Вы находитесь на странице: 1из 11

REVIEWS

Pathogenesis of antineutrophil cytoplasmic


autoantibody-mediated disease
J. Charles Jennette and Ronald J. Falk
Abstract | Antineutrophil cytoplasmic autoantibodies (ANCAs) are the probable cause of a distinct form of
vasculitis that can be accompanied by necrotizing granulomatosis. Clinical and experimental evidence supports
a pathogenesis that is driven by ANCA-induced activation of neutrophils and monocytes, producing destructive
necrotizing vascular and extravascular inflammation. Pathogenic ANCAs can originate from precursor natural
autoantibodies. Pathogenic transformation might be initiated by commensal or pathogenic microbes, legal or
illegal drugs, exogenous or endogenous autoantigen complementary peptides, or dysregulated autoantigen
expression. The ANCA autoimmune response is facilitated by insufficient Tcell and Bcell regulation.
Aputative pathogenic mechanism for vascular inflammation begins with ANCA-induced activation of primed
neutrophils and monocytes leading to activation of the alternative complement pathway, which sets in motion
an inflammatory amplification loop in the vessel wall that attracts and activates neutrophils with resultant
respiratory burst, degranulation, extrusion of neutrophil extracellular traps, apoptosis and necrosis. The
pathogenesis of extravascular granulomatosis is less clear, but a feasible scenario proposes that a prodromal
infectious or allergic condition positions primed neutrophils in extravascular tissue in which they can be
activated by ANCAs in interstitial fluid to produce extravascular necrotizing injury that would initiate an innate
granulomatous inflammatory response to wall off the necrotic debris.

Jennette, J.C. & Falk, R.J. Nat. Rev. Rheumatol. advance online publication 8 July 2014; doi:10.1038/nrrheum.2014.103

Introduction
Antineutrophil cytoplasmic autoantibodies (ANCAs) haemorrhage caused by alveolar capillaritis; glomerulo-
are associated with, and are the probable cause of, a nephritis caused by glomerular capillaritis; peripheral
distinct form of vasculitis that can affect any organ in neuropathy caused by epineural arteritis; and ocular
the body. Although small vessels are the predominant inflammation caused by vasculitis in small vessels in the
target, ANCA-associated vasculitis (AAV) can affect eye and orbit. In addition to vasculitis, some variants of
many different types of vessels including art eries, ANCA-associated disease have extravascular necrotiz-
arterioles, capillaries, venules and veins (Figure1).1 ing granulomatous inflammation (granulomatosis)
Immunopathologically, AAV has an absence or paucity seemingly not arising from vascular inflammationthat
of immunoglobulin deposition in injured vessels (pauci- most often affects the upper and lower respiratory tracts
immune vasculitis), compared with the more extensive but can affect any organ.
deposition of immunoglobulin in immune-complex- On the basis of the distribution of vascular inflam-
mediated small-vessel vasculitis.1 AAV can be limited mation and the presence or absence of granulomatosis
to one organ at the time of presentation (for example, and asthma, systemic AAV is categorized as microscopic
lung-limited disease or renal-limited disease) or involve polyangiitis (MPA) if there is vasculitis but no evidence
multiple organs. Frequent target tissues are the upper of granulomatosis or asthma, granulomatosis with poly-
and lower respiratory tract, kidneys, skin and periph- angiitis (GPA) if there is granulomatosis but no asthma,
eral nerves. Onset and exacerbations induce signs and and eosinophilic granulomatosis with polya ngiitis
symptoms of high levels of circulating inflammatory (EGPA; also known as ChurgStrauss syndrome) if
Department of cytokines, such as fever, arthralgia and myalgia. Inaddi- there is granulomatosis, asthma and blood eosinophilia.1
Pathology and tion to these nonspecific systemic inflammatory mani- Organ-limited disease, including isolated pauci-immune
Laboratory Medicine,
School of Medicine, festations, inflammation of vessel walls causes more necrotizing and crescentic glomerulonephritis, can be
University of North specific signs and symptoms of vasculitis depending considered limited variants of MPA. Limited variants of
Carolina at Chapel Hill,
308 Brinkhous-Bullitt
on which vessels and which organs are affected, for GPA and EGPA can be confined to the respiratory tract.
Building, CB#7525, example purpura caused by dermal venulitis; pulmonary AAV can also be classified on the basis of auto
Chapel Hill, antigen specificity. In patients with vasculitis, the two
NC27599-7525,
USA (J.C.J., R.J.F.).
Competing interests best-d ocumented autoantigen targets of ANCA are
J.C.J. has acted as a consultant for Amicus Therapeutics,
myelop eroxidase (MPO) 2 and proteinase3 (PR3). 35
Correspondence to: Genentech, GlaxoSmithKline and Protalix BioTherapeutics, and
J.C.J. has undertaken research in collaboration with ChemoCentryx. Classification of AAV by antigen specificity clearly
jcj@med.unc.edu R.J.F. declares no competing interests. shows differences in clinical presentations and outcomes,

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 1


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points with pauci-immune necrotizing glomerulonephritis,


including patients who were positive for MPO-ANCA
Antineutrophil cytoplasmic autoantibodies (ANCAs) are associated with and
or PR3-ANCA and others who were negative for both.
probably cause pauci-immune systemic necrotizing small-vessel vasculitis
and glomerulonephritis However, Roth etal.11 were unable to confirm this high
ANCAs specific for proteinase3 or myeloperoxidase occur with organ- frequency of LAMP2-ANCA in these patients. Similarly,
limited disease, microscopic polyangiitis, granulomatosis with polyangiitis Kawakami etal. did not detect LAMP2-ANCA in
or eosinophilic granulomatosis with polyangiitis patients with MPA, although they did report evidence for
ANCAs seem to cause vasculitis by activating circulating primed neutrophils and LAMP2-ANCA in some patients with cutaneous poly
causing them to attach to, penetrate and damage vessel walls by undergoing arteritis nodosa12 and IgA vasculitis (formerly known
respiratory burst, degranulation, NETosis, apoptosis and necrosis
as HenochSchnlein purpura).13 ANCAs also occur
ANCA-induced neutrophil activation also releases factors that activate the
alternative complement pathway, which establishes a destructive inflammatory
in patients with inflammatory bowel disease, and are
amplification loop that attracts and activates more neutrophils that, in turn, detected by indirect immunofluorescence assay in ~60%
further activates the complement system of patients with ulcerative colitis and ~20% of those with
ANCA-associated granulomatosis might result from the same pathogenic Crohn disease.1416 In this setting, ANCAs only rarely
sequence of events involving extravascular primed neutrophils and interstitial- have specificity for PR3, MPO or elastase: the major
fluid ANCAs, followed by a granulomatous reaction to wall off the resulting autoantigens are catalase, enolase, lactotransferrin
extravascular necrosis and bactericidal permeability-increasing protein. 1416
ANCA, usually MPOANCA, occurs in a small minor-
Immune complex small-vessel vasculitis ity of patients with systemic lupus erythematosus who
Cryoglobulinemic vasculitis
IgA vasculitis (HenochSchnlein)
seem to to have vasculitic features of ANCA-associated
Hypocomplementemic urticarial vasculitis disease. 17 Approximately 30% of patients with anti
(Anti-C1q vasculitis) glomerular basement membrane disease have ANCA,
Medium-vessel vasculitis Anti-GBM disease
usually MPOANCA.18
Polyarteritis nodosa An important role for ANCAs in the pathogenesis of
Kawasaki disease vasculitis and glomerulonephritis is supported by clinical
evidence and by invitro and invivo experimental data
(summarized in Box1). The leading theory proposes that
circulating neutrophils and monocytes that have been
primed by inflammatory stimuli display ANCA antigens
ANCA-associated small-vessel vasculitis at or near the cell surface, and that interaction of these
Microscopic polyangiitis
Granulomatosis with polyangiitis (Wegener)
antigens with ANCAs results in neutrophil activation
Eosinophilic granulomatosis with polyangiitis (ChurgStrauss) and initiation of vascular inflammation. An extension
of this theory proposes that primed extravascular neutro-
Large-vessel vasculitis phils interact with interstitial ANCAs, causing necrotiz-
Takayasu arteritis ing inflammation and resultant reactive granulomatous
Giant cell arteritis inflammation. This Review summarizes observations
Figure 1 | Diagram depicting the predominant types of vessels affected by major that support these pathogenic theories as well as puta-
categories of systemic vasculitis. Note that vasculitis that is associated with tive mechanisms for the origin of the pathogenic ANCA
ANCAs can target a broad range of vessel types including medium and small autoimmune response.
arteries, arterioles, capillaries, venules and veins. Large arteries, such as main
visceral arteries, are rarely affected by AAV. Thus, the types of vessels affected Pathology of ANCA-associated disease
by AAV overlap with other variants of systemic vasculitis, which results in many
Any valid theories about pathogenic mechanisms in
overlapping clinical signs and symptoms of disease. Abbreviations: AAV, ANCA-
associated vasculitis; ANCA, antineutrophil cytoplasmic antibody; GBM, glomerular
ANCA-associated disease must explain the development
basement membrane. Reproduced with permission from John Wiley & Sons, Inc. of the observed pathologic lesions. The acute and chronic
Jennette, J.C. etal. Arthritis Rheum. 65, 111 (2013). lesions described in this section provide g uidance in
formulating pathogenic theories.
organ system involvement, patterns of extravascular
inflammation, and genetic associations including HLA Vasculitis and glomerulonephritis
associations.6,7 The most informative AAV classifica- In patients1921 and experimental animal models,22 the
tion scheme includes both the ANCA antigen specific- acute vascular and extravascular lesions of ANCA-
ity and the clinicopathological phenotype, for example associated disease begin as neutrophil-rich necrotizing
MPOANCA MPA or PR3ANCA MPA.1 inflammation. In vessels, the earliest lesions have mar-
MPO and PR3 are not the only autoantigens recog- gination and diapedesis of predominantly neutrophils
nized by ANCAs. For instance, ANCAs specific for with admixed monocytes (Figure2a,b). Areas of segmen-
elastase occur, and are most often identified, in patients tal vascular necrosis also develop perivascular infiltrates
with drug-induced ANCA-associated disease, including that initially have numerous neutrophils.
disease induced by cocaine adulterated with levamisole.8,9 Within hours, neutrophils undergo apoptosis and
A controversial autoantigen specificity is for lysosomal- necrosis (Figure2a,b). The early destruction of neutro-
associated membrane protein 2 (LAMP2). Kain etal.10 phils by apoptosis and necrosis during ANCA-associated
reported finding LAMP2-ANCA in ~90% of patients glomerulonephritis and vasculitis is supported by the

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Box 1 | Evidence that ANCAs are pathogenic necrotizing inflammation are subsequently replaced by
monocytes and macrophages.22
Clinical evidence
Within 12weeks, vascular segments transition
Strong association with ANCA (MPA >90%, GPA >90%, EGPA >40%*)
Partial correlation of ANCA titre with disease activity
through the monocyte and/or macrophage-rich phase of
Correlation of ANCA epitope specificity with disease activity (MPOANCA only) inflammation to a phase of scarring that involves inter-
Disease induction by transplacental transfer of ANCA (one MPOANCA case report) stitial deposition of collagen. The extent of scarring, and
Similar disease associated with drug-induced ANCA the likelihood of return to normal structure and func-
HLA genetic associations with MPOANCA and PR3ANCA-associated disease tion, depends on the severity and duration of the inflam-
Response to immunosuppressive therapy that targets Bcells matory injury. Areas of mild segmental vascular injury
Invitro evidence can undergo complete remodelling to restore normal
Activation of cytokine-primed neutrophils by ANCA IgG architecture, whereas severe vascular injury could result
Endothelial injury by ANCA-activated neutrophils in irreversible total vascular occlusion.
Alternative complement pathway activation by ANCA-activated neutrophils
Evidence from animal models Granulomatosis
Induction of pauci-immune vasculitis, glomerulonephritis and granulomatosis
Patients with GPA and EGPA have nodular extra
in mice and rats by anti-MPO IgG
Prevention of murine anti-MPO IgG-induced disease by deficiency of neutrophils
vascular inflammatory lesions that contain numerous
Prevention of murine anti-MPO IgG-induced disease by blockade of alternative macrophages, often including multinucleated giant cells
complement pathway activation or blockade of C5a receptors (Figure3).1921 In an early granulomatous pulmonary
*More than 75% of patients with EGPA and glomerulonephritis have ANCA. Convincing animal lesion, the centre is in essence a microabscess contain-
models of MPOANCA-associated disease have been described, but only less-convincing ing neutrophils in varying stages of necrosis and apop-
animal models of PR3ANCA-associated disease have been described. Abbreviations: ANCA,
antineutrophil cytoplasmic autoantibody; EGPA, eosinophilic granulomatosis with polyangiitis; tosis, and the margin has a thin band of macrophages
GPA, granulomatosis with polyangiitis; MPA, microscopic polyangiitis; MPO, myeloperoxidase; that are responding to the necrosis (Figure3a). In a later
PR3, proteinase3. stage of granulomatosis, the centre of the lesion contains
amorphous necrotic debris that is walled off from the
adjacent lung parenchyma by a well-defined band of
frequent observation of leukocytoclasia (fragmentation epithelioid macrophages (macrophages with elongated
of nuclei) at sites of acute fibrinoid necrosis (Figure2d). nuclei and abundant acidophilic cytoplasm, resembling
Conspicuous leukocytoclasia with numerous nuclear epithelial cells) (Figure3b). At this stage, neutrophils
fragments could result from the intensity of neutrophil are no longer conspicuous, and the granulomatous
infiltration or reduced clearance of neutrophil fragments, inflammation contains progressively more lymphoid
or both. Neutrophil cytoplasmic and nuclear constitu- cells including effector Tcells, memory Tcells, Bcells,
ents, including neutrophil extracellular traps (NETs), dendritic cells, macrophages and plasma cells.30 At a still-
are present at sites of inflammation and acute necro- later stage, the necrotic debris is replaced by scar tissue,
sis.2628 NETs comprise extracellular fibrillary material and lymphocytes, including organized lymphoid folli-
containing chromatin and granule proteins extruded by cles, become components of the chronic granulomatous
activated neutrophils. inflammation. End-stage lesions are predominantly scar
Necrotizing injury to vessel walls results in haemor- tissue, often with no definitive histological features of the
rhage and release of plasma proteins into the vessel walls earlier granulomatous inflammation.
and adjacent extravascular tissue. These proteins include
coagulation factors, which are activated by thrombogenic Genesis of the ANCA autoimmune response
cellular and tissue debris, and tissue factor, which results The evidence suggests that multiple genetic and environ-
in the formation of fibrin within areas of fibrinoid necro- mental factors converge to induce the onset of ANCA-
sis (Figure2c,d). Fibrin formation also might be facili- associated disease, and these factors have modulatory
tated by tissue factor present in NETs.29 In glomeruli, effects on the clinical and pathological phenotype of
foci of segmental fibroid necrosis develop adjacent cel- disease. These factors differ among patients; for example,
lular reactions (crescents) composed predominantly in a given patient the pivotal aetiological event could be
of monocytes, macrophages and activated epithelial an infection, a drug, impaired immune regulation or
cells(Figure2d). dysregulation of genomic expression of autoantigens,
Within days, the initial acute neutrophil-rich inflam- or combinations of these and other factors.
mation and necrosis is replaced by inflammation with
a predominance of monocytes and macrophages and, Natural ANCAs
eventually, Tcells (Figure2c). Vascular and extravascu- Although Ehrlich described the horror of autotoxicus
lar inflammatory infiltrates also often contain eosino- and Burnet theorized the elimination of forbidden
phils, which are most numerous in EGPA. In glomerular clones, all healthy individuals seem to have numerous
lesions of ANCA-associated glomerulonephritis, at the circulating autoantibodies with beneficial homeostatic
time of biopsy, leukocytes are predominantly mono- functions.31 Thus, pathogenic autoimmunity might arise
cytes and macrophages with admixed Tcells and usually from dysregulation of homeostatic autoimmunity.
only scant neutrophils.2325 Within glomeruli in animal Healthy individuals have circulating autoantibodies
models of ANCA-associated disease, the neutrophils against MPO and PR3.3236 In asymptomatic individuals,
that are predominant during the first several days of such autoantibodies are often designated natural or

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 3


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

a b remission or from healthy individuals. Healthy indi


viduals had very low titres of MPOANCA that recog
nized only a few MPO epitopes. Some patients with
clinical and pathological features of ANCA-associated
disease who were determined to be ANCA-negative
E L
using conventional clinical assays reacted with a specific
N
MPO epitope when a highly sensitive epitope-excision
A method was used; this epitope was blocked from reacting
with ANCA IgG in serum because of competitive binding
A
by a fragment of ceruloplasmin, which is a natural inhibi-
tor of MPO.35 Modulation of epitope specificity (epitope
spreading) can be influenced by endogenous or exo
genous stimuli, causing nonpathogenic autoantibodies
c d to becomepathogenic.
The detection and characteristics of autoantibodies in
asymptomatic individuals might be a predictor of sub-
sequent development of disease. This predictive value
has been evaluated using the US Department of Defense
serum repository, which contains thousands of serum
samples taken from healthy military service members at
the time of enlistment as well as during their subsequent
time in the military.36 When compared with individuals
who did not develop GPA, a greater percentage of those
with GPA had at least one elevated PR3ANCA titre
before diagnosis (63% versus 0%) and a greater rate of
increase in titre (62% versus 0%). The rises in PR3ANCA
Figure 2 | Vascular and glomerular pathology of AAV. a | Leukocytoclastic venulitis titre typically occurred before the onset of elevations in
in the nasal septal mucosa of a patient with PR3ANCA-associated disease serum Creactive protein level or signs and symptoms of
(magnification 400, H&E staining) demonstrates features of early AAV lesions: GPA. These findings are consistent with the concept that
numerous neutrophils (arrows) are marginating along and penetrating the wall of endogenous or exogenous influences might modify quan-
avenule, and accumulating in the perivascular interstitium, with some undergoing titative or qualitative characteristics of natural ANCAs to
apoptosis and karyorrhexis (leukocytoclasia). b | Another venule from the same render them pathogenic.
specimen demonstrates neutrophils in the lumen, marginating neutrophils,
endothelial cells and apoptotic bodies derived from neutrophils (magnification
Pathogenic ANCAs
1,000, H&E staining). c | Necrotizing arteritis of a small artery in the kidney of
apatient with microscopic polyangiitis involves a circumferential zone of fibrinoid Factors that could induce the development of patho-
necrosis (arrow) and perivascular leukocytes that, at this phase, contain genic ANCAs include exposure to exogenous anti-
predominantly mononuclear leukocytes (magnification 400, H&E staining). gens that influence ANCA epitope specificity (such
d | A glomerulus from a patient with ANCA-associated glomerulonephritis shows as drugs or microbes), newly expressed or modified
segmental fibrinoid necrosis (lower arrow) and an adjacent cellular crescent ANCA autoantigens (such as alternatively spliced tran-
composed of mononuclear leukocytes and epithelial cells (upper arrow) scripts or antisense transcripts), immunogenic display
(magnification 400, H&E staining). Abbreviations: A, apoptotic bodies;
of ANCA autoantigens (for example, on apoptotic cells
AAV,ANCA-associated vasculitis; ANCA, antineutrophil cytoplasmic antibody;
E,endothelial cells; H&E, haematoxylin and eosin; L, lumen; N, marginating
or NETs), or loss of effective suppression of the ANCA
neutrophils; PR3, proteinase 3. autoimmune response (for example, because of defec-
tive Tcells, Bcells or myeloid-derived suppressor cells).
This pathogenic transformation could be multifactorial,
nonpathogenic autoantibodies. Presumably, quanti with genetic, environmental and immunological events
tative or qualitative differences determine autoantibody conspiring to break autoimmune homeostasis.
pathogenicity. Compared with pathogenic MPOANCA, Mapping of epitopes that are the targets of natural and
natural MPOANCA has lower titres, lower avidity, less pathogenic MPOANCAs has demonstrated that these
subclass diversity and less capability to activate neutro- two classes of epitopes occur in close proximity on the
phils invitro.34 In autoimmune diseases that have auto 3D structure of MPO.35 This finding suggests that epitope
antigens with multiple epitope specificities, the epitope spreading of ANCA specificity, from nonpathogenic to
specificity of natural autoantibodies can differ from that pathogenic epitopes, occurs as disease develops.
of pathogenic autoantibodies. Roth etal.35 identified Modulation of existing epitope specificities is known
more than 20 MPO epitopes that can be recognized by to occur during immune responses to infectious patho-
MPOANCA in patients with ANCA-associated dis gens, and several microorganisms have been implicated
ease. MPOANCA IgG from patients with active disease in the induction and modulation of ANCA-associated
recognized the largest number of epitopes, including disease, such as Staphylococcus aureus 37,38 and Ross
epitopes that were never recognized by MPOANCA River virus.39 A novel model for induction of the ANCA
IgG from patients with ANCA-associated disease in autoimmune response theorizes that the initial immune

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

a b peptides have structures and physicochemical character-


istics that give them affinity to bind together. A classic
complementary peptide pair comprises the sense and
antisense transcripts of a gene; however, peptide mimics
of an antisense peptide are similarly complementary
to the sense peptide. Codon-directed amino acids in
sense peptides have specific pairwise interactions with
the corresponding complementary amino acids in
complementary peptides.42
An immune response to one complementary protein
can result in the production of an immune response to
the paired complementary protein; thus, immunization
with an antisense complementary peptide can induce
Figure 3 | Pathology of ANCA-associated necrotizing granulomatosis. a | The edge an antibody response to the sense peptide.42,43 Patients
of an early acute pulmonary lesion in a patient with PR3ANCA-positive GPA is positive for PR3ANCA have circulating antibodies
characterized by a central zone of necrosis containing numerous neutrophils in reactive not only to PR3 sense peptides but also to PR3
varying stages of leukocytoclasia, and by an adjacent band of monocytes and complementary antisense peptides.40,41 In addition to
macrophages (arrows) (magnification 400, H&E staining). b | A later stage of antibodies that recognize complementary PR3 antigens,
granulomatous inflammation in the same pulmonary lesion specimen demonstrates
Yang etal.44 detected anticomplementary PR3-specific
a central zone of amorphous necrotic debris containing a few pyknotic leukocytes,
and an adjacent marginal zone of epithelioid macrophages (arrows) (magnification
memory Tcellsdirected against a PR3 complementary
400, H&E staining). Abbreviations: ANCA, antineutrophil cytoplasmic antibody; peptidein patients with PR3ANCA. The stimulus for
GPA, granulomatosis with polyangiitis; H&E, haematoxylin and eosin; PR3, the antibodies to complementary PR3 could have been
proteinase 3. endogenous PR3 antisense peptides45 or exogenous pep-
tides that mimic the antisense peptides, such as peptides
brought in by commensal or pathogenic microbes.40
Endogenous source
Sense
Interestingly, the bioinformatics software tool BLAST
ANCA antigen
transcription peptide (basic local alignment search tool) identifies multiple
microbial peptide homologues of complementary PR3,
Sense DNA Idiotypic Idiotope
strand antibody mimicking the including peptides from microbes that are known to be
autoantigen associated with PR3ANCAs such as Ross River virus
Neutrophil and S.aureus.40,41
A genome-wide association study has indicated that
Antisense
different HLA specificities correlate with MPOANCA
Antisense
DNA strand
peptide
Anti-idiotypic and PR3ANCA, and a gene encoding PR3 was associ-
antibody
(autoantibody) ated with PR3ANCA but not MPOANCA. 7 These
Antisense
transcription findings suggest that antigen-recognition capabilities
are important in ANCA-associated disease, and that the
Exogenous source initial antigen recognized by antigen-presenting cells is
different in PR3ANCA-associated disease as opposed
T cell to MPOANCA-associated disease. HLA specificities
seemed to be involved in the ability of ANCA to recog
nize both sense and antisense antigen peptides. 46 The
HLA DRB1*15 allele is over-represented in African
American patients with PR3ANCA-associated disease
Mimic of (odds ratio 73.3).46 The DRB1*1501 protein binds with
Microbes antisense peptide
high affinity to both sense-PR3 peptide as well as the
Figure 4 | Diagram of the induction of an ANCA-mediated autoimmune response by complementary antisense-PR3 peptide.46 This finding
an initial immune response to a peptide that is complementary to an autoantigen suggests that one factor in the induction of ANCA-
peptide. This complementary peptide immunogen could arise from antisense associated disease could be the presence of HLA antigen-
transcription of the antisense strand of the autoantigen gene, or could be a mimic binding sites that recognize the autoantigen and a
of an antisense peptide that is produced by a symbiotic or pathogenic microbe.
complementary protein.
The anticomplementary peptide antibody idiotopes would engender an anti-
idiotypic antibody response that crossreacts with the autoantigen epitopes that Thus, the theory of autoantigen complementarity sug-
are complementary to the initial immunogenic peptide. Abbreviation: ANCA, gests that a patient with a genetically determined pre-
antineutrophil cytoplasmic antibody. disposing antigen-recognition capability could produce
pathogenic ANCA as a result of an initial appropriate
response is not against the autoantigen but rather a immune response against a microbial mimic of a com-
peptide that has a complementary structure relative to plementary peptide, or against an endogenously pro-
the autoantigen, and that the anti-idiotypic response duced antisense transcript, that would be transformed
to this initial immune response results in antibodies that into an autoimmune response by an anti-idiotypic res
recognize the autoantigen (Figure4).40,41 Complementary ponse (Figure4). In support of this theory, anti-idiotypic

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 5


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

antibodies raised against autoantibodies have been effector CD4 + Tcell subset in patients with active
shown in multiple animal models to induce anti-anti- ANCA-associated disease, in comparison with patients
idiotypic antibodies that possess characteristics of the in remission and with healthy individuals. Although
initial a utoantibodies and cause autoimmune disease.47 circulating TREG cell numbers were increased in active
Induction of pathogenic ANCAs by microbial molecu- disease, these cells had decreased suppressive function
lar mimicry of autoantigens has been proposed as a cause because of expression of a FOXP3 isoform that lacks
for ANCAs with specificity for LAMP2.10 The human exon2.54 A study by Wilde etal.55 showed that increased
LAMP2 epitope has complete homology with the bacte- expression of the negative co-stimulator programmed
rial adhesin FimH, and human LAMP2-ANCA cross- cell death protein1 (PD1) on circulating Tcells in
reacts with this microbial protein. Kain etal.10 reported patients with GPA might oppose Tcell activation; how
that rats immunized with FimH develop antibodies to ever, Tcells in renal lesions in these patients mostly
rat and human LAMP2, and develop pauci-immune lacked PD1 expression.
necrotizing glomerulonephritis that resembles human B-cell regulation might also influence production of
ANCA-associated glomerulonephritis. This study pro- pathogenic ANCAs.56,57 Bcells that have high expression
poses that infection with fimbriated bacteria can cause of CD5 and produce IL10 have regulatory capabilities.
an immune response against FimH that crossreacts with Patients with active ANCA-associated disease have a
human LAMP2 and mediates ANCA-associated disease. reduced percentage of circulating CD5+ Bcells whereas
However, Roth etal.11 were not able to reproduce the most patients in remission have a normal percentage.52
findings from this e xperimental animalmodel. In addition, the degree of normalization of circulating
Another exogenous stimulus that can induce a CD5+ Bcells after therapy with rituximab correlates with
pathogenic ANCA immune response is drugs, includ- more-persistent remission.56
ing hydralazine, minocycline, propylthiouracil and ANCA-activated neutrophils might augment the
levamisole-adulterated cocaine.8,9 The mechanisms by production of ANCAs by stimulating Bcells. Acti
which drugs induce ANCAs could shed light on patho vated neutrophils release ligands for Bcell-activating
genic mechanisms that do not involve drugs. Hydralazine- factor (BAFF; also known as TNF ligand superfamily
induced ANCA-associated disease is accompanied by member13B or B lymphocyte stimulator), which stimu-
dysregulation of ANCA-antigen expression by neutro- lates Bcell proliferation and inhibits apoptosis.58 Patients
phils.8 Hydralazine is a DNA methylation inhibitor that with ANCA-associated disease have serum levels of
might reverse epigenetic silencing of PR3 and MPO, BAFF that are elevated during active disease and decline
resulting in increased expression of both autoantigens in during remission.5962
neutrophils. This increased autoantigen expression Thus, Bcell-stimulating factors released by ANCA-
in drug-induced disease is similar to a phenomenon activated neutrophils along with permissive Tcell and
observed in patients with ANCA-associated disease who Bcell regulation seem to facilitate the production of
have abnormal epigenetically deregulated overexpres- pathogenic ANCAs.
sion of MPO and PR3 in neutrophils.48,49 Increased auto
antigen expression in neutrophils could either facilitate a Pathogenesis of vascular inflammation
loss of tolerance to these proteins, or enhance the activa- Neutrophil and monocyte activation by ANCAs
tion of neutrophils by ANCAs, or both. Organized lym- Invitro studies have shown that both MPOANCA and
phoid configuration in areas of granulomatosis that bring PR3ANCA IgG can activate neutrophils that have been
together antigen-presenting cells with Tcells and Bcells,30 primed by proinflammatory stimuli, such as TNF,63 bac-
as well as effective presentation of neutrophil antigens on terial lipopolysaccharide64 or the complement anaphyla-
apoptotic neutrophils50,51 and NETs,27 could also augment toxin C5a.65 Priming causes neutrophils to release ANCA
ANCA autoimmune responses. target antigens (for example, MPO and PR3) at the cell
surface and into the microenvironment that can then
Regulation of the ANCA immune response interact with ANCAs. Full neutrophil activation results
The pathogenic ANCA autoimmune response seems to from engagement of constitutively expressed Fc recep-
be enabled by impaired Tcell and Bcell suppression, tors (FcRs) by immune complexes containing ANCA
and possibly by enhanced Bcell stimulation by ANCA- and antigen66,67 and from binding of F(ab')2 fragments of
activated neutrophils. Patients with ANCA-associated ANCA to antigen expressed on neutrophil surfaces.68,69
disease have dysfunction of regulatory T (TREG) cells that Phosphorylation of p38 mitogen-activated protein kinase
could contribute to loss of tolerance and emergence of (p38 MAPK) and the extracellular signal-regulated
a pathogenic ANCA response.5254 TREG cells are charac- kinase (ERK) are involved in neutrophil priming and
terized by high expression of CD25 and FOXP3. Even activation, and blockade of MAPK prevents this priming
though patients with active GPA have increased numbers and activation.7072
of CD4+ CD25+ Tcells, the numbers of FOXP3+ cells are ANCA-activated neutrophils generate a respiratory
decreased. 52 The suppressive function of TREG cells burst with production of toxic oxygen radicals, release
from patients with ANCA-associated disease has been destructive enzymes through degranulation, and extrude
reported to be markedly decreased in active disease and NETs that have proinflammatory properties. 27,28,6374
more normal during remission.53 Free etal.54 described These ANCA-activated cells have been shown to cause
an increase in a proinflammatory suppression-resistant injury to and death of cultured monolayers of endothelial

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Unprimed Priming after the first day or two, and there is evidence for sys-
neutrophil
temic activation of monocytes in patients with active
ANCA-associateddisease.82

Alternative
Induction of necrotizing vascular inflammation
pathway Pathogenic mechanisms of ANCA-induced vascular
activation
inflammation must be in accord with observed lesions
Activation in tissue specimens from patients with ANCA-associated
disease or in animal models. Earlier in this Review, we
Acute Chronic inflammation
inflammation and scarring described the pathological features of AAV. The histo-
pathology of the earliest acute vascular lesions reveals
a very destructive localized neutrophil-rich necrotizing
inflammation that quickly transforms into monocyte/
Epithelial cell macrophage-rich inflammation. Immunohistology
reveals a paucity, but usually not an absence, of immuno
globulin at sites of inflammation. Small amounts of
ANCA and ANCA-antigen are found at sites of vascular
NETosis inflammation, including glomerulonephritis, in patients
biopsies15,26,83 and in experimental animal models of
C5a ANCA antigen Neutrophil
C5a receptor ANCA-associated disease.84 Invitro studies have dem-
ANCA Apoptotic body Fibroblast
Cytokine onstrated that ANCA-induced neutrophil activation
Fc receptor Macrophage Fibrin
Cytokine releases into the microenvironment ANCA-antigens that
receptor T cell Monocyte Collagen can bind to nearby structures to act as targets for insitu
Figure 5 | Putative sequence of pathogenic events in ANCA-mediated vasculitis. immune-complex formation. 76 However, the amount
Circulating neutrophils are primed for activation by ANCA by inflammatory cytokines of immunoglobulin deposited at sites of inflammation in
or C5a derived from complement activation (note that monocytes can be similarly ANCA-associated disease typically is far less than is seen
primed and activated but are not illustrated). Primed neutrophils release ANCA with conventional forms of immune-complex-mediated
antigens at the cell surface and into the microenvironment, where they interact
vascular inflammation. 1 This observation supports
with ANCA. Fc receptor engagement by ANCA bound to ANCA antigens as well as
F(ab')2 binding to ANCA antigens on neutrophil surfaces cause neutrophil
the concept that the vascular inflammation of ANCA-
activation. ANCA-activated neutrophils release factors that activate the alternative associated disease is caused primarily by direct activation
complement pathway, generating C5a. C5a and ANCA create an inflammatory of neutrophils and monocytes by ANCA, with somewhat
amplification loop, with C5a attracting and priming more neutrophils for activation incidental deposition of immune complexes containing
by ANCA, which causes, in turn, further activation of the alternative complement ANCA and ANCA antigen at the sites of injury.
pathway and production of more C5a. ANCA-activated neutrophils marginate and The most compelling evidence that ANCAs are
penetrate vessel walls and undergo respiratory burst, degranulation, NETosis, pathogenic and the most informative experiments that
apoptosis and necrosis. Disruption of endothelium allows plasma to spill into
elucidate the underlying pathogenic mechanisms come
vascular and perivascular tissue where activation of the coagulation cascade
produces the fibrin strands of fibroid necrosis. The innate inflammatory response from animal models of ANCA-associated disease.8587
orchestrates the conversion of the initial acute neutrophil-rich inflammation into Multiple convincing models of MPOANCA-associated
mononuclear leukocyte-rich inflammation and subsequent collagen deposition disease have been described and confirmed.8487 How
and fibrosis. Abbreviation: ANCA, antineutrophil cytoplasmic antibody. ever, no convincing models of PR3-ANCA-associated
disease have been widely accepted, although several
cells. 7578 Release of NETs (a process referred to as have beenproposed.8891
NETosis) by ANCA-activated neutrophils27,28 contributes Mouse models of AAV and ANCA-associated glo-
to endothelial injury and death.79 merulonephritis induced by anti-MPO antibodies have
In considering pathogenic pathways in ANCA- been used most extensively to elucidate pathogenic
associated disease, it is important to note that monocytes mechanisms. 64,65,71,84,9298 Disease can be induced by
as well as neutrophils contain ANCA antigens (including injecting mouse anti-MPO IgG into immunocompetent
PR3 and MPO) and can be activated invitro by engage- or immunodeficient mice,84 injecting splenocytes con-
ment of FcRs by ANCA IgG. 80,81 ANCA-activated taining anti-MPO Bcells into immunodeficient mice,84
monocytes release proinflammatory cytokines, includ- or transplanting bone marrow that contains MPO-
ing monocyte chemoattractant protein1 (MCP1, also positive myeloid cells into MPO-knockout mice.93 In
known as CCL2)80 and IL8.81 IL8 attracts and activates these models, a pathogenic level of anti-MPO antibodies
neutrophils and thus could amplify neutrophil-mediated induces necrotizing and crescentic glomerulonephritis
injury. MCP1 attracts monocytes and macrophages and in all mice of susceptible strains, and systemic necrotiz-
could participate in the transition of ANCA-induced ing small-vessel vasculitis and granulomatous inflam-
vascular injury from predominantly neutrophil-rich mation in some, but not all, animals. The inflammatory
inflammation to predominantly monocyte and/or lesions in these mouse models closely mimic human
macrophage-rich inflammation, including granulo ANCA-associated disease.
matous inflammation. Monocytes are conspicuous Several conclusions can be drawn from experiments
at sites of ANCA vascular inflammation, 24 especially using mouse models of MPOANCA-associated disease:

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 7


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Priming identify supportive evidence in human disease. Exami


nation of glomerular and vascular inflammatory lesions
in biopsy specimens from patients with ANCA-associated
Unprimed disease revealed markers of alternative complement path
neutrophils way activation including factorB and properdin, as well
Activation as factors shared by the classical and alternative path
ways including C3d and membrane attack complex, but
the absence of components of the classical and lectin
pathways such as C4d and mannose-binding lectin.99
Although not yet substantiated by other investigators,
one research group has reported evidence of alternative
complement pathway activation in patients with active
Granulomatosis ANCA-associated disease, including elevated plasma
levels of C3a, C5a, soluble C5b9, and Bb; reduced levels
of properdin; and normal levels of C4d.100,101
On the basis of observations in experimental animal
models and supportive findings in patients, a pathogenic
Microabscess scenario can be proposed for the interaction of patho-
formation genic ANCAs with neutrophils that have been primed
by a synergistic proinflammatory condition (Figure5).
Circulating neutrophils are primed by, for example,
inflammatory cytokines or C5a derived from com-
plement activation. Primed neutrophils release small
amounts of ANCA antigens at the cell surface and in
the microenvironment, where these antigens can inter-
act with ANCAs. FcR engagement by ANCAs bound
to ANCA antigens as well as F(ab')2 binding to ANCA
C5a ANCA antigen Neutrophil antigens expressed on neutrophil surfaces cause neu-
C5a receptor ANCA
Multinucleated
trophil activation. ANCA-activated neutrophils release
Cytokine Macrophage
Cytokine
Fc receptor giant cell factors that activate the alternative complement pathway
receptor T cell Monocyte Fibrin with generation of C5a. Neutrophils, C5a and ANCAs
Figure 6 | Putative sequence of pathogenic events in ANCA-mediated necrotizing create an inflammatory amplification loop, with C5a
granulomatosis. An inflammatory prodrome, such as an infectious or allergic attracting and priming more neutrophils for activation
inflammatory respiratory tract disease, positions increased numbers of primed by ANCAs, which in turn causes further activation of
neutrophils in extravascular interstitial tissue. ANCA immunoglobulin in the the alternative complement pathway and consequent
interstitial fluid would activate primed neutrophils and initiate an inflammatory production of more C5a. ANCA-activated neutrophils
amplification loop that would attract and activate more neutrophils, resulting in the marginate and penetrate vessel walls, and undergo res-
formation of a necrotizing microabscess. This acute inflammation and necrosis
piratory burst, degranulation, NETosis, apoptosis and
would initiate an innate inflammatory response that would wall off the necrotic zone
with granulomatous inflammation containing a predominance of monocytes and
necrosis. Disruption of endothelium enables plasma to
macrophages with admixed lymphocytes. Abbreviation: ANCA, antineutrophil spill into vascular and perivascular tissue where acti-
cytoplasmic antibody. vation of the coagulation cascade produces the fibrin
strands of fibroid necrosis. Modulated by the severity and
antibody to MPO alone (in the absence of functional reversibility of the acute injury, the innate inflammatory
Tcells) is sufficient to cause acute disease; 84 bone- response orchestrates the conversion of the acute inflam-
marrow-derived cells are sufficient and necessary to mation into m ononuclear-leukocyte-rich inflammation
induce disease;92 neutrophils are required;92 genetic back- and subsequent fibrosis.
ground determines the susceptibility to and severity of
disease;94 circulating proinflammatory factors exacerbate Pathogenesis of granulomatosis
disease;64 and activation of the alternative complement There has been substantial controversy over the nature
pathway is required to induce vascular inflammation.96 of the granulomatosis seen in GPA and EGPA. The term
Studies of these mouse models also showed that disease granulomatosis has been used to describe the multiple
can be prevented or ameliorated by hydrolysis of anti- foci of extravascular inflammation that were described
MPO IgG glycans, which affects FcRantibody inter- initially by Klinger 102 and Wegener 103,104 in GPA, and
actions;95 by inhibition of p38 MAPK, which prevents by Churg and Strauss in EGPA.105 Controversy over the
priming of neutrophils by inflammatory stimuli;71 and pathogenesis of this granulomatous inflammation derives
by blockade of C597 or C5a receptor,65,98 which prevents from the fact that it can arise from at least two distinct
activation of the alternative complement pathway. pathwaysone initiated by a Tcell mediated (typeIV)
Evidence from animal models of the involvement of immune response specific for PR3 or MPO, and another
alternative complement pathway activation in the patho- by an antigen-independent innate response of macro
genesis of ANCA-associated disease prompted studies to phages to something in the tissue that needs to be

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

walledoff.106 The latter pathway is an exaggerated version patient with circulating ANCAs. ANCA-induced neutro-
of the innate monocyte and macrophage response that phil activation, possibly fuelled by activation of the alter-
closely follows any acute inflammatory event. Although native complement pathway, would induce microabscess
many investigators have proposed that ANCA-associated formation that, in turn, would engender a granulomatous
granulomatosis is caused by an antigen-specific typeIV reaction to wall off the zone of necrosis (Figure6).
adaptive immune response, we favour the theory that
the granulomatosis is an innate inflammatory response Conclusions
to acute extravascular inflammation that is initiated by Pauci-immune necrotizing small-vessel vasculitis and
ANCA-induced neutrophil activation. extravascular granulomatosis are associated with, and
The concept that the granulomatosis of GPA is a might be caused by, ANCAs. The distinctive, extremely
response to acute neutrophil-mediated necrosis is not destructive necrotizing vascular inflammation of AAV
new. For example, based on careful histopathological seems to be fuelled by an amplification loop in which
examination of lesions in patients with GPA and EGPA, ANCA-activated neutrophils activate the alterna-
Fienberg and his associates concluded that micro tive complement pathway, which, in turn, leads to the
necrosis, usually with neutrophils (microabscesses), attraction and activation of more neutrophils. Promising
constitutes the early phase in the development of the therapies are those that can target one or multiple com-
pathognomonic organized palisading granuloma. 21 ponents of this vicious cycle (for example ANCAs,
Lungs of patients with active GPA and EGPA often have ANCA antigens, neutrophil activation and complement
a range of granulomatous lesions, from acute lesions with activation). The pathogenesis of ANCA-associated
central microabscesses (Figure3a) or subacute lesions necrotizing extravascular granulomatous inflammation
with central necrosis (Figure3b) to chronic lesions with is not known, but might be fuelled by the same inflam-
a central zone of scarring and chronic inflammation.20 matory amplification loop occurring in the extravas-
Chronic lesions contain substantial accumulations of cular compartment, involving primed neutrophils and
lymphocytes and dendritic cells, including Tcells and interstitial ANCAs.
Bcells with specificity for ANCA antigens that could be
a source for autoantibody production.108
On the basis of this apparent sequence of pathologi- Review criteria
cal events, and the knowledge that ANCA IgG activates This Review is based primarily on full-text, peer-reviewed,
primed neutrophils, even invitro, a reasonable hypothesis English-language medical literature from the 1980s into
is that if an inflammatory condition (perhaps infection in 2014 reporting on vasculitis and glomerulonephritis
GPA and allergy in EGPA) positions primed neutrophils caused by antineutrophil cytoplasmic autoantibodies,
in the extravascular tissue (for example, the submucosa as well as the authors knowledge of the field acquired
over more than 30years of involvement in research on
of the respiratory tract), these primed neutrophils would
glomerulonephritis and vasculitis.
be activated by ANCA IgG in the interstitial fluid of a

1. Jennette, J.C. etal. Revised International 8. Pendergraft, W.F. 3rd & Niles, J.L. Trojan (ANCAs) in patients with inflammatory bowel
Chapel Hill Consensus Conference horses: drug culprits associated with disease show no correlation with proteinase 3,
nomenclature of vasculitides. Arthritis Rheum. antineutrophil cytoplasmic autoantibody (ANCA) lactoferrin, myeloperoxidase, elastase,
65, 111 (2013). vasculitis. Curr. Opin. Rheumatol. 26, 4249 cathepsin G and lysozyme: a Singapore study.
2. Falk, R.J. & Jennette, J.C. Anti-neutrophil (2014). Ann. Acad. Med. Singapore 29, 704707 (2000).
cytoplasmic autoantibodies with specificity for 9. Graf, J. Rheumatic manifestations of cocaine 16. Mahler, M. etal. PR3-ANCA: a promising
myeloperoxidase in patients with systemic use. Curr. Opin. Rheumatol. 25, 5055 (2013). biomarker for ulcerative colitis with extensive
vasculitis and idiopathic necrotizing and 10. Kain, R. etal. Molecular mimicry in pauci- disease. Clin. Chim. Acta. 424, 267273 (2013).
crescentic glomerulonephritis. N.Engl. J.Med. immune focal necrotizing glomerulonephritis. 17. Hervier, B. et al. Systemic lupus erythematosus
318, 16511657 (1988). Nat. Med. 14, 10881096 (2008). associated with ANCA-associated vasculitis:
3. Goldschmeding, R. etal. Wegeners 11. Roth, A.J. etal. AntiLAMP2 antibodies are not anoverlapping syndrome? Rheumatol. Int. 32,
granulomatosis autoantibodies identify a novel prevalent in patients with antineutrophil 32853290 (2012).
diisopropylfluorophosphate-binding protein in cytoplasmic autoantibody glomerulonephritis. 18. Levy, J.B., Hammad, T., Coulthart, A., Dougan, T.
the lysosomes of normal human neutrophils. J.Am. Soc. Nephrol. 23, 545555 (2012). & Pusey, C.D. Clinical features and outcome of
J.Clin. Invest. 4, 15771579 (1988). 12. Kawakami, T., Ishizu, A., Arimura,Y. & Soma, Y. patients with both ANCA and anti-GBM
4. Niles, J.L., McCluskey, T., Ahmad, M.F. & Serum antilysosomalassociated membrane antibodies. Kidney Int. 66, 15351540 (2004).
AminArnaout, M.A. Wegeners granulomatosis protein2 antibody levels in cutaneous 19. Jennette, J.C. & Thomas, D.B. in Heptinstalls
autoantigen is a novel neutrophil serine polyarteritis nodosa. Acta Derm. Venereol. Pathology of the Kidney 6th edn (eds
proteinase. Blood 74, 18881893 (1989). 93, 7073 (2013). Jennette,J.C. etal.) 643674 (Lippincott
5. Jennette, J.C., Hoidal, J.H. & Falk, R.J. 13. Kawakami, T., Takeuchi, S., Arimura, Y. & Williams & Wilkins, 2007).
Specificity of anti-neutrophil cytoplasmic Soma,Y. Elevated antilysosomal-associated 20. Jennette, J.C. Nomenclature and classification
autoantibodies for proteinase 3. Blood 75, membrane protein2 antibody levels in patients of vasculitis: lessons learned from
22632264 (1990). with adult Henoch-Schnlein purpura. Br. J. granulomatosis with polyangiitis (Wegners
6. Lionaki, S. et al. Classification of antineutrophil Dermatol. 166, 12061212 (2012). granulomatosis). Clin. Exp. Immunol.
cytoplasmic autoantibody vasculitides: the role 14. Roozendaal, C. etal. Does analysis of the 164(Suppl.1), 710 (2011).
of antineutrophil cytoplasmic autoantibody antigenic specificities of anti-neutrophil 21. Mark, E.J., Matsubara, O., Tan-Liu, N.S.
specificity for myeloperoxidase or proteinase 3 cytoplasmic antibodies contribute to their & Fienberg, R. The pulmonary biopsy
in disease recognition and prognosis. Arthritis clinical significance in the inflammatory bowel in the early diagnosis of Wegeners (pathergic)
Rheum. 64, 34523462 (2012). diseases? Scand. J. Gastroenterol. 34, granulomatosis: a study based on 35 open lung
7. Lyons, P.A. etal. Genetically distinct subsets 11231131 (1999). biopsies. Hum. Pathol. 19, 10651071 (1988).
within ANCA-associated vasculitis. N.Engl. 15. Ooi, C.J., Lim, B.L., Cheong, W.K., Ling, A.E. & 22. Jennette, J.C., Xiao, H., Falk, R. & Gasim, A.M.
J.Med. 367, 214223 (2012). Ng, H.S. Antineutrophil cytoplasmic antibodies Experimental models of vasculitis and

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 9


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

glomerulonephritis induced by antineutrophil with nasal polyps. Clin. Exp. Rheumatol. 55. Wilde, B et al. Aberrant expression of the
cytoplasmic autoantibodies. Contrib. Nephrol. 28(Suppl. 57), 5155 (2010). negative costimulator PD1 on Tcells in
169, 211220 (2011). 39. Davies, D.J., Moran, J.E., Niall, J. F. granulomatosis with polyangiitis. Rheumatology
23. Rastaldi, M.P. et al. Glomerular monocyte- & Ryan,G.B. Segmental necrotizing (Oxford) 7, 11881197 (2012).
macrophage features in ANCA-positive renal glomerulonephritis with antineutrophil antibody: 56. Bunch, D.O. etal. Decreased CD5+ Bcells
vasculitis and cryoglobulinemic nephritis. J. Am. possible arbovirus aetiology? Br. Med. J. (Clin. in active ANCA vasculitis and relapse after
Soc. Nephrol. 11, 20362043 (2000). Res. Ed.) 285, 606 (1982). rituximab. Clin. J.Am. Soc. Nephrol. 8, 382391
24. Weidner, S., Carl, M., Riess, R. & 40. Pendergraft, W.F. etal. Autoimmunity is triggered (2013).
Rupprecht,H.D. Histologic analysis of renal by cPR-3(105201), aprotein complementary to 57. Wilde B. etal. Regulatory Bcells in ANCA-
leukocyte infiltration in antineutrophil the autoantigen proteinase 3. Nat. Med. 10, associated vasculitis. Ann. Rheum. Dis. 72,
cytoplasmic antibody-associated vasculitis: 7279 (2004). 14161419 (2013).
importance of monocyte and neutrophil 41. Preston, G.A., Pendergraft, W.F. 3rd & Falk, R.J. 58. Scapini, P., Bazzoni, F. & Cassatella, M.A.
infiltration in tissue damage. Arthritis Rheum. New insights that link microbes with the Regulation of Bcellactivating factor
50, 36513657 (2004). generation of antineutrophil cytoplasmic (BAFF)/Blymphocyte stimulator (BLyS)
25. Cunningham, M.A. etal. Prominence of cell- autoantibodies: the theory of autoantigen expression in human neutrophils. Immunol.
mediated immunity effectors in pauci-immune complementarity. Curr. Opin. Nephrol. Hypertens. Letters 116, 16 (2008).
glomerulonephritis. J. Am. Soc. Nephrol. 10, 14, 217222 (2005). 59. Krumbholz, M. etal. BAFF is elevated in serum
499506 (1999). 42. Heal, J.R., Roberts, G.W., Raynes, J.G., of patients with Wegeners granulomatosis.
26. Bajema, I.M., Hagen, E.C., de Heer, E., van der Bhakoo, A. & Miller, A.D. Specific interactions J.Autoimmun. 25, 298302 (2005).
Woude, F.J. & Bruijn, J.A. Colocalization of between sense and complementary peptides: 60. Sanders, J.S., Huitma, M.G., Kallenberg, C.G.
ANCA-antigens and fibrinoid necrosis in ANCA- the basis for the proteomic code. Chembiochem. & Stegeman, C.A. Plasma levels of soluble
associated vasculitis. Kidney Int.60, 3, 136151 (2002). interleukin 2 receptor, soluble CD30, interleukin
20252030 (2001). 43. Mekler, L.B. On the specific mutual interaction 10 and Bcell activator of the tumour necrosis
27. Kessenbrock, K. etal. Netting neutrophils in of amino acid residues of polypeptide chains factor family during follow-up in vasculitis
autoimmune small-vessel vasculitis. Nat. Med. and amino acid residues with codons. Oncology associated with proteinase 3antineutrophil
15, 623625 (2009). 27, 286288 (1973). cytoplasmic antibodies: associations with
28. Abreu-Velez, A.M., Smith, J.G. Jr & Howard,M.S. 44. Yang, J.J. etal. ANCA patients have Tcells disease activity and relapse. Ann. Rheum. Dis.
Presence of neutrophil extracellular traps and responsive to complementary PR3 autoantigen. 65, 14841489 (2006).
antineutrophil cytoplasmic antibodies associated Kidney Int. 74, 11591169 (2008). 61. Nagai, M. etal. Serum levels of BAFF and APRIL
with vasculitides. North Am. J. Med. Sci. 1, 45. Badhwar, A. K., Simmons, J., Jennette, J. C., in myeloperoxidase anti-neutrophil cytoplasmic
309313 (2009). Falk,R. J. & Preston, G. A. Characterization of autoantibody-associated renal vasculitis:
29. Kambas, K. etal. Tissue factor expression in an antisense transcript from the proteinase3 association with disease activity. Nephron Clin.
neutrophil extracellular traps and neutrophil gene locus in patients with ANCA-vasculitis Pract. 118, c339c345 (2011).
derived microparticles in antineutrophil utilizing strand-specific RTPCR and 5'RACE 62. Bader, L., Koldingsnes, W. & Nossent, J.
cytoplasmic antibody associated vasculitis may [abstract]. J.Am. Soc. Nephrol. 19, 188A Blymphocyte activating factor levels are
promote thromboinflammation and the (2008). increased in patients with Wegeners
thrombophilic state associated with the disease. 46. Cao, Y. etal. DRB1*15 allele is a risk factor for granulomatosis and inversely correlated with
Ann. Rheum. Dis. http://dx.doi.org/10.1136/ PR3-ANCA disease in African Americans. J. Am. ANCA titer. Clin. Rheumatol. 29, 10311035
annrheumdis-2013-203430 (2013). Soc. Nephrol. 22, 11611167 (2011). (2010).
30. Lamprecht, P. & Gross, W.L. Current knowledge 47. Shoenfeld, Y. Idiotypic induction of 63. Falk, R.J., Terrell, R.S., Charles, L.A. &
on cellular interactions in the WGgranuloma. autoimmunity: a new aspect of the idiotypic Jennette, J.C. Anti-neutrophil cytoplasmic
Clin. Exp. Rheumatol. 25 (Suppl. 44), S49S51 network. FASEB J. 8, 12961301 (1994). autoantibodies induce neutrophils to
(2007). 48. Yang, J.J. etal. Circumvention of normal degranulate and produce oxygen radicals
31. Jennette, J.C. & Falk, R.J. The rise and fall of constraints on granule protein gene expression invitro. Proc. Natl Acad. Sci. USA 87,
horror autotoxicus and forbidden clones. Kidney in peripheral blood neutrophils and monocytes 41154119 (1990).
Int. 78, 533535 (2010). of patients with antineutrophil cytoplasmic 64. Huugen, D. etal. Aggravation of anti-
32. Cui, Z., Zhao, M.H., Segelmark, M. & autoantibody-associated glomerulonephritis. myeloperoxidase antibody induced
Hellmark,T. Natural autoantibodies to J.Am. Soc. Nephrol. 15, 21032114 (2004). glomerulonephritis by bacterial
myeloperoxidase, proteinase 3, and the 49. Ciavatta, D.J. etal. Epigenetic basis for aberrant lipopolysaccharide: role of tumor necrosis
glomerular basement membrane are present upregulation of autoantigen genes in humans factor . Am. J.Pathol. 167, 4758 (2005).
in normal individuals. Kidney Int. 78, 590597 with ANCA vasculitis. J.Clin. Invest. 120, 65. Schreiber, A. etal. C5a receptor mediates
(2010). 32093219 (2010). neutrophil activation and ANCA-induced
33. Olson, S.W. et al. Asymptomatic autoantibodies 50. Durant, S. etal. Apoptosis-induced proteinase 3 glomerulonephritis. J.Am. Soc. Nephrol. 20,
associate with future anti-glomerular basement membrane expression is independent from 289298 (2009).
membrane disease. J. Am. Soc. Nephrol. 22, degranulation. J.Leukoc. Biol. 75, 8798 66. Porges, A.J. etal. Anti-neutrophil cytoplasmic
19461952 (2011). (2004). antibodies engage and activate human
34. Xu, P.C., Cui, Z., Chen, M., Hellmark, T. & 51. Kantari, C. etal. Proteinase 3, the Wegener neutrophils via Fc RIIa. J.Immunol. 153,
Zhao,M.H. Comparison of characteristics of autoantigen, is externalized during neutrophil 12711280 (1994).
natural autoantibodies against myeloperoxidase apoptosis: evidence for a functional association 67. Kocher, M., Siegel, M.E., Edberg, J.C. &
and anti-myeloperoxidase autoantibodies from with phospholipid scramblase 1 and interference Kimberly, R.P. Cross-linking of Fc receptor IIa
patients with microscopic polyangiitis. with macrophage phagocytosis. Blood 110, and Fc receptor IIIb induces different
Rheumatology (Oxford) 50, 12361243 (2011). 40864095 (2007). proadhesive phenotypes on human neutrophils.
35. Roth, A.J. etal. ANCA epitope specificity 52. Morgan, M.D. etal. Patients with Wegeners J.Immunol. 159, 39403948 (1997).
determines pathogenicity, detectability and granulomatosis demonstrate a relative 68. Kettritz, R., Jennette, J.C. & Falk, R.J. Cross-
clinical predictive value. J.Clin. Invest. 123, deficiency and functional impairment of linking of ANCA-antigens stimulates superoxide
17731783 (2013). Tregulatory cells. Immunology 130, 6473 release by human neutrophils. J.Am. Soc.
36. Olson, S.W. et al. Relation between (2010). Nephrol. 8, 386394 (1997).
asymptomatic proteinase 3 antibodies and 53. Rimbert, M. etal. Decreased numbers of blood 69. Williams, J.M. etal. Activation of the Gi
future granulomatosis with polyangiitis. Clin. J. dendritic cells and defective function of heterotrimeric G protein by ANCA IgG F(ab')2
Am. Soc. Nephrol. 8, 13121318 (2013). regulatory Tcells in antineutrophil cytoplasmic fragments is necessary but not sufficient to
37. Popa, E.R., Stegeman, C.A., Kallenberg, C.G. antibody-associated vasculitis. PLoS ONE 6, stimulate the recruitment of those downstream
&Tervaert, J.W. Staphylococcus aureus and e18734 (2011). mediators used by intact ANCA IgG.J.Am. Soc.
Wegeners granulomatosis. Arthritis Res. 4, 54. Free, M.E. etal. Patients with antineutrophil Nephrol. 14, 661669 (2003).
7779 (2002). cytoplasmic antibody-associated vasculitis have 70. Kettritz, R., Schreiber, A., Luft, F.C. & Haller, H.
38. Laudien, M. etal. Nasal carriage of defective TREG cell function exacerbated by the Role of mitogen-activated protein kinases in
Staphylococcus aureus and endonasal activity presence of a suppression-resistant effector cell activation of human neutrophils by antineutrophil
inWegeners granulomatosis as compared to population. Arthritis Rheum. 65, 19221933 cytoplasmic antibodies. J. Am. Soc. Nephrol. 12,
rheumatoid arthritis and chronic rhinosinusitis (2013). 3746 (2001).

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

71. van der Veen, B.S. etal. Effects of p38 mitogen- Wegeners granulomatosis. Ann. Rheum. Dis. 58, 96. Xiao, H., Schreiber, A., Heeringa, P., Falk, R.J. &
activated protein kinase inhibition on anti- 237245 (1999). Jennette, J.C. Alternative complement pathway
neutrophil cytoplasmic autoantibody 83. Arimura, Y. et al. The role of myeloperoxidase in the pathogenesis of disease mediated by
pathogenicity invitro and invivo. Ann. Rheum. and myeloperoxidase-antineutrophil cytoplasmic antineutrophil cytoplasmic autoantibodies.
Dis. 70, 356365 (2011). antibodies (MPO-ANCAs) in the pathogenesis Am.J.Pathol. 170, 5264 (2007).
72. Hao, J., Meng, L. Q., Xu, P.C., Chen, M. & ofhuman MPOANCAassociated 97. Huugen, D. etal. Inhibition of complement factor
Zhao,M.H. p38MAPK, ERK and PI3K signaling glomerulonephritis. Clin. Exp. Nephrol. 17, C5 protects against anti-myeloperoxidase
pathways are involved in C5a-primed neutrophils 634637 (2013). antibody-mediated glomerulonephritis in mice.
for ANCA-mediated activation. PLoS ONE 7, 84. Xiao, H. etal. Antineutrophil cytoplasmic Kidney Int. 71, 646654 (2007).
e38317 (2012). autoantibodies specific for myeloperoxidase 98. Xiao, H. etal. C5a receptor (CD88) blockade
73. Charles, L.A., Caldas, M.L., Falk, R.J., cause glomerulonephritis and vasculitis in mice. protects against MPO-ANCA glomerulonephritis.
Terrell,R.S. & Jennette, J.C. Antibodies against J.Clin. Invest. 110, 955963 (2002). J.Am. Soc. Nephrol. 25, 225231 (2014).
granule proteins activate neutrophils invitro. 85. Jennette, J.C., Xiao, H., Falk, R. & Gasim, A.M. 99. Xing, G.Q. etal. Complement activation
J.Leukoc. Biol. 50, 539546 (1991). Experimental models of vasculitis and is involved in renal damage in human
74. Csernok, E., Ernst, M., Schmitt, W., Bainton, D.F. glomerulonephritis induced by antineutrophil antineutrophil cytoplasmic autoantibody
& Gross, W.L. Activated neutrophils express cytoplasmic autoantibodies. Contrib. Nephrol. associated pauci-immune vasculitis. J.Clin.
proteinase 3 on their plasma membrane invitro 169, 211220 (2011). Immunol. 29, 282291 (2009).
and invivo. Clin. Exp. Immunol. 95, 244250 86. van Timmeren, M. M. & Heeringa, P. 100. Yuan, J. et al. C5a and its receptors in human
(1994). Pathogenesis of ANCA-associated vasculitis: anti-neutrophil cytoplasmic antibody (ANCA)-
75. Ewert, B.H., Jennette, J.C. & Falk, R.J. recent insights from animal models. Curr. Opin. associated vasculitis. Arthritis Res. Ther. 14,
Anti-myeloperoxidase antibodies stimulate Rheumatol. 24, 814 (2012). R140 (2012).
neutrophils to damage human endothelial cells. 87. Salama, A.D. & Little, M.A. Animal models of 101. Gou, S.J., Yuan, J., Chen, M., Yu, F. &
Kidney Int. 41, 375383 (1992). antineutrophil cytoplasm antibody-associated Zhao,M.H. Circulating complement activation
76. Savage, C.O., Gaskin, G., Pusey, C.D. & vasculitis. Curr. Opin. Rheumatol. 24, 17 (2012). inpatients with anti-neutrophil cytoplasmic
Pearson, J.D. Myeloperoxidase binds to 88. van der Geld, Y.M. etal. Rats and mice antibody-associated vasculitis. Kidney Int. 83,
vascular endothelial cells, is recognized by ANCA immunized with chimeric human/mouse 129137 (2012).
and can enhance complement dependent proteinase 3 produce autoantibodies to mouse 102. Klinger, H. Grenzformen der periarteriitis nodosa
cytotoxicity. Adv. Exp. Med. Biol. 336, 121123 Pr3 and rat granulocytes. Ann. Rheum. Dis. 66, [German]. Frankf. Z. Pathol. 42, 455480 (1931).
(1993). 16791682 (2007). 103. Wegener, F. ber generalisierte septische
77. Ewert, B.H., Becker, M.E., Jennette, J.C. 89. Pfister, H. etal. Antineutrophil cytoplasmic Gefsserkrankungen [German]. Verh. Deut.
& Falk,R.J. Antimyeloperoxidase antibodies autoantibodies against the murine homolog Pathol. Ges. 29, 202210 (1936).
induce neutrophil adherence to cultured human ofproteinase 3 (Wegener autoantigen) are 104. Wegener, F. ber eine eigenartige rhinogene
endothelial cells. Ren. Fail. 17, 125133 pathogenic invivo. Blood 104, 14111418 Granulomatose mit besonderer Beteiligung des
(1995). (2004). Arteriensystems und der Nieren [German]. Beitr.
78. Lu, X., Garfield, A., Rainger, G.E., Savage, C.O. 90. Primo, V.C. etal. Anti-PR3 immune responses Pathol. Anat. 102, 3068 (1939).
&Nash, G.B. Mediation of endothelial cell induce segmental and necrotizing 105. Churg, J. & Strauss, L. Allergic granulomatosis,
damage by serine proteases, but not superoxide glomerulonephritis. Clin. Exp. Immunol. 159, allergic antiitis, and periarteritis nodosa. Am. J.
released from antineutrophil cytoplasmic 327337 (2010). Pathol. 27, 277294 (1951).
antibody-stimulated neutrophils. Arthritis 91. Little, M.A. et al. Anti-proteinase 3 anti-neutrophil 106. Petersen, H.J. & Smith, A.M. The role of the
Rheum. 54, 16191628 (2006). cytoplasm autoantibodies recapitulate systemic innate immune system in granulomatous
79. Gupta, A.K. etal. Activated endothelial cells vasculitis in mice with ahumanized immune disorders. Front. Immunol. 4, 120 (2013).
induce neutrophil extracellular traps and are system. PLoS ONE 7, e28626 (2012). 107. Voswinkel, J., Mller, A. & Lamprecht, P.
susceptible to NETosis-mediated cell death. 92. Xiao, H. etal. The role of neutrophils in the Is PR3-ANCA formation initiated in Wegeners
FEBS Letters 584, 31933197 (2010). induction of glomerulonephritis by anti- granulomatosis lesions? Granulomas as
80. Casselman, B.L., Kilgore, K.S., Miller, B.F. myeloperoxidase antibodies. Am. J.Pathol. 167, potential lymphoid tissue maintaining
&Warren, J.S. Antibodies to neutrophil 3945 (2005). autoantibody production. Ann. NY Acad. Sci.
cytoplasmic antigens induce monocyte 93. Schreiber, A., Xiao, H., Falk, R.J. & 1051, 1219 (2005).
chemoattractant protein1 secretion from human Jennette,J.C. Bone marrow-derived cells are
monocytes. J. Lab. Clin. Med. 126, 495502 sufficient and necessary targets to mediate Acknowledgements
(1995). glomerulonephritis and vasculitis induced by The authors are supported by a grant from the NIH
81. Ralston, D.R., Marsh, C.B., Lowe, M.P. & anti-myeloperoxidase antibodies. J.Am. Soc. National Institute of Diabetes and Digestive and
Wewers, M.D. Antineutrophil cytoplasmic Nephrol. 17, 33553364 (2006). Kidney Diseases (P01-DK05833511).
antibodies induce monocyte IL8 release. Role 94. Xiao, H. etal. Genetically determined severity of
ofsurface proteinase3, 1-antitrypsin, and Fc anti-myeloperoxidase glomerulonephritis. Am. J. Author contributions
receptors. J. Clin. Invest. 100, 14161424 Pathol. 8, 139160 (2013). J.C.J. researched data for the article, and both
(1997). 95. van Timmeren, M.M. etal. IgG glycan hydrolysis authors contributed equally to discussions of
82. Muller Kobold, A.C., Kallenberg, C.G. & Tervaert, attenuates ANCA-mediated glomerulonephritis. content,writing and review/editing of the manuscript
J.W. Monocyte activation in patients with J.Am. Soc. Nephrol. 21, 11031114 (2010). before submission.

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 11


2014 Macmillan Publishers Limited. All rights reserved

Вам также может понравиться