You are on page 1of 5

Lupus (1998) 7, 280 ± 284

ß 1998 Stockton Press. All rights reserved 0961-2033/98 $12.00


http://www.stockton-press.co.uk/lup

REVIEW

Pathogenesis of vasculitis
CGM Kallenberg and P Heeringa
Department of Clinical Immunology, University Hospital Groningen, The Netherlands

This review discusses current thoughts on the pathogenesis of vasculitis. Secondary vasculitides,
frequently associated with infections or systemic autoimmune diseases, are, in most cases,
characterized by immune deposits in the vessel wall, which probably underlies the development of
lesions. In the primary vasculitides, immune deposits are generally absent. A group of primary
vasculitides is, however, strongly associated with anti-neutrophil cytoplasmic autoantibodies
(ANCA). Various in vitro and in vivo experimental data suggest that those ANCA are involved in
the pathogenesis of lesions in the associated disorders.

Keywords: systemic vasculitis; anti-neutrophil cytoplasmic antibodies; Wegener's granulomato-


sis; pathophysiology

Introduction secondary vasculitides will be brie¯y discussed. The


main focus will, however, be on recent studies on the
pathogenesis of the primary vasculitides.
The systemic vasculitides comprise a diverse group of
disorders characterized by in¯ammation of the blood
vessel wall. The localization and size of the in¯amed Secondary vasculitides
vessels and the nature of the in¯ammatory process, as
well as its systemic effects, determine the clinical
presentation of the disorder. As stated before, many cases of secondary vasculitis
Vasculitis may, incidentally, result from direct demonstrate immune deposits within the vessel wall
invasion of the blood vessel wall by micro-organisms by direct immuno¯uorescence. A classical example is
or from microbial embolization.1 Many cases of the occurrence of leucocytoclastic vasculitis, clini-
vasculitis are secondary to other diseases. These cally apparent as purpura, in patients with cryoglobu-
secondary vasculitides are frequently characterized by linemia as part of Sjùgren's syndrome. Cryoglobulins
immune complex deposition, as will be discussed later composed of monoclonal rheumatoid factor and
on. In the remainder of cases in which vasculitis polyclonal immunoglobulin G are deposited in the
is histopathologically demonstrable, no underlying vessel wall and activate the complement cascade
disorder or direct infection of the vessel wall can be along the classical pathway. Neutrophils are attracted
detected. Those primary or idiopathic vasculitides and activated, and invade the vessel wall resulting
constitute a heterogeneous group of disorders that are in leucocytoclastic vasculitis. Activation of the
classi®ed based on the size of the vessels involved, the complement cascade with consumption of com-
histopathological ®ndings and the clinical symptoms plement components is demonstrable by decreased
(Table 1).2 ± 4 The pathogenesis of the primary levels of complement C3 and C4 and increased levels
vasculitides is far from understood, but recent of C3d, a breakdown product of C3 after activation. In
developments have shed some light on the patho- addition, circulating immune complexes can be
physiological processes underlying those disorders. detected in serum.
Immune complex-mediated vasculitis has been
In this review the pathogenetic pathways in the
suggested to occur in association with systemic
autoimmune diseases and infections.5 In the systemic
Correspondence: Prof.dr. CGM Kallenberg, Department of Clinical
autoimmune diseases, particularly systemic lupus
Immunology, University Hospital, Hanzeplein 1, 9713 GZ Groningen, erythematosus (SLE), nuclear antigens and their
The Netherlands. corresponding antibodies have been thought to be

Downloaded from lup.sagepub.com at Bobst Library, New York University on June 4, 2015
Pathogenesis of vasculitis
CGM Kallenberg and P Heeringa
281
Table 1 Classi®cation of primary vasculitides according to the IgG non-speci®cally,9 also resulting in immune com-
Chapel Hill Consensus Conference plex formation. Thus, microbial agents are de®nitely
Large vessel vasculitis involved in the etiopathogenesis of vasculitis. The
Giant cell (temporal) arteritis precise pathophysiology of the associated secondary
Takayasu arteritis vasculitides is still under study. Also, in other forms of
Medium sized vessel vasculitis
Polyarteritis nodosa secondary vasculitis, for example, drug-related, the
Kawasaki disease exact pathophysiological mechanisms have not been
Small vessel vasculitis fully elucidated.
Wegener's granulomatosisa
Churg ± Strauss syndromea
Microscopic polyangiitisa
SchoÈnlein ± Henoch purpura Primary vasculitides
Essential cryoglobulinemia vasculitis
Cutaneous leukocytoclastic angiitis
a
Strongly associated with anti-neutrophil cytoplasmic antibodies (adapted
In classical polyarteritis nodosa (PAN) viral infections
from Ref. 3). seem to be involved in some 20% of cases.8 Hepatitis
B virus (HBV) and HCV, as well as HIV and,
pathogenetically involved. Recent ®ndings do, how- possibly, parvovirus, have been thought to be causally
ever, support an (additional) mechanism in which the related to the development of PAN in these cases. The
antigen is ®rst planted in the vessel wall. Binding of pathophysiological mechanisms are only partly under-
nucleohistones to the (glomerular) basement mem- stood, as discussed in the previous section. As
brane in lupus nephritis, due to charge interaction, is mentioned before, HBV-, HCV-, and HIV-infection
followed by binding of antibodies.6 This in situ are in many more cases associated with mixed
immune complex formation may be operative in cryoglobulinemic vasculitis.7,8 How those viruses
many cases of immune complex-mediated vasculitis. induce cryoglobulins is still open for study.
Besides systemic autoimmune diseases, infections In the majority of cases with primary vasculitis,
are associated with vasculitis. Bacterial and viral exogenous factors are not directly demonstrable.
infections may induce the formation of immune There is increasing evidence that autoimmune re-
complexes consisting of microbial antigens and their sponses are operative in those disorders. Although the
corresponding antibodies which, either by deposition possible pathogenic role of anti-endothelial cell
from the circulation or by in situ formation, results in antibodies and the anti-phospholipid antibodies in
vasculitides. The association of cryoglobulinemia with, systemic vasculitis (reviewed in Ref. 10) should be
particularly, hepatitis C virus (HCV)7,8 is remarkable in mentioned, the focus has been on anti-neutrophil
this respect. Different mechanisms may, however, be cytoplasmic antibodies in recent years.
operative. First, microbes may directly invade the
vessel wall followed by an infectious in¯ammatory
process.1 Secondly, certain viruses, such as cytomega-
Anti-neutrophil cytoplasmic antibodies
lovirus, can penetrate endothelial cells and activate
(ANCA) and the pathophysiology of systemic
those cells leading, along pathways that still have to be
vasculitis
explored, to vasculitis.8 Thirdly, bacterial antigens from
Staphylococci, such as Staphylococcal neutral phos- As shown in Table 1, Wegener's granulomatosis
phatase, can attach to basement membranes and bind (WG), microscopic polyangiitis (MPA), Churg

Table 2 Pathogenic effects of anti-neutrophil cytoplasmic antibodies (ANCA) as derived from in vitro studies
1. Activation of neutrophils primed with low dose TNFa resulting in the production of reactive oxygen intermediates (ROI) and the release of lysosomal
enzymes.13 This process only occurs when neutrophils are allowed to adhere to a surface; this adherence particularly involves the b2-integrins.14
Priming (and apoptosis) results in expression of the target antigens of ANCA on the cell surface making them accessible for ANCA.15;16
Neutrophil activation by ANCA is also dependent, probably, on the interaction of the Fc-part of ANCA with, particularly, the FcgRIIa-receptor on
neutrophils.17
ANCA-induced ROI production and degranulation is enhanced in the presence of extracellular arachidonic acid.18
2. Activation of the 5-lipoxygenase pathway in neutrophils inducing the production of large amounts of leukotriene B4, a potent chemo-attractant for
neutrophils.18
3. Induction of cytokine production such as IL-1b by neutrophils.19
4. Induction of the production of ROI20 and monocyte chemo-attractant protein-1,21 by monocytes.
5. Lysis of endothelial cells previously incubated with myeloperoxidase (MPO) or proteinase 3 (Pr3).22,23
Upregulation of adhesion molecules on (primed) endothelial cells.24
Lysis of (primed) endothelial cells in the presence of neutrophils.25
6. Interference with the inactivation of Pr3 by a1-antitrypsin26 which may result in persistently active Pr3.

Downloaded from lup.sagepub.com at Bobst Library, New York University on June 4, 2015
Pathogenesis of vasculitis
CGM Kallenberg and P Heeringa
282
Strauss Syndrome (CSS) and idiopathic necrotizing

Pauci-immune character disputed by


Pre-treatment with antibiotics gives
crescentic glomerulonephritis (NCGN) are closely

Selective breeding for crescent


Scanty immune deposits in the
associated with autoantibodies to either proteinase 3

Anti-anti-idiotypic responses
severe reduction of lesions
(Pr3) or myeloperoxidase (MPO).11 Changes in titers

No lesions in the kidneys


Remarks
of those autoantibodies seem to re¯ect changes in
disease activity.12 Those ®ndings suggest that the
autoantibodies are involved in the pathophysiology of
the associated disorders. Experimental evidence for

formation

others36
the pathogenic potential of ANCA has been derived

kidney
mainly from in vitro studies. Those studies are
summarized in Table 2. One may conclude from
those studies, that ANCA are able to aggravate a

Pauci-immune necrotizing crescentic


subclinical in¯ammatory process by activating, along

Small vessel vasculitis in the gut,

Systemic vasculitis (kidney, gall

glomerulonephritis, vasculitis in

Sterile micro-abscesses in lungs


several lines, neutrophils and monocytes. In addition,

in¯ammatory changes in many

bladder), lymphoproliferation
endothelial cells are potentially a target of ANCA,

Vasculitis in lungs and gut


Pauci-immune crescentic
either indirectly via endothelial binding of Pr3=MPO

Lesions
released from neutrophils, to which the antibodies can

glomerulonephritis
bind, or directly by reacting with Pr3 expressed on the
surface of endothelial cells as demonstrated by Mayet

other organs
Table 3 Animal models of anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis/glomerulonephritis
et al,27 but disputed by others.28

organs
Although all of the aforementioned data suggest
that ANCA are pathogenic, more direct proof
should come from in vivo studies on exper-

Multiple autoantibodies, polyreactive


imental animals. Until now, a fully satisfactory

Amongst others, autoantibodies to


with MPO, DNA, lactoferrin, etc.
animal model for ANCA-associated vasculitis has

Mouse antibodies to human Pr3


Anti-MPO as part of polyclonal
Autoantibody response

cross-reacting with rat MPO

cross-reacting with rat MPO


not been developed. Nevertheless, several in vivo

Antibodies to human MPO

Antibodies to human MPO


models are presently available that support a patho-
autoantibody responses

genic role for ANCA,29 summarized in Table 3. In


certain models,30 ± 34 vasculitis and=or crescentic
glomerulonephritis develops spontaneously in con-
junction with autoantibodies to MPO. The autoanti-
body response to MPO is, however, part of a more
MPO

polyclonal autoantibody response which does not


allow the analysis of the speci®c contribution of anti-
MPO to the development of vasculitis. The anti-anti- Immunization with human anti-Pr3
Immunization with human MPO,

Immunization with human MPO,


systemic injection of products of
products of activated neutrophils
ex vivo perfusion of kidney with
Injection with mercuric chloride

idiotypic model38 for anti-Pr3 is interesting but awaits


con®rmation as well as further analysis of the lesions.
It is important to mention that induction of ANCA
Induction

activated neutrophils

(anti-MPO) alone does not result in lesions.35 ± 37


When, however, products of activated neutrophils
are present in the circulation, pauci-immune crescen-
Spontaneous

Spontaneous

tic glomerulonephritis35 and systemic vasculitis37 can


develop.
MPO ˆ myeloperoxidase; Pr3 ˆ proteinase 3.

All of the aforementioned in vitro and in vivo


experimental data lead to the conclusion that ANCA
Brown-Norway rat

Brown-Norway rat

Brown-Norway rat

in combination with exogenous factor(s) that induce


MRL=lpr mouse
Animal strain

pre-activation (priming) of neutrophils, may result in


Balb-c mouse
SCG mouse

systemic vasculitis and glomerulonephritis. The


exogenous factors have not yet been de®ned, but the
observations that carriage of Staphylococcus aureus
is an important risk factor for relapses in WG39
and that maintenance treatment with trimethoprim=
Various31 ± 33
30

Heeringa37
Brouwer35
Mathieson

sulfamethoxazole prevents relapses40 suggest that


34

Blank38
Author

Kinjoh

microbial factors are also involved in the primary


vasculitides.

Downloaded from lup.sagepub.com at Bobst Library, New York University on June 4, 2015
Pathogenesis of vasculitis
CGM Kallenberg and P Heeringa
283
T-lymphocytes in the systemic vasculitides 7 Agnello V, Chung RT, Kaplan LM. A role for hepatitis C virus infection in
type II cryoglobulinemia. N Engl J Med 1992; 327: 1490 ± 1495.
8 Guillevin L, Lhote F, Gherardi R. The spectrum and treatment of virus-
associated vasculitides. Curr Op Rheumatol 1997; 9: 31 ± 36.
Whereas many experimental and clinical data are 9 Yousif Y et al. Staphylococcal neutral phosphatase. A highly cationic
available on the role of autoantibodies in systemic molecule with binding properties for immunoglobulin. APMIS 1991;
vasculitis, the role of T-cells has been explored to a 102: 897 ± 900.
10 Kallenberg CGM. Laboratory ®ndings in the vasculitides. Bailliere's
limited extent only. In the ANCA-associated vasculi- Clin Rheumatol 1997; 11: 395 ± 421.
tides T-cell responses of peripheral blood mono- 11 Kallenberg CGM, Brouwer E, Weening JJ, Cohen Tervaert JW. Anti-
nuclear cells to Pr3 but hardly to MPO have been neutrophil cytoplasmic antibodies: current diagnostic and pathophy-
siological potential. Kidney Int 1994; 46: 1 ± 15.
documented in patients but also, albeit to a lesser 12 Cohen Tervaert JW, Stegeman CA, Kallenberg CGM. Serial ANCA
extent, in healthy controls.41 The functional character- testing is useful in monitoring disease activity of patients with ANCA-
istics of those cells, the epitopes involved, and the associated vasculitides. Sarcoidosis 1996; 13: 241 ± 245.
13 Falk RJ, Terrell RS, Charles LA, Jennette JC. Anti-neutrophil cytoplasmic
analysis of tissue in®ltrating lymphocytes, are still autoantibodies induce neutrophils to degranulate and produce oxygen
open to study. More interestingly, skewing of the T- radicals in vitro. Proc Natl Acad Sci 1990; 87: 4115 ± 4119.
14 Reumaux D, Vossebeld PJM, Roos D, Verhoeven AJ. Effects of tumor
cell repertoire due to preferential use of certain Vb necrosis factor-induced integrin activation on Fcg receptor II-mediated
genes have been noted in a number of vasculitides, signal transduction: relevance for activation of neutrophils by anti-
notably in Kawasaki disease,42 microscopic polyan- proteinase 3 or anti-myeloperoxidase antibodies. Blood 1995; 86:
giitis,43 and, very recently, in WG.44 These data can be 3189 ± 3195.
15 Csernok E et al. Activated neutrophils express proteinase 3 on their
consistent with antigen or superantigen, particularly plasma membrane in vitro and in vivo. Clin Exp Immunol 1994; 95:
staphylococcal superantigen, stimulation. It should, 244 ± 250.
16 Gilligan HM et al. Antineutrophil cytoplasmic autoantibodies interact
however, be kept in mind that shaping of the with primary granule constituents on the surface of apoptotic
peripheral blood T-cell repertoire, also occurs with neutrophils in the absence of neutrophil priming. J Exp Med 1996;
increasing age.45 Certainly, more work should be done 184: 2231 ± 2241.
17 Mulder AHL et al. Activation of granulocytes by anti-neutrophil
on T-cells in systemic vasculitis. In this respect, the cytoplasmic antibodies (ANCA); a FcgRII-dependent process. Clin
studies of Weyand and Goronzy46 on T-cell macro- Exp Immunol 1994; 78: 270 ± 278.
phage interactions in the vessel wall of giant cell 18 Grimminger F et al. Neutrophil activation by anti-proteinase 3
antibodies in Wegener's Granulomatosis: Role of exogenous arachi-
arteritis are promising. donic acid and leukotriene B4 generation. J Exp Med 1996; 184:
1567 ± 1572.
19 Brooks CJ et al. IL-1b production by human polymorpho-
Conclusion nuclear leukocytes stimulated by anti-neutrophil cytoplasmic autoanti-
bodies: relevance to systemic vasculitis. Clin Exp Immunol 1996; 106:
273 ± 279.
20 Ewert BH, Jennette JC, Falk RJ. The pathogenic role of anti-neutrophil
The etiopathogenesis of the primary systemic vasculi- cytoplasmic autoantibodies. Am J Kidney Dis 1991; 18 188 ± 195.
tides has not yet been clari®ed in detail. The detection 21 Casselman BL, Kilgore KS, Miller BF, Warren JS. Antibodies to
of anti-neutrophil cytoplasmic antibodies in a number neutrophil cytoplasmic antigens induce monocyte chemoattractant
protein-1 secretion from human monocytes. J Lab Clin Med 1995; 126:
of those diseases has, however, given a huge impulse 495 ± 502.
to the unraveling of pathogenetic pathways operative 22 Savage CO, Pottinger BE, Gaskin G, Pusey CD, Pearson JD.
in the vasculitides. In addition, further analysis of the Autoantibodies developing to myeloperoxidase and proteinase 3 in
systemic vasculitis stimulate neutrophil cytotoxicity towards cultured
speci®c immune responses underlying the idiopathic endothelial cells. Am J Path 1992; 141: 335 ± 342.
vasculitides may bene®t from recent developments in 23 Ewert BH, Jennette JC, Falk RJ. Anti-myeloperoxidase antibodies
molecular immunology. stimulate neutrophils to damage human endothelial cells. Kidney Int
1992; 41: 375 ± 383.
24 Mayet WJ, Meyer zum Buschenfelde KH. Antibodies to proteinase 3
increase adhesion of neutrophils to human endothelial cells. Clin Exp
References Immunol 1993; 94: 440 ± 446.
25 Mayet WJ, Schwarting A, Meyer zum Buschenfelde KH. Cytotoxic
effects of antibodies to proteinase 3 (C-ANCA) on human endothelial
1 Somer T, Finegold SM. Vasculitides associated with infections, cells. Clin Exp Immunol 1994; 97: 458 ± 465.
immunization and antimicrobial drugs. Clin Inf Dis 1995; 20: 1010 ± 26 Van der Wiel BA et al. Interference of Wegener's granulomatosis
1036. autoantibodies with neutrophil proteinase 3 activity. Clin Exp Immunol
2 Hunder GG et al. The American College of Rheumatology 1990 1992; 90: 409 ± 414.
criteria for the classi®cation of vasculitis. Arthritis Rheum 1990; 33: 27 Mayet WJ et al. Human endothelial cells express proteinase 3, the
1065 ± 1067. target antigen of anticytoplasmic antibodies in Wegener's Granulo-
3 Jennette JC et al. Nomenclature of systemic vasculitides: the proposal matosis. Blood 1993; 82: 1221 ± 1229.
of an international consensus conference. Arthritis Rheum 1994; 37: 28 King WJ et al. Endothelial cells and renal epithelial cells do not
187 ± 192. express the Wegener's autoantigen, proteinase 3. Clin Exp Immunol
4 Lie JT. Nomenclature and classi®cation of vasculitis: plus cËa change, 1995; 102: 98 ± 105.
plus c'est la meÃme chose. Arthritis Rheum 1994; 37: 181 ± 186. 29 Heeringa P et al. Animal models of anti-neutrophil cytoplasmic
5 Theo®lopoulos AN, Dixon FJ. Immune complexes in human diseases. antibody associated vasculitis. Kidney Int (in press).
Am J Pathol 1980; 100: 530 ± 594. 30 Mathieson PW, Thiru S, Oliveira DBG. Mercuric chloride-treated
6 Bruggen MCJ van, Berden JHM. Central role for nucleosomes in Brown Norway rats develop widespread tissue injury including
lupus. Nephrol Dial Transplant 1996; 11: 1219 ± 1222. necrotizing vasculitis. Lab Invest 1992; 67: 121 ± 129.

Downloaded from lup.sagepub.com at Bobst Library, New York University on June 4, 2015
Pathogenesis of vasculitis
CGM Kallenberg and P Heeringa
284
31 Hewicker M, Trautwein G. Sequential study of vasculitis in MRL 39 Stegeman CA et al. Association of chronic nasal carriage of
mice. Lab Anim 1987; 21: 335 ± 341. Staphylococcus aureus and higher relapse rates in Wegener's
32 Moyer CF, Strandberg DJ, Reinish CL. Systemic mononuclear cell granulomatosis. Ann Intern Med 1994; 113: 12 ± 17.
vasculitis in MRL=Mp-lpr=lpr mice: a histological and immunocyto- 40 Stegeman CA, Cohen Tervaert JW, Jong de PE, Kallenberg CGM.
chemical analysis. Am J Path 1987; 127: 229 ± 242. Trimethoprim (Cotrimoxazole) for the prevention of relapses of
33 Harper JM, Lockwood CM, Cooke A. Anti-neutrophil cytoplasm Wegener's granulomatosis. N Engl J Med 1996; 335: 16 ± 20.
antibody in MRL-lpr=lpr mice. Clin Exp Immunol 1993; 93(suppl 1): 22. 41 Brouwer E et al. T-cell reactivity to proteinase 3 and myeloperoxidase
34 Kinjoh K, Kyogoku M, Good RA. Genetic selection for crescent in patients with Wegener's granulomatosis. Clin Exp Immunol 1994;
formation yields mouse strain with rapidly progressive glomerulo- 98: 448 ± 453.
nephritis and small vessel vasculitis. Proc Natl Acad Sci 1993; 90: 42 Leung DYM et al. Evidence for superantigen involvement in
3413 ± 3417. cardiovascular injury due to Kawasaki syndrome. J Immunol 1995;
35 Brouwer E et al. Anti-myeloperoxidase associated proliferative 155: 5018 ± 5021.
glomerulonephritis; an animal model. J Exp Med 1993; 177: 905 ± 914. 43 Simpson IJ et al. Peripheral blood T lymphocytes in systemic
36 Yang JJ, Jennette JC, Falk RJ. Immune complex glomerulonephritis is vasculitis: increased T cell receptor Vb2 gene usage in microscopic
induced in rats immunized with heterologous myeloperoxidase. Clin polyarteritis. Clin Exp Immunol 1995; 101: 220 ± 226.
Exp Immunol 1994; 97: 466 ± 473. 44 Popa ER et al. Skewed TCR Vb usage in Wegener's Granulomatosis
37 Heeringa P et al. Systemic injection of products of activated neutrophils suggests stimulation by superantigens. J Am Soc Nephrol 1997; 8:
and H2O2 in myeloperoxidase-immunized rats leads to necrotizing 543A.
vasculitis in the lungs and gut. Am J Path 1997; 151: 131 ± 139. 45 Posnett DN, Sinha R, Kabak S, Russo C. Clonal populations of T cells
38 Blank M et al. Immunization with anti-neutrophil cytoplasmic in normal elderly humans: the T cell equivalents to `benign
antibody (ANCA) induces the production of mouse ANCA and monoclonal gammopathy'. J Exp Med 1994; 179: 609 ± 618.
perivascular lymphocyte in®ltration. Clin Exp Immunol 1995; 102: 46 Weyand CM, Goronzy JJ. Multisystem interactions in the pathogenesis
120 ± 130. of vasculitis. Curr Op Rheumatol 1997; 9: 3 ± 11.

Downloaded from lup.sagepub.com at Bobst Library, New York University on June 4, 2015

You might also like