gradient, and disrupted by multiple freeze–thaw against MPO, and from eight against PR3. cycles followed by sonication. The granule contents were sepa- rated on a C4 reverse phase high performance liquid chromato- graphy (HPLC) column (Alltech, Carnforth, UK), with an Isolation of PBMC acetonitrile gradient. The resulting fractions were freeze-dried, The PBMC were separated from blood or buffy coat by and those containing PR3 were identified by SDS–PAGE and density gradient centrifugation to Lymphoprep (Nycomed, Bir- Western blotting against sera containing C-ANCA specific for mingham, UK). Cells were resuspended in freezing PR3 by ELISA (Fig. 1). The fractions selected were confirmed to be PR3 by N-terminal amino acid sequencing. The PR3 was medium, consisting of 40% RPMI 1640 (GIBCO, Paisley, UK), with 10% dimethylsulphoxide (BDH, Dagenham, UK) and resuspended in PBS and the concentration of protein estimated by 50% fetal calf serum (FCS; M. B. Meldrum, Bourne End, UK), comparison of optical density with bovine serum albumin (BSA) and stored in liquid nitrogen. Before use, the cells were rapidly solutions of known concentration. thawed, washed in PBS pH 7.2, and their viability checked by with ethidium bromide and acridine orange. Assays for ANCA The method used for detection of ANCA by indirect immuno- fluorescence was based on that described by Wiik [19,20]. Table 2. Diagnoses of controls with anti-neutrophil cytoplasmic antibodies The ELISAs used for measurement of antibodies to MPO, (ANCA) not specific for MPO or PR3 PR3, and GBM were performed as previously described by our laboratory [17,18,21]. The antigens used in the MPO and

PR3 ELISAs were identical to those used in the prolifera- ‡

P-ANCA‡ C-ANCA tion assays.

ÿ Diagnosis MPOÿ PR3

Microscopic polyangiitis 1 0 T cell proliferation assays

5

Wegener’s granulomatosis 0 3 The isolated PBMC were cultured at 2  10 cells/well in flat-

Idiopathic rapidly progressive nephritis 1 0 bottomed 96-well plates (GIBCO), suspended in 200 "l of RPMI Cryoglobulinaemia 1 0 1640 medium (GIBCO) containing 10% AB‡ human serum (North

Scleroderma 1 0 West London Transfusion Centre, Edgware, UK), 100 U/ml

penicillin, 100 "g/ml streptomycin (GIBCO) and 4 mM glutamine 5 1996 Blackwell Science Ltd, Clinical and Experimental Immunology, 103:253–258 T cell responses in vasculitis 255

Fig. 2. Proliferative responses of peripheral blood mononuclear cells (PBMC) from patients and controls to MPO and PR3. g, Controls with P-ANCA or C-ANCA not specific for MPO or PR3, respectively.

Fig. 3. Proliferation of peripheral blood mononuclear cells (PBMC) from

h patients and controls to MPO is dose-dependent. g, Patient; , control. Error bars show  s.e.m. (GIBCO). All assays were performed in triplicate, and included

control wells with no antigen and wells containing phytohae- magglutinin (PHA; Sigma Aldrich) at 2 "g/ml, as a non-specific T cell mitogen. Specific antigen was added to the wells in between those who responded to the specific autoantigens and

.

" concentrations varying from 0 1 "g/ml to 30 g/ml. Cells from those who did not.

patients with MPO-specific ANCA were tested against MPO, and The optimum autoantigen concentration was 10 "g/ml for those with PR3-specific ANCA were tested against PR3. The MPO, and 20 "g/ml for PR3. PBMC from patients and controls three patients with P-ANCA not specific for MPO were also that responded to the antigens did so in a dose-dependent tested against MPO, and those with C-ANCA not specific for manner (Fig. 3). Proliferation was seen at all three time points, PR3 were tested against PR3. Twelve normal controls were and was usually maximal at 7 or 10 days (Fig. 4). All batches of tested against MPO, and eight against PR3. Proliferative cells tested were viable, as shown by their response to PHA responses were measured at 5, 7 and 10 days. Tritiated thymi- (mean SI 78). There was no difference in the response to PHA

dine (Amersham, Little Chalfont, UK) was added to the wells at between those who responded to MPO or PR3 and those who did 1 "Ci/well, and 16 h later the cells were harvested onto glassfibre not. filters using an automated cell harvester (Pharmacia, Milton Cells from nine of 13 P-ANCA-positive, anti-MPO-positive Keynes, UK). Thymidine incorporation was measured using an patients proliferated in response to MPO, with a median SI of

. automated  counter (Pharmacia). Results were expressed as 4 7, compared with five of 16 controls (five of 12 normal

either ct/min or as stimulation indices (SI: calculated as the ratio controls and nil of four ANCA-positive, anti-MPO-negative

. X of ct/min with antigen to ct/min with medium alone). An SI of patients), with a median SI of 1 7(Pˆ004). The highest SI 2.5 or more was regarded as positive. in the ANCA-positive, anti-MPO-positive group was 21.3, whereas that of controls was 9.5. Eight of the nine anti-MPO- positive patients who responded had cells collected during their Statistical analysis first presentation with active vasculitis, and all had started The Wilcoxon rank sum test was used to test for significant therapy with prednisolone and cyclophosphamide within the differences in proliferation between the study groups and previous 72 h. Patient 9 (SI 7.1) was clinically in remission controls. Analysis of variance was used to test for differences when the cells were taken and not on any treatment, although in responses to PHA, and background proliferation counts, she remained P-ANCA-positive and has since relapsed. Of the between those who responded to MPO or PR3 and those who four anti-MPO-positive patients who did not respond, three were did not. also positive for anti-GBM antibodies, and clinically were diagnosed as having Goodpasture’s disease. Cells from patients with Goodpasture’s were collected during their first presentation RESULTS with active disease, and all had started prednisolone and cyclo- T cell proliferation was observed to both MPO and PR3 in a phosphamide within the previous 72 h. The fourth non-responder proportion of patients and controls (Fig. 2). Overall, cells from had MP that was clinically in remission, and was on low dose 14 of 21 patients proliferated in response to their specific prednisolone.

autoantigen (median SI 4.63), which was significantly greater Cells from five of eight C-ANCA-positive, anti-PR3-positive

. X than the seven of 27 controls (median SI 1 7, P ˆ 0 005). The patients showed significant proliferation in response to PR3, the

mean background proliferation was 257 ct/min, and did not differ median SI being 3.8, compared with two of 11 controls (two of 5 1996 Blackwell Science Ltd, Clinical and Experimental Immunology, 103:253–258 256 M. E. Griffith, A. Coulthart & C. D. Pusey

is also increasing evidence for the pathogenicity of ANCA [27], the production of which is likely to depend on T cell help. Furthermore, success in obtaining remission in patients with intractable vasculitis has been achieved by treatment with a combination of MoAbs to CD4 and CD52 [28]. T cell proliferation in response to specific B cell autoantigens has been demonstrated in other autoimmune diseases, such as myasthenia gravis [29], autoimmune thyroiditis [30,31], and Goodpasture’s disease [32]. We have now demonstrated sig- nificant T cell proliferation to MPO and PR3 in patients with ANCA specific for these antigens. Proliferation was also seen in cells from some healthy controls, a phenomenon observed in several other autoimune diseases [32–34]. Further analysis is required to ascertain whether these autoreactive T cells are present at a lower precursor frequency in controls than in patients, as shown in myasthenia gravis and multiple sclerosis [33,34]. Previous studies of T cell responses to MPO and PR3 in patients with vasculitis have been hampered by difficulties with antigen preparation, and results have been inconsistent. An early study failed to demonstrate proliferation to a crude acid

Fig. 4. Proliferation of peripheral blood mononuclear cells (PBMC) from extract of neutrophil granules, which was not separated into

h l

one patient to PR3 at three different time points: g, day 5; , day 7; , day individual components [12]. Convincing T cell proliferation has 10. Error bars show  s.e.m. been observed in response to purified PR3, but not to MPO. Using boiled inactivated , Brouwer et al. demonstrated significant proliferation to PR3, but obtained a response to

eight normal controls and nil of three C-ANCA-positive, anti- MPO in only one of five patients [14]. More recently, Ballieux

. X

PR3-negative controls), with a median SI of 1 3(Pˆ005). Of et al. [15] demonstrated proliferation to native PR3, but only at

X the five anti-PR3-positive patients who responded, three had a low level (maximum SI `3 5); however, their method of cells collected during their first presentation with active vascu- purification involved detergents which, they suggest, may litis, and had started treatment with prednisolone and cyclopho- have disrupted intra- and intermolecular bonds of PR3 and sphamide within the previous 72 h. Two patients were clinically resulted in alteration of antigen recognition and processing. in remission; one was taking prednisolone and azathioprine (SI Some neutrophil enzymes may be mitogenic and stimulate 4.6), and the other was not on treatment, but has always lymphocytes non-specifically [35]. However, as reported by remained C-ANCA-positive (SI 3.0). The three anti-PR3-positive the other group using native purified antigens [15], we did patients who did not respond also had cells collected during not observe this in our experiments. their first presentation with active vasculitis. Two had started All patients in the present study had their ANCA specificity prednisolone and cyclophosphamide in the previous 72 h, and determined by ELISA to ensure that the correct B cell auto- one had cells taken before starting any treatment (SI 1.7). The antigen was being investigated. Controls with ANCA not specific highest SI obtained in the C-ANCA positive, anti-PR3-positive for MPO or PR3 were also tested, to ensure that ANCA group was 18.6, whereas that of controls was 3.7. positivity itself was not affecting the results. Many (13/21) of Four individuals that were retested showed consistent our patients were undergoing plasma exchange at the time of responses. (e.g. maximum SIs were 9.2 and 8.4 in one patient, obtaining their PBMC, and may have had earlier or more severe and 2.8 and 3.1 in another). In view of the possible effects disease than patients in other studies; the higher proliferative of enzymatically active MPO and PR3 on T cell proliferation, responses obtained may therefore reflect an increased population three subjects were also tested with MPO or PR3 which was of autoreactive cells. boiled for 15 min to remove enzymatic activity [14]; the The isolation of T cell lines and clones in other autoim- responses obtained were the same as those with unheated mune diseases has been facilitated by the use of recombinant antigen. antigens [33]. The production of recombinant MPO has already been reported [36], and production of an antigenic form of recombinant PR3 is currently underway in our laboratory. These reagents should allow us to establish autoreactive lines and DISCUSSION clones from proliferating T cell populations, which will The role of T cells in many other autoimmune diseases is allow further investigation of the specificity and characteristics now well established [22], and evidence for their involve- of autoreactive T cells in systemic vasculitis. The character- ment in systemic vasculitis is increasing. The level of soluble ization of T cell responses at a molecular level has led to the IL-2 receptor, which is a marker of T cell activation, has been development of new immunotherapeutic approaches, for found to rise at times of active vasculitis [23,24]. More example, the use of tolerogenic peptide analogues based on indirect evidence for T cell involvement comes from reports key autoantigenic peptides, or immunization with peptides of associations between HLA class II and systemic based on the sequence of the relevant T cell receptors [37].

vasculitis, although there is as yet no consensus [25,26]. There These approaches have been shown to prevent or treat 5 1996 Blackwell Science Ltd, Clinical and Experimental Immunology, 103:253–258 T cell responses in vasculitis 257 autoimmune disease in a variety of animal models, and trials A hypothesis based on both humoral and cellular mechanisms. Neth in some human diseases are underway [38–40]. Systemic J Med 1990; 36:169–71. vasculitis is a relapsing disease with considerable morbidity 14 Brouwer E, Stegeman CA, Huitema MG, Limburg PC, Kallenberg and mortality, which is contributed to by the adverse effects CGM. T cell reactivity to proteinase 3 and myeloperoxidase in of non-specific immunosuppressive therapy required for dis- patients with Wegener’s granulomatosis (WG). Clin Exp Immunol 1994; 98:448–53. ease control. The development of specific immunotherapy 15 Ballieux BEPB, van der Burg SH, Hagen EC, van der Woude FJ, directed at autoreactive T cells in systemic vasculitis is there- Melief CJM, Daha MR. Cell mediated autoimmunity in patients fore of great potential importance. with Wegener’s granulomatosis (WG). Clin Exp Immunol 1995; 100:186–93. 16 Jennette JC, Falk RJ, Andrassy K et al. Nomenclature of systemic vasculitides. Proposal of an international consensus conference. ACKNOWLEDMENTS Arthritis Rheum 1994; 37:187–92. 17 Katbamna I, Turner N, Ross CN, Pusey CD. Development and M.E.G. is a Wellcome Trust Research Training Fellow. We are grateful to application of an ELISA for Goodpasture’s disease based on sheep

Dr G. Gaskin for advice on the purification of PR3, and to Dr C. J. Derry 3(IV)NC1 domains. J Clin Lab Immunol 1995; (in press). for help in establishing the proliferation assays. We thank Ms J. S. Hollyer 18 Gaskin G, Kendal H, Coulthart A, Turner N, Pusey CD. Use of and Ms A. Gosling for statistical advice. proteinase 3 purified by reverse phase HPLC to detect auto- antibodies in systemic vasculitis. J Immunol Methods 1995; 180:25–33. 19 Wiik A. Delineation of a standard procedure for indirect immuno- REFERENCES fluorescence detection of anti-neutrophil cytoplasmic antibodies 1 Hagen EC, Ballieux BE, van Es LA, Daha MR, van der Woude FJ. (ANCA). APMIS 1989; 97(Suppl. 6):12–13. Antineutrophil cytoplasmic autoantibodies: a review of the antigens 20 Gaskin G, Savage COS, Ryan JJ et al. Anti-neutrophil cytoplasmic involved, the assays, and the clinical and possible pathogenetic antibodies and disease activity during long-term follow-up of 70 consequences. (Review.) Blood 1993; 81:1996–2002. patients with systemic vasculitis. Nephrol Dial Transplant 1991; 2 Pusey CD, Gaskin G. Disease associations with anti-neutrophil 6:689–94. cytoplasmic antibodies. In: Gross WL, ed. ANCA-associated 21 De’Oliviera J, Gaskin G, Dash A, Rees AJ, Pusey CD. Relationship vasculitides: immunological and clinical aspects. New York: between disease activity and ANCA concentration by ELISA in Plenum Press, 1993; 145–55. long-term management of systemic vasculitis. Am J Kid Dis 1995; 3 Falk RJ, Jennette JC. Anti-neutrophil cytoplasmic autoantibodies 25:380–9. with specificity for myeloperoxidase in patients with systemic 22 Wraith DC, McDevitt HO, Steinman L, Acha-Orbea H. T cell vasculitis and idiopathic necrotizing and crescentic glomerulone- recognition as the target for immune intervention in autoimmune phritis. N Engl J Med 1988; 318:1651–7. disease. Cell 1989; 57:709–15. 4 Goldschmeding R, van der Schoot CE, ten Bokkel Huinink D 23 Stegeman CA, Cohen Tervaert JW, Huitema MG, Kallenberg CGM. et al. Wegener’s granulomatosis autoantibodies identify a novel Serum markers of T cell activation in relapses of Wegener’s diisopropylfluorophosphate-binding protein in the of granulomatosis. Clin Exp Immunol 1993; 91:415–20. normal human neutrophils. J Clin Invest 1989; 84:1577–87. 24 Schmitt WH, Heeson C, Csernok E, Rautmann A, Gross WL. 5 Niles JL, McCluskey RT, Ahmad MF, Amin Arnaout M. Wegener’s Elevated serum levels of soluble interleukin-2 receptor in patients granulomatosis autoantigen is a novel neutrophil serine proteinase. with Wegener’s granulomatosis. Arthritis Rheum 1992; 35:1088–96. Blood 1989; 74:1888–93. 25 Spencer SJW, Burns A, Gaskin G, Pusey CD, Rees AJ. HLA class 6 Bories D, Raynal M-C, Solomon DH, Darzynkiewicz Z, Cayre YE. II specificities in vasculitis with antibodies to neutrophil cytoplasmic Down-regulation of a , myeloblastin, causes growth antigens. Kidney Int 1992; 41:1059–63. arrest and differentiation of promyelocytic leukaemia cells. Cell 26 Zhang L, Jayne D, Zhao MH, Lockwood CM, Oliviera DBG. 1989; 59:959–68. Distribution of MHC class II alleles in primary systemic vasculitis. 7 Cohen Tervaert JW, van der Woude FJ, Fauci AS et al. Association Kidney Int 1995; 47:294–8. between active Wegener’s granulomatosis and anticytoplasmic 27 Kallenberg CGM, Brouwer E, Mulder AHL, Stegeman CA, antibodies. Arch Intern Med 1989; 149:2461–5. Weening JJ, Cohen Tervaert JW. ANCA-pathophysiology revisited. 8 Falk RJ, Terrell RS, Charles LA, Jennette JC. Anti-neutrophil Clin Exp Immunol 1995; 100:1–3. cytoplasmic autoantibodies induce neutrophils to degranulate and 28 Lockwood CM, Stewart S, Thiru S, Isaacs JD, Hale G, Waldmann produce oxygen radicals in vitro. Proc Natl Acad Sci USA 1990; H. Humanised monoclonal antibody therapy for Wegener’s 87:4115–9. granulomatosis. Clin Exp Immunol 1995; 101:43 (Abstr.). 9 Savage COS, Pottinger BE, Gaskin G, Pusey CD, Pearson JD. 29 Willcox N. Myaesthenia gravis. Curr Opin Immunol 1993; 5:910–7. Autoantibodies developing to myeloperoxidase and proteinase 3 30 Tandon N, Weetman AP. T cells and thyroid autoimmunity. J Roy in systemic vasculitis stimulate neutrophil cytotoxicity towards Coll Phys 1994; 28:10–18. cultured endothelial cells. Am J Pathol 1992; 141:335–42. 31 McGregor AM. Autoimmunity in the thyroid—can the molecular 10 Mayet WJ, Schwarting A, Meyer zum Buschenfelde KH. Cytotoxic revolution contribute to our understanding? Quart J Med 1992; effect of antibodies to proteinase 3 (cANCA) on human endothelial 82:1–13. cells. Clin Exp Immunol 1994; 97:458–65. 32 Derry CJ, Ross CN, Lombardi G et al. Analysis of T cell responses 11 Bolton WK, Innes D, Sturgill BC, Kaiser DL. T cells and to the autoantigen in Goodpasture’s disease. Clin Exp Immunol macrophages in rapidly progressive glomerulonephritis: clinico- 1995; 100:202–8. pathological correlates. Kidney Int 1987; 32:869–76. 33 Sun J, Harcourt G, Wang ZY et al. T cell responses to human 12 Mathieson PW, Lockwood CM, Oliviera DGB. T and B cell recombinant acetylcholine receptor—a subunit in myaesthenia gravis responses to cytoplasmic antigens in systemic vasculitis. Clin and controls. Eur J of Immunol 1992; 22:1553–9. Immunol Immunopathol 1992; 63:135–41. 34 Olsson T, Sun J, Hillert J et al. Increased numbers of T cells 13 Van de Woude FJ, van Es LA, Daha MR. The role of the recognizing multiple myelin basic protein in multiple sclerosis. Eur

cANCA antigen in the pathogenesis of Wegener’s granulomatosis. J Immunol 1992; 22:1083–7. 5 1996 Blackwell Science Ltd, Clinical and Experimental Immunology, 103:253–258 258 M. E. Griffith, A. Coulthart & C. D. Pusey

35 Vischer TL, Bretz U, Baggiolini M. In vitro stimulation of 38 Kaufman DL, Clare-Salzler M, Tian J et al. Spontaneous loss of T- lymphocytes by neutral proteinases from human polymorphonuclear cell tolerance to glutamic acid decarboxylase in murine insulin- leucocyte granules. J Exp Med 1976; 144:863–72. dependent diabetes. Nature 1993; 366:69–72. 36 Pedrello E, Bleil L, Bautz FA, Kalden JR, Bautz E. ANCA 39 Elias D, Cohen IR. Peptide therapy for diabetes in NOD mice. recognising a recombinant MPO subunit. Adv Exp Med Biol Lancet 1994; 343:704–7. 1993; 336:87–92. 40 Ransohoff RM, Tuohy V, Lehmann P. The immunology of multiple 37 Gaur A, Fathman CG. Immunotherapeutic strategies directed at the sclerosis: new intricacies and insights. Curr Opin Neurol 1994;

trimolecular complex. Adv Immunol 1995; 56:219–65. 7:242–9. 5 1996 Blackwell Science Ltd, Clinical and Experimental Immunology, 103:253–258