SlideShare a Scribd company logo
1 of 11
Download to read offline
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  AUGUST 2014  |  463
Department of
Pathology and
Laboratory Medicine,
School of Medicine,
University of North
Carolina at Chapel Hill,
308 Brinkhous-Bullitt
Building, CB#7525,
Chapel Hill,
NC 27599-7525,
USA (J.C.J., R.J.F.).
Correspondence to:
J.C.J.
jcj@med.unc.edu
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 T‑cell and B‑cell regulation.
A putative 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. 10, 463–473 (2014); published online 8 July 2014; doi:10.1038/nrrheum.2014.103
Introduction
Antineutrophil cytoplasmic autoantibodies (ANCAs)
are associated with, and are the probable cause of, a
distinct form of vasculitis that can affect any organ in
the body. Although small vessels are the predominant
target, ANCA-associated vasculitis (AAV) can affect
many different types of vessels including ar­teries,
arterioles, capillaries, venules and veins (Figure 1).1
Immuno­pathologically, AAV has an absence or paucity
of immunoglobulin deposition in injured vessels (pauci-
immune vasculitis), compared with the more extensive
deposition of immunoglobulin in immune-complex-
mediated small-vessel vasculitis.1
AAV can be limited
to one organ at the time of presentation (for example,
lung-limited disease or renal-limited disease) or involve
multiple organs. Frequent target tissues are the upper
and lower respiratory tract, kidneys, skin and periph-
eral nerves. Onset and exacerbations induce signs and
symptoms of high levels of circulating inflammatory
cytokines, such as fever, arthralgia and myalgia. In addi-
tion to these nonspecific systemic inflammatory mani-
festations, inflammation of vessel walls causes more
specific signs and symptoms of vasculitis depending
on which vessels and which organs are affected, for
example purpura caused by dermal venulitis; pulmonary
haemorrhage caused by alveolar capillaritis; glomerulo-
nephritis caused by glomerular capillaritis; peripheral
neuropathy caused by epineural arteritis; and ocular
inflammation caused by vasculitis in small vessels in the
eye and orbit. In addition to vasculitis, some variants of
ANCA-associated disease have extravascular necrotiz-
ing granulomatous inflammation (granulomatosis)—­
seemingly not arising from vascular inflammation—that
most often affects the upper and lower respiratory tracts
but can affect any organ.
On the basis of the distribution of vascular inflam-
mation and the presence or absence of granulomatosis
and asthma, systemic AAV is categorized as microscopic
polyangiitis (MPA) if there is vasculitis but no evidence
of granulomatosis or asthma, granulomatosis with poly-
angiitis (GPA) if there is granulomatosis but no asthma,
and eosinophilic granulomatosis with poly­angiitis
(EGPA; also known as Churg–Strauss syndrome) if
there is granulomatosis, asthma and blood eosinophilia.1
Organ-limited disease, including isolated pauci-immune
necrotizing and crescentic glomerulonephritis, can be
considered limited variants of MPA. Limited variants of
GPA and EGPA can be confined to the respiratory tract.
AAV can also be classified on the basis of auto­
antigen specificity. In patients with vasculitis, the two
best-­documented autoantigen targets of ANCA are
myelo­peroxidase (MPO)2
and proteinase 3 (PR3).3–5
Classification of AAV by antigen specificity clearly
shows differences in clinical presentations and outcomes,
Competing interests
J.C.J. has acted as a consultant for Amicus Therapeutics,
Genentech, GlaxoSmithKline and Protalix BioTherapeutics, and
has undertaken research in collaboration with ChemoCentryx.
R.J.F. declares no competing interests.
FOCUS ON VASCULITIS
© 2014 Macmillan Publishers Limited. All rights reserved
464  |  AUGUST 2014  |  VOLUME 10 www.nature.com/nrrheum
organ system involvement, patterns of extravascular
inflammation, and genetic associations including HLA
associ­ations.6,7
The most informative AAV classifica-
tion scheme includes both the ANCA antigen specifi­
city and the clinicopathological phenotype, for example
MPO‑ANCA MPA or PR3‑ANCA MPA.1
MPO and PR3 are not the only autoantigens recog-
nized by ANCAs. For instance, ANCAs specific for
elastase occur, and are most often identified, in patients
with drug-induced ANCA-associated disease, including
disease induced by cocaine adulterated with leva­misole.8,9
A controversial autoantigen specificity is for lysosomal-
associated membrane protein 2 (LAMP‑2). Kain et al.10
reported finding LAMP‑2-ANCA in ~90% of patients
Key points
■■ Antineutrophil cytoplasmic autoantibodies (ANCAs) are associated with and
probably cause pauci-immune systemic necrotizing small-vessel vasculitis
and glomerulonephritis
■■ ANCAs specific for proteinase 3 or myeloperoxidase occur with organ-
limited disease, microscopic polyangiitis, granulomatosis with polyangiitis
or eosinophilic granulomatosis with polyangiitis
■■ ANCAs seem to cause vasculitis by activating circulating primed neutrophils and
causing them to attach to, penetrate and damage vessel walls by undergoing
respiratory burst, degranulation, NETosis, apoptosis and necrosis
■■ ANCA-induced neutrophil activation also releases factors that activate the
alternative complement pathway, which establishes a destructive inflammatory
amplification loop that attracts and activates more neutrophils that, in turn,
further activates the complement system
■■ ANCA-associated granulomatosis might result from the same pathogenic
sequence of events involving extravascular primed neutrophils and interstitial-
fluid ANCAs, followed by a granulomatous reaction to wall off the resulting
extravascular necrosis
with pauci-immune necrotizing glomerulonephritis,
including patients who were positive for MPO-ANCA
or PR3-ANCA and others who were negative for both.
However, Roth et al.11
were unable to confirm this high
frequency of LAMP‑2-ANCA in these patients. Similarly,
Kawakami et al. did not detect LAMP‑2-ANCA in
patients with MPA, although they did report evidence for
LAMP‑2-ANCA in some patients with cutaneous poly­
arteritis nodosa12
and IgA vasculitis (formerly known
as Henoch–Schönlein purpura).13
ANCAs also occur
in patients with inflammatory bowel disease, and are
detected by indirect immunofluorescence assay in ~60%
of patients with ulcerative colitis and ~20% of those with
Crohn disease.14–16
In this setting, ANCAs only rarely
have specificity for PR3, MPO or elastase: the major
autoantigens are catalase, α‑enolase, lacto­transferrin
and bactericidal permeability-increasing protein.14–16
ANCA, usually MPO‑ANCA, occurs in a small minor-
ity of patients with systemic lupus erythe­matosus who
seem to to have vasculitic features of ANCA-associated
disease.17
Approximately 30% of patients with anti­
glomerular basement membrane disease have ANCA,
usually MPO‑ANCA.18
An important role for ANCAs in the pathogenesis of
vasculitis and glomerulonephritis is supported by clinical
evidence and by in vitro and in vivo experimental data
(summarized in Box 1). The leading theory proposes that
circulating neutrophils and monocytes that have been
primed by inflammatory stimuli display ANCA antigens
at or near the cell surface, and that interaction of these
antigens with ANCAs results in neutrophil activation
and initiation of vascular inflammation. An extension
of this theory proposes that primed extravascular neutro-
phils interact with interstitial ANCAs, causing necrotiz-
ing inflammation and resultant reactive granulomatous
inflammation. This Review summarizes observations
that support these pathogenic theories as well as puta-
tive mechanisms for the origin of the pathogenic ANCA
autoimmune response.
Pathology of ANCA-associated disease
Any valid theories about pathogenic mechanisms in
ANCA-associated disease must explain the development
of the observed pathologic lesions. The acute and chronic
lesions described in this section provide ­guidance in
­formulating pathogenic theories.
Vasculitis and glomerulonephritis
In patients19–21
and experimental animal models,22
the
acute vascular and extravascular lesions of ANCA-
associated disease begin as neutrophil-rich necrotizing
inflammation. In vessels, the earliest lesions have mar-
gination and diapedesis of predominantly neutrophils
with admixed monocytes (Figure 2a,b). Areas of segmen-
tal vascular necrosis also develop perivascular infiltrates
that initially have numerous neutrophils.
Within hours, neutrophils undergo apoptosis and
necrosis (Figure 2a,b). The early destruction of neutro-
phils by apoptosis and necrosis during ANCA-associated
glomerulonephritis and vasculitis is supported by the
Medium-vessel vasculitis
Polyarteritis nodosa
Kawasaki disease
Immune complex small-vessel vasculitis
Cryoglobulinemic vasculitis
IgA vasculitis (Henoch–Schönlein)
Hypocomplementemic urticarial vasculitis
(Anti-C1q vasculitis)
ANCA-associated small-vessel vasculitis
Microscopic polyangiitis
Granulomatosis with polyangiitis (Wegener)
Eosinophilic granulomatosis with polyangiitis (Churg–Strauss)
Large-vessel vasculitis
Takayasu arteritis
Giant cell arteritis
Anti-GBM disease
Figure 1 | Diagram depicting the predominant types of vessels affected by major
categories of systemic vasculitis. Note that vasculitis that is associated with
ANCAs can target a broad range of vessel types including medium and small
arteries, arterioles, capillaries, venules and veins. Large arteries, such as main
visceral arteries, are rarely affected by AAV. Thus, the types of vessels affected
by AAV overlap with other variants of systemic vasculitis, which results in many
overlapping clinical signs and symptoms of disease. Abbreviations: AAV, ANCA-
associated vasculitis; ANCA, antineutrophil cytoplasmic antibody; GBM, glomerular
basement membrane. Reproduced with permission from John Wiley  Sons, Inc.
© Jennette, J. C. et al. Arthritis Rheum. 65, 1–11 (2013).
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  AUGUST 2014  |  465
frequent observation of leukocytoclasia (fragmentation
of nuclei) at sites of acute fibrinoid necrosis (Figure 2d).
Conspicuous leukocytoclasia with numerous nuclear
fragments could result from the intensity of neutrophil
infiltration or reduced clearance of neutrophil fragments,
or both. Neutrophil cytoplasmic and nuclear constitu-
ents, including neutrophil extracellular traps (NETs),
are present at sites of inflammation and acute necro-
sis.26–28
NETs comprise extracellular fibrillary material
containing chromatin and granule proteins extruded by
­activated neutrophils.
Necrotizing injury to vessel walls results in haemor-
rhage and release of plasma proteins into the vessel walls
and adjacent extravascular tissue. These proteins include
coagulation factors, which are activated by thrombogenic
cellular and tissue debris, and tissue factor, which results
in the formation of fibrin within areas of fibrinoid necro-
sis (Figure 2c,d). Fibrin formation also might be facili-
tated by tissue factor present in NETs.29
In glo­meruli,
foci of segmental fibroid necrosis develop adjacent cel-
lular reactions (crescents) composed predominantly
of monocytes, macrophages and activated epithelial
cells (Figure 2d).
Within days, the initial acute neutrophil-rich inflam-
mation and necrosis is replaced by inflammation with
a predominance of monocytes and macrophages and,
eventually, T cells (Figure 2c). Vascular and extravascu-
lar inflammatory infiltrates also often contain eosino-
phils, which are most numerous in EGPA. In glomerular
lesions of ANCA-associated glomerulonephritis, at the
time of biopsy, leukocytes are predominantly mono-
cytes and macrophages with admixed T cells and usually
only scant neutrophils.23–25
Within glomeruli in animal
models of ANCA-associated disease, the neutrophils
that are predominant during the first several days of
necrotizing inflammation are subsequently replaced by
­monocytes and macrophages.22
Within 1–2 weeks, vascular segments transition
through the monocyte and/or macrophage-rich phase of
inflammation to a phase of scarring that involves inter-
stitial deposition of collagen. The extent of scarring, and
the likelihood of return to normal structure and func-
tion, depends on the severity and duration of the inflam-
matory injury. Areas of mild segmental vascular injury
can undergo complete remodelling to restore normal
architecture, whereas severe vascular injury could result
in irreversible total vascular occlusion.
Granulomatosis
Patients with GPA and EGPA have nodular extra­
vascu­lar inflammatory lesions that contain numerous
macro­phages, often including multinucleated giant cells
(Figure 3).19–21
In an early granulomatous pulmonary
lesion, the centre is in essence a microabscess contain-
ing neutrophils in varying stages of necrosis and apop-
tosis, and the margin has a thin band of macrophages
that are responding to the necrosis (Figure 3a). In a later
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
epithelioid macrophages (macrophages with elongated
nuclei and abundant acidophilic cytoplasm, resembling
epithelial cells) (Figure 3b). At this stage, neutrophils
are no longer conspicuous, and the granulomatous
inflammation contains progressively more lymphoid
cells including effector T cells, memory T cells, B cells,
dendritic cells, macrophages and plasma cells.30
At a still-
later stage, the necrotic debris is replaced by scar tissue,
and lymphocytes, including organized lymphoid folli-
cles, become components of the chronic granulo­matous
inflammation. End-stage lesions are predominantly scar
tissue, often with no definitive histological features of the
earlier granulomatous inflammation.
Genesis of the ANCA autoimmune response
The evidence suggests that multiple genetic and environ-
mental factors converge to induce the onset of ANCA-
associated disease, and these factors have modulatory
effects on the clinical and pathological phenotype of
disease. These factors differ among patients; for example,
in a given patient the pivotal aetiological event could be
an infection, a drug, impaired immune regulation or
dysregulation of genomic expression of autoantigens,
or combinations of these and other factors.
Natural ANCAs
Although Ehrlich described the horror of autotoxicus
and Burnet theorized the elimination of ‘forbidden
clones’, all healthy individuals seem to have numerous
circulating autoantibodies with beneficial homeostatic
functions.31
Thus, pathogenic autoimmunity might arise
from dysregulation of homeostatic autoimmunity.
Healthy individuals have circulating autoantibodies
against MPO and PR3.32–36
In asymptomatic indi­viduals,
such autoantibodies are often designated ‘natural’ or
Box 1 | Evidence that ANCAs are pathogenic
Clinical evidence
■■ Strong association with ANCA (MPA 90%, GPA 90%, EGPA 40%*)
■■ Partial correlation of ANCA titre with disease activity
■■ Correlation of ANCA epitope specificity with disease activity (MPO‑ANCA only)
■■ Disease induction by transplacental transfer of ANCA (one MPO‑ANCA case report)
■■ Similar disease associated with drug-induced ANCA
■■ HLA genetic associations with MPO‑ANCA and PR3‑ANCA-associated disease
■■ Response to immunosuppressive therapy that targets B cells
In vitro evidence
■■ Activation of cytokine-primed neutrophils by ANCA IgG
■■ Endothelial injury by ANCA-activated neutrophils
■■ Alternative complement pathway activation by ANCA-activated neutrophils
Evidence from animal models‡
■■ Induction of pauci-immune vasculitis, glomerulonephritis and granulomatosis
in mice and rats by anti-MPO IgG
■■ Prevention of murine anti-MPO IgG-induced disease by deficiency of neutrophils
■■ Prevention of murine anti-MPO IgG-induced disease by blockade of alternative
complement pathway activation or blockade of C5a receptors
*More than 75% of patients with EGPA and glomerulonephritis have ANCA. ‡
Convincing animal
models of MPO‑ANCA-associated disease have been described, but only less-convincing
animal models of PR3‑ANCA-associated disease have been described. Abbreviations: ANCA,
antineutrophil cytoplasmic autoantibody; EGPA, eosinophilic granulomatosis with polyangiitis;
GPA, granulomatosis with polyangiitis; MPA, microscopic polyangiitis; MPO, myeloperoxidase;
PR3, proteinase 3.
FOCUS ON VASCULITIS
© 2014 Macmillan Publishers Limited. All rights reserved
466  |  AUGUST 2014  |  VOLUME 10 www.nature.com/nrrheum
nonpathogenic autoantibodies. Presumably, quanti­
tative or qualitative differences determine autoantibody
pathogenicity. Compared with pathogenic MPO‑ANCA,
natural MPO‑ANCA has lower titres, lower avidity, less
subclass diversity and less capability to activate neutro-
phils in vitro.34
In autoimmune diseases that have auto­
antigens with multiple epitope specificities, the epitope
specificity of natural autoantibodies can differ from that
of pathogenic autoantibodies. Roth et al.35
identified
more than 20 MPO epitopes that can be recognized by
MPO‑ANCA in patients with ANCA-associated dis­
ease. MPO‑ANCA IgG from patients with active disease
recognized the largest number of epitopes, including
epitopes that were never recognized by MPO‑ANCA
IgG from patients with ANCA-associated disease in
remission or from healthy individuals. Healthy indi­
viduals had very low titres of MPO‑ANCA 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
using conventional clinical assays reacted with a specific
MPO epitope when a highly sensitive epitope-excision
method was used; this epitope was blocked from reacting
with ANCA IgG in serum because of competitive binding
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
to become pathogenic.
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 PR3‑ANCA titre
before diagnosis (63% versus 0%) and a greater rate of
increase in titre (62% versus 0%). The rises in PR3‑ANCA
titre typically occurred before the onset of elevations in
serum C‑reactive protein level or signs and symptoms of
GPA. These findings are consistent with the concept that
endogenous or exogenous influences might modify quan-
titative or qualitative characteristics of natural ANCAs to
render them pathogenic.
Pathogenic ANCAs
Factors that could induce the development of patho-
genic ANCAs include exposure to exogenous anti-
gens that influence ANCA epitope specificity (such
as drugs or microbes), newly expressed or modified
ANCA autoantigens (such as alternatively spliced tran-
scripts or antisense transcripts), immunogenic display
of ANCA autoantigens (for example, on apoptotic cells
or NETs), or loss of effective suppression of the ANCA
autoimmune response (for example, because of defec-
tive T cells, B cells or myeloid-derived suppressor cells).
This pathogenic transformation could be multifactorial,
with genetic, environmental and immunological events
conspiring to break autoimmune homeostasis.
Mapping of epitopes that are the targets of natural and
pathogenic MPO‑ANCAs has demonstrated that these
two classes of epitopes occur in close proximity on the
3D structure of MPO.35
This finding suggests that epitope
spreading of ANCA specificity, from nonpathogenic to
pathogenic epitopes, occurs as disease develops.
Modulation of existing epitope specificities is known
to occur during immune responses to infectious patho-
gens, and several microorganisms have been implicated
in the induction and modulation of ANCA-associated
disease, such as Staphylococcus aureus37,38
and Ross
River virus.39
A novel model for induction of the ANCA
autoimmune response theorizes that the initial immune
a
c d
b
E L
A
A
N
Figure 2 | Vascular and glomerular pathology of AAV. a | Leukocytoclastic venulitis
in the nasal septal mucosa of a patient with PR3‑ANCA-associated disease
(magnification ×400, HE staining) demonstrates features of early AAV lesions:
numerous neutrophils (arrows) are marginating along and penetrating the wall of
a venule, and accumulating in the perivascular interstitium, with some undergoing
apoptosis and karyorrhexis (leukocytoclasia). b | Another venule from the same
specimen demonstrates neutrophils in the lumen, marginating neutrophils,
endothelial cells and apoptotic bodies derived from neutrophils (magnification
×1,000, HE staining). c | Necrotizing arteritis of a small artery in the kidney of
a patient with microscopic polyangiitis involves a circumferential zone of fibrinoid
necrosis (arrow) and perivascular leukocytes that, at this phase, contain
predominantly mononuclear leukocytes (magnification ×400, HE staining).
d | A glomerulus from a patient with ANCA-associated glomerulonephritis shows
segmental fibrinoid necrosis (lower arrow) and an adjacent cellular crescent
composed of mononuclear leukocytes and epithelial cells (upper arrow)
(magnification ×400, HE staining). Abbreviations: A, apoptotic bodies;
AAV, ANCA-associated vasculitis; ANCA, antineutrophil cytoplasmic antibody;
E, endothelial cells; HE, haematoxylin and eosin; L, lumen; N, marginating
neutrophils; PR3, proteinase 3.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  AUGUST 2014  |  467
response is not against the autoantigen but rather a
peptide that has a complementary structure relative to
the auto­antigen, and that the anti-idiotypic response
to this initial immune response results in antibodies that
recognize the autoantigen (Figure 4).40,41
Complementary
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
an antibody response to the sense peptide.42,43
Patients
positive for PR3‑ANCA have circulating antibodies
reactive not only to PR3 sense peptides but also to PR3
complementary antisense peptides.40,41
In addition to
antibodies that recognize complementary PR3 antigens,
Yang et al.44
detected anticomplementary PR3-specific
memory T cells—directed against a PR3 complementary
peptide—in patients with PR3‑ANCA. The stimulus for
the antibodies to complementary PR3 could have been
endogenous PR3 antisense peptides45
or exogenous pep-
tides that mimic the antisense peptides, such as peptides
brought in by commensal or pathogenic microbes.40
Interestingly, the bioinformatics software tool BLAST
(basic local alignment search tool) identifies multiple
microbial peptide homologues of complementary PR3,
including peptides from microbes that are known to be
associated with PR3‑ANCAs such as Ross River virus
and S. aureus.40,41
A genome-wide association study has indicated that
different HLA specificities correlate with MPO‑ANCA
and PR3‑ANCA, and a gene encoding PR3 was associ-
ated with PR3‑ANCA but not MPO‑ANCA.7
These
findings suggest that antigen-recognition capabilities
are important in ANCA-associated disease, and that the
initial antigen recognized by antigen-presenting cells is
different in PR3‑ANCA-associated disease as opposed
to MPO‑ANCA-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 PR3‑ANCA-associated disease
(odds ratio 73.3).46
The DRB1*1501 protein binds with
high affinity to both sense-PR3 peptide as well as the
complementary antisense-PR3 peptide.46
This finding
suggests that one factor in the induction of ANCA-
associated disease could be the presence of HLA antigen-­
binding sites that recognize the autoantigen and a
complementary protein.
Thus, the theory of autoantigen complementarity sug-
gests that a patient with a genetically determined pre-
disposing antigen-recognition capability could produce
pathogenic ANCA as a result of an initial appropriate
immune response against a microbial mimic of a com-
plementary peptide, or against an endogenously pro-
duced antisense transcript, that would be transformed
into an autoimmune response by an anti-idiotypic res­
ponse (Figure 4). In support of this theory, anti-idiotypic
a b
Figure 3 | Pathology of ANCA-associated necrotizing granulomatosis. a | The edge
of an early acute pulmonary lesion in a patient with PR3‑ANCA-positive GPA is
characterized by a central zone of necrosis containing numerous neutrophils in
varying stages of leukocytoclasia, and by an adjacent band of monocytes and
macrophages (arrows) (magnification ×400, HE staining). b | A later stage of
granulomatous inflammation in the same pulmonary lesion specimen demonstrates
a central zone of amorphous necrotic debris containing a few pyknotic leukocytes,
and an adjacent marginal zone of epithelioid macrophages (arrows) (magnification
×400, HE staining). Abbreviations: ANCA, antineutrophil cytoplasmic antibody;
GPA, granulomatosis with polyangiitis; HE, haematoxylin and eosin; PR3,
proteinase 3.
Endogenous source
Exogenous source
Microbes
Neutrophil
Anti-idiotypic
antibody
(autoantibody)
Mimic of
antisense peptide
Antisense
transcription
Sense
transcription
Sense DNA
strand
Antisense
DNA strand
ANCA antigen
peptide
Antisense
peptide
T cell
Idiotypic
antibody
Idiotope
mimicking the
autoantigen
Figure 4 | Diagram of the induction of an ANCA-mediated autoimmune response by
an initial immune response to a peptide that is complementary to an autoantigen
peptide. This complementary peptide immunogen could arise from antisense
transcription of the antisense strand of the autoantigen gene, or could be a mimic
of an antisense peptide that is produced by a symbiotic or pathogenic microbe.
The anticomplementary peptide antibody idiotopes would engender an anti-
idiotypic antibody response that crossreacts with the autoantigen epitopes that
are complementary to the initial immunogenic peptide. Abbreviation: ANCA,
antineutrophil cytoplasmic antibody.
FOCUS ON VASCULITIS
© 2014 Macmillan Publishers Limited. All rights reserved
468  |  AUGUST 2014  |  VOLUME 10 www.nature.com/nrrheum
antibodies raised against autoantibodies have been
shown in multiple animal models to induce anti-­anti-
idiotypic antibodies that possess characteristics of the
initial ­autoantibodies and cause autoimmune disease.47
Induction of pathogenic ANCAs by microbial molecu-
lar mimicry of autoantigens has been proposed as a cause
for ANCAs with specificity for LAMP‑2.10
The human
LAMP‑2 epitope has complete homology with the bacte-
rial adhesin FimH, and human LAMP‑2-ANCA cross-
reacts with this microbial protein. Kain et al.10
reported
that rats immunized with FimH develop antibodies to
rat and human LAMP‑2, and develop pauci-immune
necrotizing glomerulonephritis that resembles human
ANCA-associated glomerulonephritis. This study pro-
poses that infection with fimbriated bac­teria can cause
an immune response against FimH that crossreacts with
human LAMP‑2 and mediates ANCA-associated disease.
However, Roth et al.11
were not able to ­reproduce the
findings from this ­experimental animal model.
Another exogenous stimulus that can induce a
patho­genic ANCA immune response is drugs, includ-
ing hydralazine, minocycline, propylthiouracil and
levamisole-­adulterated cocaine.8,9
The mecha­nisms by
which drugs induce ANCAs could shed light on patho­
genic mechanisms that do not involve drugs. Hydralazine-
induced ANCA-associated disease is ac­companied by
dysregulation of ANCA-antigen expression by neutro-
phils.8
Hydralazine is a DNA methylation inhibi­tor that
might reverse epigenetic silencing of PR3 and MPO,
resulting in increased expression of both autoantigens in
neutrophils. This increased autoantigen expression
in drug-induced disease is similar to a phenomenon
observed in patients with ANCA-associated disease who
have abnormal epigenetically deregulated overexpres-
sion of MPO and PR3 in neutrophils.48,49
Increased auto­
antigen expression in neutrophils could either facilitate a
loss of tolerance to these proteins, or enhance the activa-
tion of neutrophils by ANCAs, or both. Organized lym-
phoid configuration in areas of granulomatosis that bring
together antigen-presenting cells with T cells and B cells,30
as well as effective presentation of neutrophil antigens on
apoptotic neutrophils50,51
and NETs,27
could also augment
ANCA autoimmune responses.
Regulation of the ANCA immune response
The pathogenic ANCA autoimmune response seems to
be enabled by impaired T‑cell and B‑cell suppression,
and possibly by enhanced B‑cell stimulation by ANCA-
activated neutrophils. Patients with ANCA-associated
disease have dysfunction of regulatory T (TREG
) cells that
could contribute to loss of tolerance and emergence of
a pathogenic ANCA response.52–54
TREG
cells are charac-
terized by high expression of CD25 and FOXP3. Even
though patients with active GPA have increased numbers
of CD4+
CD25+
T cells, the numbers of FOXP3+
cells are
decreased.52
The suppressive function of TREG
cells
from patients with ANCA-associated disease has been
reported to be markedly decreased in active disease and
more normal during remission.53
Free et al.54
described
an increase in a proinflammatory suppression-­resistant
effector CD4+
T‑cell subset in patients with active
ANCA-associated disease, in comparison with patients
in remission and with healthy individuals. Although
circulating TREG
cell numbers were increased in active
dis­ease, these cells had decreased suppressive function
because of expression of a FOXP3 isoform that lacks
exon 2.54
A study by Wilde et al.55
showed that increased
expression of the negative co-stimulator programmed
cell death protein 1 (PD‑1) on circulating T cells in
patients with GPA might oppose T‑cell activation; how­
ever, T cells in renal lesions in these patients mostly
lacked PD‑1 expression.
B-cell regulation might also influence production of
pathogenic ANCAs.56,57
B cells that have high expression
of CD5 and produce IL‑10 have regulatory capabilities.
Patients with active ANCA-associated disease have a
reduced percentage of circulating CD5+
B cells whereas
most patients in remission have a normal percentage.52
In addition, the degree of normalization of circulating
CD5+
B cells after therapy with rituximab correlates with
more-persistent remission.56
ANCA-activated neutrophils might augment the
production of ANCAs by stimulating B cells. Acti­
vated neutrophils release ligands for B‑cell-activating
fac­tor (BAFF; also known as TNF ligand superfamily
mem­ber 13B or B lymphocyte stimulator), which stimu-
lates B‑cell proliferation and inhibits apoptosis.58
Patients
with ANCA-associated disease have serum levels of
BAFF that are elevated during active disease and decline
during remission.59–62
Thus, B‑cell-stimulating factors released by ANCA-
activated neutrophils along with permissive T‑cell and
B‑cell regulation seem to facilitate the production of
pathogenic ANCAs.
Pathogenesis of vascular inflammation
Neutrophil and monocyte activation by ANCAs
In vitro studies have shown that both MPO‑ANCA and
PR3‑ANCA IgG can activate neutrophils that have been
primed by proinflammatory stimuli, such as TNF,63
bac-
terial lipopolysaccharide64
or the complement anaphyla-
toxin C5a.65
Priming causes neutrophils to release ANCA
target antigens (for example, MPO and PR3) at the cell
surface and into the microenvironment that can then
interact with ANCAs. Full neutrophil activation results
from engagement of constitutively expressed Fcγ recep-
tors (FcγRs) by immune complexes containing ANCA
and antigen66,67
and from binding of F(ab')2
fragments of
ANCA to antigen expressed on neutrophil surfaces.68,69
Phosphorylation of p38 mitogen-activated protein kinase
(p38 MAPK) and the extracellular signal-regulated
kinase (ERK) are involved in neutrophil priming and
activation, and ­blockade of MAPK prevents this priming
and activation.70–72
ANCA-activated neutrophils generate a respiratory
burst with production of toxic oxygen radicals, release
des­tructive enzymes through degranulation, and extrude
NETs that have proinflammatory properties.27,28,63–74
These ANCA-activated cells have been shown to cause
injury to and death of cultured monolayers of endo­thelial
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  AUGUST 2014  |  469
cells.75–78
Release of NETs (a process referred to as
NETosis) by ANCA-activated neutrophils27,28
contributes
to endothelial injury and death.79
In considering pathogenic pathways in ANCA-
associated disease, it is important to note that monocytes
as well as neutrophils contain ANCA antigens (including
PR3 and MPO) and can be activated in vitro by engage-
ment of FcγRs by ANCA IgG.80,81
ANCA-activated
monocytes release proinflammatory cytokines, includ-
ing monocyte chemoattractant protein‑1 (MCP‑1, also
known as CCL2)80
and IL‑8.81
IL‑8 attracts and activates
neutrophils and thus could amplify neutrophil-­mediated
injury. MCP‑1 attracts monocytes and macrophages and
could participate in the transition of ANCA-induced
vascular injury from predominantly neutrophil-rich
inflammation to predominantly monocyte and/or
macrophage-rich inflammation, including granulo­
matous inflammation. Monocytes are conspicuous
at sites of ANCA vascular inflammation,24
especially
after the first day or two, and there is evidence for sys-
temic activation of monocytes in patients with active
ANCA-associated disease.82
Induction of necrotizing vascular inflammation
Pathogenic mechanisms of ANCA-induced vascular
inflammation must be in accord with observed lesions
in tissue specimens from patients with ANCA-associated
disease or in animal models. Earlier in this Review, we
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/
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
inflammation, including glomerulonephritis, in patients’
biopsies15,26,83
and in experimental animal models of
ANCA-associated disease.84
In vitro studies have dem-
onstrated that ANCA-induced neutrophil activation
releases into the microenvironment ANCA-antigens that
can bind to nearby structures to act as targets for in situ
immune-complex formation.76
However, the amount
of immunoglobulin deposited at sites of inflammation in
ANCA-associated disease typically is far less than is seen
with conventional forms of immune-complex-mediated
vascular inflammation.1
This observation supports
the concept that the vascular inflammation of ANCA-
associated disease is caused primarily by direct activation
of neutrophils and monocytes by ANCA, with somewhat
incidental deposition of immune complexes containing
ANCA and ANCA antigen at the sites of injury.
The most compelling evidence that ANCAs are
pathogenic and the most informative experiments that
elucidate the underlying pathogenic mechanisms come
from animal models of ANCA-associated disease.85–87
Multiple convincing models of MPO‑ANCA-associated
dis­ease have been described and confirmed.84–87
How­
ever, no convincing models of PR3-ANCA-associated
disease have been widely accepted, although several
have been proposed.88–91
Mouse models of AAV and ANCA-associated glo-
merulonephritis induced by anti-MPO antibodies have
been used most extensively to elucidate pathogenic
mechanisms.64,65,71,84,92–98
Disease can be induced by
injecting mouse anti-MPO IgG into immunocompetent
or immunodeficient mice,84
injecting splenocytes con-
taining anti-MPO B cells into immunodeficient mice,84
or transplanting bone marrow that contains MPO-
positive myeloid cells into MPO-knockout mice.93
In
these models, a pathogenic level of anti-MPO antibodies
induces necrotizing and crescentic glomerulonephritis
in all mice of susceptible strains, and systemic necrotiz-
ing small-vessel vasculitis and granulomatous inflam-
mation in some, but not all, animals. The inflammatory
lesions in these mouse models closely mimic human
­ANCA-associated disease.
Several conclusions can be drawn from experiments
using mouse models of MPO‑ANCA-associated disease:
C5a
C5a receptor
Cytokine
Cytokine
receptor
Neutrophil
Apoptotic body
Macrophage
Monocyte
Unprimed
neutrophil
Priming
Activation
Alternative
pathway
activation
NETosis
Acute
inflammation
Chronic inflammation
and scarring
ANCA antigen
ANCA
Fc receptor
T cell
Fibroblast
Fibrin
Collagen
Epithelial cell
Figure 5 | Putative sequence of pathogenic events in ANCA-mediated vasculitis.
Circulating neutrophils are primed for activation by ANCA by inflammatory cytokines
or C5a derived from complement activation (note that monocytes can be similarly
primed and activated but are not illustrated). Primed neutrophils release ANCA
antigens at the cell surface and into the microenvironment, where they interact
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
activation. ANCA-activated neutrophils release factors that activate the alternative
complement pathway, generating C5a. C5a and ANCA create an inflammatory
amplification loop, with C5a attracting and priming more neutrophils for activation
by ANCA, which causes, in turn, further activation of the alternative complement
pathway and production of more C5a. ANCA-activated neutrophils marginate and
penetrate vessel walls and undergo respiratory burst, degranulation, NETosis,
apoptosis and necrosis. Disruption of endothelium allows plasma to spill into
vascular and perivascular tissue where activation of the coagulation cascade
produces the fibrin strands of fibroid necrosis. The innate inflammatory response
orchestrates the conversion of the initial acute neutrophil-rich inflammation into
mononuclear leukocyte-rich inflammation and subsequent collagen deposition
and fibrosis. Abbreviation: ANCA, antineutrophil cytoplasmic antibody.
FOCUS ON VASCULITIS
© 2014 Macmillan Publishers Limited. All rights reserved
470  |  AUGUST 2014  |  VOLUME 10 www.nature.com/nrrheum
antibody to MPO alone (in the absence of functional
T cells) is sufficient to cause acute disease;84
bone-­
marrow-derived cells are sufficient and necessary to
induce disease;92
neutrophils are required;92
genetic back-
ground determines the susceptibility to and severity of
dis­­ease;94
circulating proinflammatory factors exacer­bate
dis­ease;64
and activation of the alternative complement
path­way is required to induce vascular inflammation.96
Studies of these mouse models also showed that disease
can be prevented or ameliorated by hydrolysis of anti-
MPO IgG glycans, which affects FcγR–antibody inter-
actions;95
by inhibition of p38 MAPK, which prevents
priming of neutrophils by inflammatory stimuli;71
and
by blockade of C597
or C5a receptor,65,98
which prevents
activation of the alternative complement pathway.
Evidence from animal models of the involvement of
alternative complement pathway activation in the patho-
genesis of ANCA-associated disease prompted studies to
identify supportive evidence in human disease. Exami­
nation of glomerular and vascular inflammatory lesions
in biopsy specimens from patients with ANCA-associated
dis­ease revealed markers of alternative complement path­
way activation including factor B and properdin, as well
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
ANCA-associated disease, including elevated plasma
levels of C3a, C5a, soluble C5b‑9, 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
scenario can be proposed for the interaction of patho-
genic ANCAs with neutrophils that have been primed
by a synergistic proinflammatory condition (Figure 5).
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. FcγR engagement by ANCAs bound
to ANCA antigens as well as F(ab')2
binding to ANCA
antigens expressed on neutrophil surfaces cause neu-
trophil activation. ANCA-activated neutrophils release
factors that activate the alternative complement pathway
with generation of C5a. Neutrophils, C5a and ANCAs
create an inflammatory amplification loop, with C5a
attracting and priming more neutrophils for activation
by ANCAs, which in turn causes further activation of
the alternative complement pathway and consequent
production of more C5a. ANCA-activated neutrophils
marginate and penetrate vessel walls, and undergo res-
piratory burst, degranulation, NETosis, apoptosis and
necrosis. Disruption of endo­thelium enables plasma to
spill into vascular and perivascular tissue where acti-
vation of the coagulation cascade produces the fibrin
strands of fibroid necrosis. Modulated by the severity and
reversibility of the acute injury, the innate inflammatory
response orchestrates the conversion of the acute inflam-
mation into ­mononuclear-leukocyte-rich inflammation
and subsequent fibrosis.
Pathogenesis of granulomatosis
There has been substantial controversy over the nature
of the granulomatosis seen in GPA and EGPA. The term
‘granulomatosis’ has been used to describe the multiple
foci of extravascular inflammation that were described
initially by Klinger102
and Wegener103,104
in GPA, and
by Churg and Strauss in EGPA.105
Controversy over the
pathogenesis of this granulomatous inflammation derives
from the fact that it can arise from at least two distinct
pathways—one initiated by a T‑cell mediated (type IV)
immune response specific for PR3 or MPO, and another
by an antigen-independent innate response of macro­
phages to something in the tissue that needs to be
Unprimed
neutrophils
Activation
Microabscess
formation
Granulomatosis
Priming
C5a
C5a receptor
Cytokine
Cytokine
receptor
Neutrophil
Macrophage
Monocyte
ANCA antigen
ANCA
Fc receptor
T cell
Multinucleated
giant cell
Fibrin
Figure 6 | Putative sequence of pathogenic events in ANCA-mediated necrotizing
granulomatosis. An inflammatory prodrome, such as an infectious or allergic
inflammatory respiratory tract disease, positions increased numbers of primed
neutrophils in extravascular interstitial tissue. ANCA immunoglobulin in the
interstitial fluid would activate primed neutrophils and initiate an inflammatory
amplification loop that would attract and activate more neutrophils, resulting in the
formation of a necrotizing microabscess. This acute inflammation and necrosis
would initiate an innate inflammatory response that would wall off the necrotic zone
with granulomatous inflammation containing a predominance of monocytes and
macrophages with admixed lymphocytes. Abbreviation: ANCA, antineutrophil
cytoplasmic antibody.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  AUGUST 2014  |  471
walled off.106
The latter pathway is an exaggerated version
of the innate monocyte and macrophage response that
closely follows any acute inflammatory event. Although
many investigators have proposed that ANCA-associated
granulomatosis is caused by an antigen-specific type IV
adaptive immune response, we favour the theory that
the granulomatosis is an innate inflammatory response
to acute extravascular inflammation that is initiated by
ANCA-induced neutrophil activation.
The concept that the granulomatosis of GPA is a
res­ponse to acute neutrophil-mediated necrosis is not
new. For example, based on careful histopathological
examination of lesions in patients with GPA and EGPA,
Fienberg and his associates concluded that “micro­
necrosis, usually with neutrophils (microabscesses),
constitutes the early phase in the development of the
pathognomonic organized palisading granuloma”.21
Lungs of patients with active GPA and EGPA often have
a range of granulomatous lesions, from acute lesions with
central microabscesses (Figure 3a) or subacute lesions
with central necrosis (Figure 3b) to chronic lesions with
a central zone of scarring and chronic inflammation.20
Chronic lesions contain substantial accumulations of
lymphocytes and dendritic cells, including T cells and
B cells with specificity for ANCA antigens that could be
a source for autoantibody production.108
On the basis of this apparent sequence of pathologi-
cal events, and the knowledge that ANCA IgG activates
primed neutrophils, even in vitro, a reasonable hypothesis
is that if an inflammatory condition (perhaps infection in
GPA and allergy in EGPA) positions primed neutrophils
in the extravascular tissue (for example, the submucosa
of the respiratory tract), these primed neutrophils would
be activated by ANCA IgG in the interstitial fluid of a
patient with circulating ANCAs. ANCA-induced neutro-
phil activation, possibly fuelled by activation of the alter-
native complement path­way, would induce microabscess
formation that, in turn, would engender a granulomatous
reaction to wall off the zone of necrosis (Figure 6).
Conclusions
Pauci-immune necrotizing small-vessel vasculitis and
extra­vascular granulomatosis are associated with, and
might be caused by, ANCAs. The distinctive, extremely
des­tructive necrotizing vascular inflammation of AAV
seems to be fuelled by an amplification loop in which
ANCA-activated neutrophils activate the alterna-
tive complement pathway, which, in turn, leads to the
attraction and activation of more neutrophils. Promising
therapies are those that can target one or multiple com-
ponents of this vicious cycle (for example ANCAs,
ANCA antigens, neutrophil activation and complement
activation). The pathogenesis of ANCA-associated
necrotizing extra­vascular granulomatous inflammation
is not known, but might be fuelled by the same inflam-
matory amplification loop occurring in the extravas-
cular compartment, in­volving primed neutrophils and
­interstitial ANCAs.
Review criteria
This Review is based primarily on full-text, peer-reviewed,
English-language medical literature from the 1980s into
2014 reporting on vasculitis and glomerulonephritis
caused by antineutrophil cytoplasmic autoantibodies,
as well as the authors’ knowledge of the field acquired
over more than 30 years of involvement in research on
glomerulonephritis and vasculitis.
1.	 Jennette, J. C. et al. Revised International
Chapel Hill Consensus Conference
nomenclature of vasculitides. Arthritis Rheum.
65, 1–11 (2013).
2.	 Falk, R. J.  Jennette, J. C. Anti-neutrophil
cytoplasmic autoantibodies with specificity for
myeloperoxidase in patients with systemic
vasculitis and idiopathic necrotizing and
crescentic glomerulonephritis. N. Engl. J. Med.
318, 1651–1657 (1988).
3.	 Goldschmeding, R. et al. Wegener’s
granulomatosis autoantibodies identify a novel
diisopropylfluorophosphate-binding protein in
the lysosomes of normal human neutrophils.
J. Clin. Invest. 4, 1577–1579 (1988).
4.	 Niles, J. L., McCluskey, T., Ahmad, M. F. 
Amin Arnaout, M. A. Wegener’s granulomatosis
autoantigen is a novel neutrophil serine
proteinase. Blood 74, 1888–1893 (1989).
5.	 Jennette, J. C., Hoidal, J. H.  Falk, R. J.
Specificity of anti-neutrophil cytoplasmic
autoantibodies for proteinase 3. Blood 75,
2263–2264 (1990).
6.	 Lionaki, S. et al. Classification of antineutrophil
cytoplasmic autoantibody vasculitides: the role
of antineutrophil cytoplasmic autoantibody
specificity for myeloperoxidase or proteinase 3
in disease recognition and prognosis. Arthritis
Rheum. 64, 3452–3462 (2012).
7.	 Lyons, P. A. et al. Genetically distinct subsets
within ANCA-associated vasculitis. N. Engl.
J. Med. 367, 214–223 (2012).
8.	 Pendergraft, W. F. 3rd
 Niles, J. L. Trojan
horses: drug culprits associated with
antineutrophil cytoplasmic autoantibody (ANCA)
vasculitis. Curr. Opin. Rheumatol. 26, 42–49
(2014).
9.	 Graf, J. Rheumatic manifestations of cocaine
use. Curr. Opin. Rheumatol. 25, 50–55 (2013).
10.	 Kain, R. et al. Molecular mimicry in pauci-
immune focal necrotizing glomerulonephritis.
Nat. Med. 14, 1088–1096 (2008).
11.	 Roth, A. J. et al. Anti‑LAMP‑2 antibodies are not
prevalent in patients with antineutrophil
cytoplasmic autoantibody glomerulonephritis.
J. Am. Soc. Nephrol. 23, 545–555 (2012).
12.	 Kawakami, T., Ishizu, A., Arimura,Y.  Soma, Y.
Serum anti‑lysosomal‑associated membrane
protein‑2 antibody levels in cutaneous
polyarteritis nodosa. Acta Derm.Venereol.
93, 70–73 (2013).
13.	 Kawakami, T., Takeuchi, S., Arimura, Y. 
Soma, Y. Elevated antilysosomal-associated
membrane protein‑2 antibody levels in patients
with adult Henoch-Schönlein purpura. Br. J.
Dermatol. 166, 1206–1212 (2012).
14.	 Roozendaal, C. et al. Does analysis of the
antigenic specificities of anti-neutrophil
cytoplasmic antibodies contribute to their
clinical significance in the inflammatory bowel
diseases? Scand. J. Gastroenterol. 34,
1123–1131 (1999).
15.	 Ooi, C. J., Lim, B. L., Cheong, W. K., Ling, A. E. 
Ng, H. S. Antineutrophil cytoplasmic antibodies
(ANCAs) in patients with inflammatory bowel
disease show no correlation with proteinase 3,
lactoferrin, myeloperoxidase, elastase,
cathepsin G and lysozyme: a Singapore study.
Ann. Acad. Med. Singapore 29, 704–707 (2000).
16.	 Mahler, M. et al. PR3-ANCA: a promising
biomarker for ulcerative colitis with extensive
disease. Clin. Chim.Acta. 424, 267–273 (2013).
17.	 Hervier, B. et al. Systemic lupus erythematosus
associated with ANCA-associated vasculitis:
an overlapping syndrome? Rheumatol. Int. 32,
3285–3290 (2012).
18.	 Levy, J. B., Hammad, T., Coulthart, A., Dougan, T.
 Pusey, C. D. Clinical features and outcome of
patients with both ANCA and anti-GBM
antibodies. Kidney Int. 66, 1535–1540 (2004).
19.	 Jennette, J. C.  Thomas, D. B. in Heptinstall’s
Pathology of the Kidney 6th
edn (eds
Jennette, J. C. et al.) 643–674 (Lippincott
Williams  Wilkins, 2007).
20.	 Jennette, J. C. Nomenclature and classification
of vasculitis: lessons learned from
granulomatosis with polyangiitis (Wegner’s
granulomatosis). Clin. Exp. Immunol.
164 (Suppl. 1), 7–10 (2011).
21.	 Mark, E. J., Matsubara, O., Tan-Liu, N. S.
 Fienberg, R. The pulmonary biopsy
in the early diagnosis of Wegener’s (pathergic)
granulomatosis: a study based on 35 open lung
biopsies. Hum. Pathol. 19, 1065–1071 (1988).
22.	 Jennette, J. C., Xiao, H., Falk, R.  Gasim, A. M.
Experimental models of vasculitis and
FOCUS ON VASCULITIS
© 2014 Macmillan Publishers Limited. All rights reserved
472  |  AUGUST 2014  |  VOLUME 10 www.nature.com/nrrheum
glomerulonephritis induced by antineutrophil
cytoplasmic autoantibodies. Contrib. Nephrol.
169, 211–220 (2011).
23.	 Rastaldi, M. P. et al. Glomerular monocyte-
macrophage features in ANCA-positive renal
vasculitis and cryoglobulinemic nephritis. J.Am.
Soc. Nephrol. 11, 2036–2043 (2000).
24.	 Weidner, S., Carl, M., Riess, R. 
Rupprecht, H. D. Histologic analysis of renal
leukocyte infiltration in antineutrophil
cytoplasmic antibody-associated vasculitis:
importance of monocyte and neutrophil
infiltration in tissue damage. Arthritis Rheum.
50, 3651–3657 (2004).
25.	 Cunningham, M. A. et al. Prominence of cell-
mediated immunity effectors in “pauci-immune”
glomerulonephritis. J.Am. Soc. Nephrol. 10,
499–506 (1999).
26.	 Bajema, I. M., Hagen, E. C., de Heer, E., van der
Woude, F. J.  Bruijn, J. A. Colocalization of
ANCA-antigens and fibrinoid necrosis in
ANCA-associated vasculitis. Kidney Int. 60,
2025–2030 (2001).
27.	 Kessenbrock, K. et al. Netting neutrophils in
autoimmune small-vessel vasculitis. Nat. Med.
15, 623–625 (2009).
28.	 Abreu-Velez, A. M., Smith, J. G. Jr  Howard, M. S.
Presence of neutrophil extracellular traps and
antineutrophil cytoplasmic antibodies associated
with vasculitides. North Am. J. Med. Sci. 1,
309–313 (2009).
29.	 Kambas, K. et al. Tissue factor expression in
neutrophil extracellular traps and neutrophil
derived microparticles in antineutrophil
cytoplasmic antibody associated vasculitis may
promote thromboinflammation and the
thrombophilic state associated with the disease.
Ann. Rheum. Dis. http://dx.doi.org/10.1136/
annrheumdis-2013-203430 (2013).
30.	 Lamprecht, P.  Gross, W. L. Current knowledge
on cellular interactions in the WG‑granuloma.
Clin. Exp. Rheumatol. 25 (Suppl. 44), S49–S51
(2007).
31.	 Jennette, J. C.  Falk, R. J. The rise and fall of
horror autotoxicus and forbidden clones. Kidney
Int. 78, 533–535 (2010).
32.	 Cui, Z., Zhao, M. H., Segelmark, M. 
Hellmark, T. Natural autoantibodies to
myeloperoxidase, proteinase 3, and the
glomerular basement membrane are present
in normal individuals. Kidney Int. 78, 590–597
(2010).
33.	 Olson, S. W. et al. Asymptomatic autoantibodies
associate with future anti-glomerular basement
membrane disease. J.Am. Soc. Nephrol. 22,
1946–1952 (2011).
34.	 Xu, P. C., Cui, Z., Chen, M., Hellmark, T. 
Zhao, M. H. Comparison of characteristics of
natural autoantibodies against myeloperoxidase
and anti-myeloperoxidase autoantibodies from
patients with microscopic polyangiitis.
Rheumatology (Oxford) 50, 1236–1243 (2011).
35.	 Roth, A. J. et al. ANCA epitope specificity
determines pathogenicity, detectability and
clinical predictive value. J. Clin. Invest. 123,
1773–1783 (2013).
36.	 Olson, S. W. et al. Relation between
asymptomatic proteinase 3 antibodies and
future granulomatosis with polyangiitis. Clin. J.
Am. Soc. Nephrol. 8, 1312–1318 (2013).
37.	 Popa, E. R., Stegeman, C. A., Kallenberg, C. G.
 Tervaert, J. W. Staphylococcus aureus and
Wegener’s granulomatosis. Arthritis Res. 4,
77–79 (2002).
38.	 Laudien, M. et al. Nasal carriage of
Staphylococcus aureus and endonasal activity
in Wegener’s granulomatosis as compared to
rheumatoid arthritis and chronic rhinosinusitis
with nasal polyps. Clin. Exp. Rheumatol.
28 (Suppl. 57), 51–55 (2010).
39.	 Davies, D. J., Moran, J. E., Niall, J. F.
 Ryan, G. B. Segmental necrotizing
glomerulonephritis with antineutrophil antibody:
possible arbovirus aetiology? Br. Med. J. (Clin.
Res. Ed.) 285, 606 (1982).
40.	 Pendergraft, W. F. et al. Autoimmunity is triggered
by cPR-3(105–201), a protein complementary to
the autoantigen proteinase 3. Nat. Med. 10,
72–79 (2004).
41.	 Preston, G. A., Pendergraft, W. F. 3rd
 Falk, R. J.
New insights that link microbes with the
generation of antineutrophil cytoplasmic
autoantibodies: the theory of autoantigen
complementarity. Curr. Opin. Nephrol. Hypertens.
14, 217–222 (2005).
42.	 Heal, J. R., Roberts, G. W., Raynes, J. G.,
Bhakoo, A.  Miller, A. D. Specific interactions
between sense and complementary peptides:
the basis for the proteomic code. Chembiochem.
3, 136–151 (2002).
43.	 Mekler, L. B. On the specific mutual interaction
of amino acid residues of polypeptide chains
and amino acid residues with codons. Oncology
27, 286–288 (1973).
44.	 Yang, J. J. et al. ANCA patients have T cells
responsive to complementary PR‑3 autoantigen.
Kidney Int. 74, 1159–1169 (2008).
45.	 Badhwar, A. K., Simmons, J., Jennette, J. C.,
Falk, R. J.  Preston, G. A. Characterization of
an antisense transcript from the proteinase‑3
gene locus in patients with ANCA-vasculitis
utilizing strand-specific RT‑PCR and 5'RACE
[abstract]. J. Am. Soc. Nephrol. 19, 188A
(2008).
46.	 Cao, Y. et al. DRB1*15 allele is a risk factor for
PR3-ANCA disease in African Americans. J.Am.
Soc. Nephrol. 22, 1161–1167 (2011).
47.	 Shoenfeld, Y. Idiotypic induction of
autoimmunity: a new aspect of the idiotypic
network. FASEB J. 8, 1296–1301 (1994).
48.	 Yang, J. J. et al. Circumvention of normal
constraints on granule protein gene expression
in peripheral blood neutrophils and monocytes
of patients with antineutrophil cytoplasmic
autoantibody-associated glomerulonephritis.
J. Am. Soc. Nephrol. 15, 2103–2114 (2004).
49.	 Ciavatta, D. J. et al. Epigenetic basis for aberrant
upregulation of autoantigen genes in humans
with ANCA vasculitis. J. Clin. Invest. 120,
3209–3219 (2010).
50.	 Durant, S. et al. Apoptosis-induced proteinase 3
membrane expression is independent from
degranulation. J. Leukoc. Biol. 75, 87–98
(2004).
51.	 Kantari, C. et al. Proteinase 3, the Wegener
autoantigen, is externalized during neutrophil
apoptosis: evidence for a functional association
with phospholipid scramblase 1 and interference
with macrophage phagocytosis. Blood 110,
4086–4095 (2007).
52.	 Morgan, M. D. et al. Patients with Wegener’s
granulomatosis demonstrate a relative
deficiency and functional impairment of
T‑regulatory cells. Immunology 130, 64–73
(2010).
53.	 Rimbert, M. et al. Decreased numbers of blood
dendritic cells and defective function of
regulatory T cells in antineutrophil cytoplasmic
antibody-associated vasculitis. PLoS ONE 6,
e18734 (2011).
54.	 Free, M. E. et al. Patients with antineutrophil
cytoplasmic antibody-associated vasculitis have
defective TREG
cell function exacerbated by the
presence of a suppression-resistant effector cell
population. Arthritis Rheum. 65, 1922–1933
(2013).
55.	 Wilde, B et al. Aberrant expression of the
negative costimulator PD‑1 on T cells in
granulomatosis with polyangiitis. Rheumatology
(Oxford) 7, 1188–1197 (2012).
56.	 Bunch, D. O. et al. Decreased CD5+
B cells
in active ANCA vasculitis and relapse after
rituximab. Clin. J. Am. Soc. Nephrol. 8, 382–391
(2013).
57.	 Wilde B. et al. Regulatory B cells in ANCA-
associated vasculitis. Ann. Rheum. Dis. 72,
1416–1419 (2013).
58.	 Scapini, P., Bazzoni, F.  Cassatella, M. A.
Regulation of B‑cell‑activating factor
(BAFF)/B lymphocyte stimulator (BLyS)
expression in human neutrophils. Immunol.
Letters 116, 1–6 (2008).
59.	 Krumbholz, M. et al. BAFF is elevated in serum
of patients with Wegener’s granulomatosis.
J. Autoimmun. 25, 298–302 (2005).
60.	 Sanders, J. S., Huitma, M. G., Kallenberg, C. G.
 Stegeman, C. A. Plasma levels of soluble
interleukin 2 receptor, soluble CD30, interleukin
10 and B cell activator of the tumour necrosis
factor family during follow-up in vasculitis
associated with proteinase 3‑antineutrophil
cytoplasmic antibodies: associations with
disease activity and relapse. Ann. Rheum. Dis.
65, 1484–1489 (2006).
61.	 Nagai, M. et al. Serum levels of BAFF and APRIL
in myeloperoxidase anti-neutrophil cytoplasmic
autoantibody-associated renal vasculitis:
association with disease activity. Nephron Clin.
Pract. 118, c339–c345 (2011).
62.	 Bader, L., Koldingsnes, W.  Nossent, J.
B‑lymphocyte activating factor levels are
increased in patients with Wegener’s
granulomatosis and inversely correlated with
ANCA titer. Clin. Rheumatol. 29, 1031–1035
(2010).
63.	 Falk, R. J., Terrell, R. S., Charles, L. A. 
Jennette, J. C. Anti-neutrophil cytoplasmic
autoantibodies induce neutrophils to
degranulate and produce oxygen radicals
in vitro. Proc. Natl Acad. Sci. USA 87,
4115–4119 (1990).
64.	 Huugen, D. et al. Aggravation of anti-
myeloperoxidase antibody induced
glomerulonephritis by bacterial
lipopolysaccharide: role of tumor necrosis
factor α. Am. J. Pathol. 167, 47–58 (2005).
65.	 Schreiber, A. et al. C5a receptor mediates
neutrophil activation and ANCA-induced
glomerulonephritis. J. Am. Soc. Nephrol. 20,
289–298 (2009).
66.	 Porges, A. J. et al. Anti-neutrophil cytoplasmic
antibodies engage and activate human
neutrophils via Fcγ RIIa. J. Immunol. 153,
1271–1280 (1994).
67.	 Kocher, M., Siegel, M. E., Edberg, J. C. 
Kimberly, R. P. Cross-linking of Fcγ receptor IIa
and Fcγ receptor IIIb induces different
proadhesive phenotypes on human neutrophils.
J. Immunol. 159, 3940–3948 (1997).
68.	 Kettritz, R., Jennette, J. C.  Falk, R. J. Cross-
linking of ANCA-antigens stimulates superoxide
release by human neutrophils. J. Am. Soc.
Nephrol. 8, 386–394 (1997).
69.	 Williams, J. M. et al. Activation of the Gi
heterotrimeric G protein by ANCA IgG F(ab')2
fragments is necessary but not sufficient to
stimulate the recruitment of those downstream
mediators used by intact ANCA IgG. J. Am. Soc.
Nephrol. 14, 661–669 (2003).
70.	 Kettritz, R., Schreiber, A., Luft, F. C.  Haller, H.
Role of mitogen-activated protein kinases in
activation of human neutrophils by antineutrophil
cytoplasmic antibodies. J.Am. Soc. Nephrol. 12,
37–46 (2001).
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  AUGUST 2014  |  473
71.	 van der Veen, B. S. et al. Effects of p38 mitogen-
activated protein kinase inhibition on anti-
neutrophil cytoplasmic autoantibody
pathogenicity in vitro and in vivo. Ann. Rheum.
Dis. 70, 356–365 (2011).
72.	 Hao, J., Meng, L. Q., Xu, P. C., Chen, M. 
Zhao, M. H. p38MAPK, ERK and PI3K signaling
pathways are involved in C5a-primed neutrophils
for ANCA-mediated activation. PLoS ONE 7,
e38317 (2012).
73.	 Charles, L. A., Caldas, M. L., Falk, R. J.,
Terrell, R. S.  Jennette, J. C. Antibodies against
granule proteins activate neutrophils in vitro.
J. Leukoc. Biol. 50, 539–546 (1991).
74.	 Csernok, E., Ernst, M., Schmitt, W., Bainton, D. F.
 Gross, W. L. Activated neutrophils express
proteinase 3 on their plasma membrane in vitro
and in vivo. Clin. Exp. Immunol. 95, 244–250
(1994).
75.	 Ewert, B. H., Jennette, J. C.  Falk, R. J.
Anti-myeloperoxidase antibodies stimulate
neutrophils to damage human endothelial cells.
Kidney Int. 41, 375–383 (1992).
76.	 Savage, C. O., Gaskin, G., Pusey, C. D. 
Pearson, J. D. Myeloperoxidase binds to
vascular endothelial cells, is recognized by ANCA
and can enhance complement dependent
cytotoxicity. Adv. Exp. Med. Biol. 336, 121–123
(1993).
77.	 Ewert, B. H., Becker, M. E., Jennette, J. C.
 Falk, R. J. Antimyeloperoxidase antibodies
induce neutrophil adherence to cultured human
endothelial cells. Ren. Fail. 17, 125–133
(1995).
78.	 Lu, X., Garfield, A., Rainger, G. E., Savage, C. O.
 Nash, G. B. Mediation of endothelial cell
damage by serine proteases, but not superoxide
released from antineutrophil cytoplasmic
antibody-stimulated neutrophils. Arthritis
Rheum. 54, 1619–1628 (2006).
79.	 Gupta, A. K. et al. Activated endothelial cells
induce neutrophil extracellular traps and are
susceptible to NETosis-mediated cell death.
FEBS Letters 584, 3193–3197 (2010).
80.	 Casselman, B. L., Kilgore, K. S., Miller, B. F.
 Warren, J. S. Antibodies to neutrophil
cytoplasmic antigens induce monocyte
chemoattractant protein‑1 secretion from human
monocytes. J. Lab. Clin. Med. 126, 495–502
(1995).
81.	 Ralston, D. R., Marsh, C. B., Lowe, M. P. 
Wewers, M. D. Antineutrophil cytoplasmic
antibodies induce monocyte IL‑8 release. Role
of surface proteinase‑3, α1-antitrypsin, and Fcγ
receptors. J. Clin. Invest. 100, 1416–1424
(1997).
82.	 Muller Kobold, A. C., Kallenberg, C. G. 
Tervaert, J. W. Monocyte activation in patients
with Wegener’s granulomatosis. Ann. Rheum. Dis.
58, 237–245 (1999).
83.	 Arimura, Y. et al. The role of myeloperoxidase
and myeloperoxidase-antineutrophil cytoplasmic
antibodies (MPO-ANCAs) in the pathogenesis
of human MPO‑ANCA‑associated
glomerulonephritis. Clin. Exp. Nephrol. 17,
634–637 (2013).
84.	 Xiao, H. et al. Antineutrophil cytoplasmic
autoantibodies specific for myeloperoxidase
cause glomerulonephritis and vasculitis in mice.
J. Clin. Invest. 110, 955–963 (2002).
85.	 Jennette, J. C., Xiao, H., Falk, R.  Gasim, A. M.
Experimental models of vasculitis and
glomerulonephritis induced by antineutrophil
cytoplasmic autoantibodies. Contrib. Nephrol.
169, 211–220 (2011).
86.	 van Timmeren, M. M.  Heeringa, P.
Pathogenesis of ANCA-associated vasculitis:
recent insights from animal models. Curr. Opin.
Rheumatol. 24, 8–14 (2012).
87.	 Salama, A. D.  Little, M. A. Animal models of
antineutrophil cytoplasm antibody-associated
vasculitis. Curr. Opin. Rheumatol. 24, 1–7 (2012).
88.	 van der Geld, Y. M. et al. Rats and mice
immunized with chimeric human/mouse
proteinase 3 produce autoantibodies to mouse
Pr3 and rat granulocytes. Ann. Rheum. Dis. 66,
1679–1682 (2007).
89.	 Pfister, H. et al. Antineutrophil cytoplasmic
autoantibodies against the murine homolog
of proteinase 3 (Wegener autoantigen) are
pathogenic in vivo. Blood 104, 1411–1418
(2004).
90.	 Primo, V. C. et al. Anti-PR3 immune responses
induce segmental and necrotizing
glomerulonephritis. Clin. Exp. Immunol. 159,
327–337 (2010).
91.	 Little, M. A. et al. Anti-proteinase 3 anti-neutrophil
cytoplasm autoantibodies recapitulate systemic
vasculitis in mice with a humanized immune
system. PLoS ONE 7, e28626 (2012).
92.	 Xiao, H. et al. The role of neutrophils in the
induction of glomerulonephritis by anti-
myeloperoxidase antibodies. Am. J. Pathol. 167,
39–45 (2005).
93.	 Schreiber, A., Xiao, H., Falk, R. J. 
Jennette, J. C. Bone marrow-derived cells are
sufficient and necessary targets to mediate
glomerulonephritis and vasculitis induced by
anti-myeloperoxidase antibodies. J. Am. Soc.
Nephrol. 17, 3355–3364 (2006).
94.	 Xiao, H. et al. Genetically determined severity of
anti-myeloperoxidase glomerulonephritis. Am. J.
Pathol. 8, 139–160 (2013).
95.	 van Timmeren, M. M. et al. IgG glycan hydrolysis
attenuates ANCA-mediated glomerulonephritis.
J. Am. Soc. Nephrol. 21, 1103–1114 (2010).
96.	 Xiao, H., Schreiber, A., Heeringa, P., Falk, R. J. 
Jennette, J. C. Alternative complement pathway
in the pathogenesis of disease mediated by
antineutrophil cytoplasmic autoantibodies.
Am. J. Pathol. 170, 52–64 (2007).
97.	 Huugen, D. et al. Inhibition of complement factor
C5 protects against anti-myeloperoxidase
antibody-mediated glomerulonephritis in mice.
Kidney Int. 71, 646–654 (2007).
98.	 Xiao, H. et al. C5a receptor (CD88) blockade
protects against MPO-ANCA glomerulonephritis.
J. Am. Soc. Nephrol. 25, 225–231 (2014).
99.	 Xing, G. Q. et al. Complement activation
is involved in renal damage in human
antineutrophil cytoplasmic autoantibody
associated pauci-immune vasculitis. J. Clin.
Immunol. 29, 282–291 (2009).
100.	Yuan, J. et al. C5a and its receptors in human
anti-neutrophil cytoplasmic antibody (ANCA)-
associated vasculitis. Arthritis Res.Ther. 14,
R140 (2012).
101.	Gou, S. J., Yuan, J., Chen, M., Yu, F. 
Zhao, M. H. Circulating complement activation
in patients with anti-neutrophil cytoplasmic
antibody-associated vasculitis. Kidney Int. 83,
129–137 (2012).
102.	Klinger, H. Grenzformen der periarteriitis nodosa
[German]. Frankf. Z. Pathol. 42, 455–480 (1931).
103.	Wegener, F. Über generalisierte septische
Gefässerkrankungen [German]. Verh. Deut.
Pathol. Ges. 29, 202–210 (1936).
104.	Wegener, F. Über eine eigenartige rhinogene
Granulomatose mit besonderer Beteiligung des
Arteriensystems und der Nieren [German]. Beitr.
Pathol.Anat. 102, 30–68 (1939).
105.	Churg, J.  Strauss, L. Allergic granulomatosis,
allergic antiitis, and periarteritis nodosa. Am. J.
Pathol. 27, 277–294 (1951).
106.	Petersen, H. J.  Smith, A. M. The role of the
innate immune system in granulomatous
disorders. Front. Immunol. 4, 120 (2013).
107.	Voswinkel, J., Müller, A.  Lamprecht, P.
Is PR3-ANCA formation initiated in Wegener’s
granulomatosis lesions? Granulomas as
potential lymphoid tissue maintaining
autoantibody production. Ann. NY Acad. Sci.
1051, 12–19 (2005).
Acknowledgements
The authors are supported by a grant from the NIH
National Institute of Diabetes and Digestive and
Kidney Diseases (P01-DK058335‑11).
Author contributions
J.C.J. researched data for the article, and both
authors contributed equally to discussions of
content, writing and review/editing of the manuscript
before submission.
FOCUS ON VASCULITIS
© 2014 Macmillan Publishers Limited. All rights reserved

More Related Content

What's hot

Vasculitis syndrome an approach -and-basic principles of treatment
Vasculitis syndrome an approach -and-basic principles of treatmentVasculitis syndrome an approach -and-basic principles of treatment
Vasculitis syndrome an approach -and-basic principles of treatmentSachin Verma
 
Approach to the patient vasculitis (2)
Approach to the patient vasculitis (2)Approach to the patient vasculitis (2)
Approach to the patient vasculitis (2)Manjunath Anvekar
 
Econdary glomerular nephropathies
Econdary glomerular nephropathiesEcondary glomerular nephropathies
Econdary glomerular nephropathiesmedicinaingles1
 
Complement system
Complement  system Complement  system
Complement system Ajay Kurian
 
Pulmonary Langerhans Cell Histiocytosis (Plch), Eosinophilic Granuloma Of The...
Pulmonary Langerhans Cell Histiocytosis (Plch), Eosinophilic Granuloma Of The...Pulmonary Langerhans Cell Histiocytosis (Plch), Eosinophilic Granuloma Of The...
Pulmonary Langerhans Cell Histiocytosis (Plch), Eosinophilic Granuloma Of The...Bassel Ericsoussi, MD
 
Econdary glomerular nephropathies
Econdary glomerular nephropathiesEcondary glomerular nephropathies
Econdary glomerular nephropathiesMedicinaIngles
 
Pediatric vasculitis dr inayat ullah
Pediatric vasculitis dr inayat ullahPediatric vasculitis dr inayat ullah
Pediatric vasculitis dr inayat ullahDr Inayat Ullah
 
Childhood vasculitis
Childhood vasculitisChildhood vasculitis
Childhood vasculitisArnab Nandy
 
Small vessel vasculitis
Small vessel vasculitisSmall vessel vasculitis
Small vessel vasculitisJulfikar Saif
 
Feltys syndrome
Feltys syndromeFeltys syndrome
Feltys syndromeramarawand
 
Lupus nephritis 2016
Lupus nephritis 2016Lupus nephritis 2016
Lupus nephritis 2016drsamianik
 
Pulmonary Renal Syndromes
Pulmonary Renal SyndromesPulmonary Renal Syndromes
Pulmonary Renal SyndromesZunaira Islam
 

What's hot (20)

Vasculitis syndrome an approach -and-basic principles of treatment
Vasculitis syndrome an approach -and-basic principles of treatmentVasculitis syndrome an approach -and-basic principles of treatment
Vasculitis syndrome an approach -and-basic principles of treatment
 
Vasculitis
VasculitisVasculitis
Vasculitis
 
Approach to the patient vasculitis (2)
Approach to the patient vasculitis (2)Approach to the patient vasculitis (2)
Approach to the patient vasculitis (2)
 
Econdary glomerular nephropathies
Econdary glomerular nephropathiesEcondary glomerular nephropathies
Econdary glomerular nephropathies
 
Complement system
Complement  system Complement  system
Complement system
 
Pulmonary Langerhans Cell Histiocytosis (Plch), Eosinophilic Granuloma Of The...
Pulmonary Langerhans Cell Histiocytosis (Plch), Eosinophilic Granuloma Of The...Pulmonary Langerhans Cell Histiocytosis (Plch), Eosinophilic Granuloma Of The...
Pulmonary Langerhans Cell Histiocytosis (Plch), Eosinophilic Granuloma Of The...
 
Econdary glomerular nephropathies
Econdary glomerular nephropathiesEcondary glomerular nephropathies
Econdary glomerular nephropathies
 
Pediatric vasculitis dr inayat ullah
Pediatric vasculitis dr inayat ullahPediatric vasculitis dr inayat ullah
Pediatric vasculitis dr inayat ullah
 
Vasculitis
VasculitisVasculitis
Vasculitis
 
Vasculitis undergrad: diagnosis & treatment.
Vasculitis undergrad: diagnosis & treatment.Vasculitis undergrad: diagnosis & treatment.
Vasculitis undergrad: diagnosis & treatment.
 
THE VASCULITIS SYNDROME
THE VASCULITIS SYNDROMETHE VASCULITIS SYNDROME
THE VASCULITIS SYNDROME
 
Lecture samy- 2-4-16
Lecture  samy- 2-4-16Lecture  samy- 2-4-16
Lecture samy- 2-4-16
 
Childhood vasculitis
Childhood vasculitisChildhood vasculitis
Childhood vasculitis
 
Small vessel vasculitis
Small vessel vasculitisSmall vessel vasculitis
Small vessel vasculitis
 
Tutorial vasculitis
Tutorial vasculitisTutorial vasculitis
Tutorial vasculitis
 
Rheumatoid arthritis
Rheumatoid arthritisRheumatoid arthritis
Rheumatoid arthritis
 
Feltys syndrome
Feltys syndromeFeltys syndrome
Feltys syndrome
 
Lupus nephritis 2016
Lupus nephritis 2016Lupus nephritis 2016
Lupus nephritis 2016
 
Vasculitis
VasculitisVasculitis
Vasculitis
 
Pulmonary Renal Syndromes
Pulmonary Renal SyndromesPulmonary Renal Syndromes
Pulmonary Renal Syndromes
 

Viewers also liked

Mapping Inhibitory Neuronal Ircuits By Laser Scanning Photostimula Tion
Mapping Inhibitory Neuronal Ircuits By Laser Scanning Photostimula TionMapping Inhibitory Neuronal Ircuits By Laser Scanning Photostimula Tion
Mapping Inhibitory Neuronal Ircuits By Laser Scanning Photostimula TionTaruna Ikrar
 
A Double Point Mutation In The Selectivity Filter Site
A Double Point Mutation In The Selectivity Filter SiteA Double Point Mutation In The Selectivity Filter Site
A Double Point Mutation In The Selectivity Filter SiteTaruna Ikrar
 
Circulation Journal
Circulation JournalCirculation Journal
Circulation JournalTaruna Ikrar
 
3th First Author Publishcation With Animal Models
3th First Author Publishcation With Animal Models3th First Author Publishcation With Animal Models
3th First Author Publishcation With Animal ModelsTaruna Ikrar
 
Evaluation Of Channel Function After Alteration Of Amino Acid Residues At The...
Evaluation Of Channel Function After Alteration Of Amino Acid Residues At The...Evaluation Of Channel Function After Alteration Of Amino Acid Residues At The...
Evaluation Of Channel Function After Alteration Of Amino Acid Residues At The...Taruna Ikrar
 
Genetics Screening Of Human Cardiac Ryanodine Receptor Mutations In Ion Channel
Genetics Screening Of Human Cardiac Ryanodine Receptor Mutations In Ion ChannelGenetics Screening Of Human Cardiac Ryanodine Receptor Mutations In Ion Channel
Genetics Screening Of Human Cardiac Ryanodine Receptor Mutations In Ion ChannelTaruna Ikrar
 
International Journal of Cardiology
International Journal of CardiologyInternational Journal of Cardiology
International Journal of CardiologyTaruna Ikrar
 
The American Journal of Cardiology
The American Journal of CardiologyThe American Journal of Cardiology
The American Journal of CardiologyTaruna Ikrar
 
Nogo receptor1confinesadisinhibitorymicrocircuittothe criticalperiodinvisualc...
Nogo receptor1confinesadisinhibitorymicrocircuittothe criticalperiodinvisualc...Nogo receptor1confinesadisinhibitorymicrocircuittothe criticalperiodinvisualc...
Nogo receptor1confinesadisinhibitorymicrocircuittothe criticalperiodinvisualc...Taruna Ikrar
 
A disinhibitory microcircuit initiates critical period plasticity in the visu...
A disinhibitory microcircuit initiates critical period plasticity in the visu...A disinhibitory microcircuit initiates critical period plasticity in the visu...
A disinhibitory microcircuit initiates critical period plasticity in the visu...Taruna Ikrar
 

Viewers also liked (10)

Mapping Inhibitory Neuronal Ircuits By Laser Scanning Photostimula Tion
Mapping Inhibitory Neuronal Ircuits By Laser Scanning Photostimula TionMapping Inhibitory Neuronal Ircuits By Laser Scanning Photostimula Tion
Mapping Inhibitory Neuronal Ircuits By Laser Scanning Photostimula Tion
 
A Double Point Mutation In The Selectivity Filter Site
A Double Point Mutation In The Selectivity Filter SiteA Double Point Mutation In The Selectivity Filter Site
A Double Point Mutation In The Selectivity Filter Site
 
Circulation Journal
Circulation JournalCirculation Journal
Circulation Journal
 
3th First Author Publishcation With Animal Models
3th First Author Publishcation With Animal Models3th First Author Publishcation With Animal Models
3th First Author Publishcation With Animal Models
 
Evaluation Of Channel Function After Alteration Of Amino Acid Residues At The...
Evaluation Of Channel Function After Alteration Of Amino Acid Residues At The...Evaluation Of Channel Function After Alteration Of Amino Acid Residues At The...
Evaluation Of Channel Function After Alteration Of Amino Acid Residues At The...
 
Genetics Screening Of Human Cardiac Ryanodine Receptor Mutations In Ion Channel
Genetics Screening Of Human Cardiac Ryanodine Receptor Mutations In Ion ChannelGenetics Screening Of Human Cardiac Ryanodine Receptor Mutations In Ion Channel
Genetics Screening Of Human Cardiac Ryanodine Receptor Mutations In Ion Channel
 
International Journal of Cardiology
International Journal of CardiologyInternational Journal of Cardiology
International Journal of Cardiology
 
The American Journal of Cardiology
The American Journal of CardiologyThe American Journal of Cardiology
The American Journal of Cardiology
 
Nogo receptor1confinesadisinhibitorymicrocircuittothe criticalperiodinvisualc...
Nogo receptor1confinesadisinhibitorymicrocircuittothe criticalperiodinvisualc...Nogo receptor1confinesadisinhibitorymicrocircuittothe criticalperiodinvisualc...
Nogo receptor1confinesadisinhibitorymicrocircuittothe criticalperiodinvisualc...
 
A disinhibitory microcircuit initiates critical period plasticity in the visu...
A disinhibitory microcircuit initiates critical period plasticity in the visu...A disinhibitory microcircuit initiates critical period plasticity in the visu...
A disinhibitory microcircuit initiates critical period plasticity in the visu...
 

Similar to Vasculitis nrrheum.2014.103

anca vasculitis.pdf
anca vasculitis.pdfanca vasculitis.pdf
anca vasculitis.pdftaxxus1
 
Granulomatosis polyangitis GPA
Granulomatosis polyangitis GPAGranulomatosis polyangitis GPA
Granulomatosis polyangitis GPAMarwa Besar
 
Churg -Strauss syndrome
Churg -Strauss syndromeChurg -Strauss syndrome
Churg -Strauss syndromeJoisy Aloor
 
Vasculitides AND ANTI-GBM
Vasculitides AND ANTI-GBMVasculitides AND ANTI-GBM
Vasculitides AND ANTI-GBMAnass Qasem
 
Pulmonary manifestations of collagen vascular diseases
Pulmonary manifestations of collagen vascular diseasesPulmonary manifestations of collagen vascular diseases
Pulmonary manifestations of collagen vascular diseasesHemant Kumar Agarwal
 
Vasculitis nrrheum.2014.78
Vasculitis nrrheum.2014.78Vasculitis nrrheum.2014.78
Vasculitis nrrheum.2014.78Elsa von Licy
 
Diagnosis, Etiology, and Treatment of PACNS
Diagnosis, Etiology, and Treatment of PACNSDiagnosis, Etiology, and Treatment of PACNS
Diagnosis, Etiology, and Treatment of PACNSZachary Kassirer
 
Polyarteritis nodasa and microscopic polyangitis
Polyarteritis nodasa and microscopic polyangitisPolyarteritis nodasa and microscopic polyangitis
Polyarteritis nodasa and microscopic polyangitisMarwa Besar
 
Autoimmune liver disease laboratory diagnosis
Autoimmune liver disease laboratory diagnosisAutoimmune liver disease laboratory diagnosis
Autoimmune liver disease laboratory diagnosisDr. Rajesh Bendre
 
Cutaneous Vasculitis
Cutaneous VasculitisCutaneous Vasculitis
Cutaneous VasculitisDr Yugandar
 
Genetically Distinct differences of ANCA-Associated Vasculitis Disorders
Genetically Distinct differences of ANCA-Associated Vasculitis Disorders Genetically Distinct differences of ANCA-Associated Vasculitis Disorders
Genetically Distinct differences of ANCA-Associated Vasculitis Disorders Blake Bordelon
 
Pathogenesis of Interstitial cystitis
Pathogenesis of Interstitial  cystitisPathogenesis of Interstitial  cystitis
Pathogenesis of Interstitial cystitisRohan Sharma
 

Similar to Vasculitis nrrheum.2014.103 (20)

ANCA GN.pptx
ANCA GN.pptxANCA GN.pptx
ANCA GN.pptx
 
Renal vasculitis
Renal vasculitisRenal vasculitis
Renal vasculitis
 
Vasculitis anca positivo 2015
Vasculitis anca positivo 2015Vasculitis anca positivo 2015
Vasculitis anca positivo 2015
 
anca vasculitis.pdf
anca vasculitis.pdfanca vasculitis.pdf
anca vasculitis.pdf
 
MOIZ IFA
MOIZ IFAMOIZ IFA
MOIZ IFA
 
Granulomatosis polyangitis GPA
Granulomatosis polyangitis GPAGranulomatosis polyangitis GPA
Granulomatosis polyangitis GPA
 
Acalasia
AcalasiaAcalasia
Acalasia
 
Churg -Strauss syndrome
Churg -Strauss syndromeChurg -Strauss syndrome
Churg -Strauss syndrome
 
Vasculitides AND ANTI-GBM
Vasculitides AND ANTI-GBMVasculitides AND ANTI-GBM
Vasculitides AND ANTI-GBM
 
Pulmonary manifestations of collagen vascular diseases
Pulmonary manifestations of collagen vascular diseasesPulmonary manifestations of collagen vascular diseases
Pulmonary manifestations of collagen vascular diseases
 
Vasculitis
VasculitisVasculitis
Vasculitis
 
Myasthenia Gravis e-Medicine Article
Myasthenia Gravis e-Medicine Article Myasthenia Gravis e-Medicine Article
Myasthenia Gravis e-Medicine Article
 
Vasculitis nrrheum.2014.78
Vasculitis nrrheum.2014.78Vasculitis nrrheum.2014.78
Vasculitis nrrheum.2014.78
 
Diagnosis, Etiology, and Treatment of PACNS
Diagnosis, Etiology, and Treatment of PACNSDiagnosis, Etiology, and Treatment of PACNS
Diagnosis, Etiology, and Treatment of PACNS
 
Polyarteritis nodasa and microscopic polyangitis
Polyarteritis nodasa and microscopic polyangitisPolyarteritis nodasa and microscopic polyangitis
Polyarteritis nodasa and microscopic polyangitis
 
Autoimmune liver disease laboratory diagnosis
Autoimmune liver disease laboratory diagnosisAutoimmune liver disease laboratory diagnosis
Autoimmune liver disease laboratory diagnosis
 
Cutaneous Vasculitis
Cutaneous VasculitisCutaneous Vasculitis
Cutaneous Vasculitis
 
Genetically Distinct differences of ANCA-Associated Vasculitis Disorders
Genetically Distinct differences of ANCA-Associated Vasculitis Disorders Genetically Distinct differences of ANCA-Associated Vasculitis Disorders
Genetically Distinct differences of ANCA-Associated Vasculitis Disorders
 
Pathogenesis of Interstitial cystitis
Pathogenesis of Interstitial  cystitisPathogenesis of Interstitial  cystitis
Pathogenesis of Interstitial cystitis
 
Omer
OmerOmer
Omer
 

More from Elsa von Licy

Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...Elsa von Licy
 
Strategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfiStrategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfiElsa von Licy
 
Neuropsychophysiologie
NeuropsychophysiologieNeuropsychophysiologie
NeuropsychophysiologieElsa von Licy
 
L agressivite en psychanalyse (21 pages 184 ko)
L agressivite en psychanalyse (21 pages   184 ko)L agressivite en psychanalyse (21 pages   184 ko)
L agressivite en psychanalyse (21 pages 184 ko)Elsa von Licy
 
C1 clef pour_la_neuro
C1 clef pour_la_neuroC1 clef pour_la_neuro
C1 clef pour_la_neuroElsa von Licy
 
Vuillez jean philippe_p01
Vuillez jean philippe_p01Vuillez jean philippe_p01
Vuillez jean philippe_p01Elsa von Licy
 
Spr ue3.1 poly cours et exercices
Spr ue3.1   poly cours et exercicesSpr ue3.1   poly cours et exercices
Spr ue3.1 poly cours et exercicesElsa von Licy
 
Plan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 pPlan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 pElsa von Licy
 
Bioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutionsBioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutionsElsa von Licy
 
Poly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicalesPoly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicalesElsa von Licy
 
Methodes travail etudiants
Methodes travail etudiantsMethodes travail etudiants
Methodes travail etudiantsElsa von Licy
 
Atelier.etude.efficace
Atelier.etude.efficaceAtelier.etude.efficace
Atelier.etude.efficaceElsa von Licy
 
There is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_introThere is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_introElsa von Licy
 

More from Elsa von Licy (20)

Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
 
Strategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfiStrategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfi
 
Rainville pierre
Rainville pierreRainville pierre
Rainville pierre
 
Neuropsychophysiologie
NeuropsychophysiologieNeuropsychophysiologie
Neuropsychophysiologie
 
L agressivite en psychanalyse (21 pages 184 ko)
L agressivite en psychanalyse (21 pages   184 ko)L agressivite en psychanalyse (21 pages   184 ko)
L agressivite en psychanalyse (21 pages 184 ko)
 
C1 clef pour_la_neuro
C1 clef pour_la_neuroC1 clef pour_la_neuro
C1 clef pour_la_neuro
 
Hemostase polycop
Hemostase polycopHemostase polycop
Hemostase polycop
 
Antiphilos
AntiphilosAntiphilos
Antiphilos
 
Vuillez jean philippe_p01
Vuillez jean philippe_p01Vuillez jean philippe_p01
Vuillez jean philippe_p01
 
Spr ue3.1 poly cours et exercices
Spr ue3.1   poly cours et exercicesSpr ue3.1   poly cours et exercices
Spr ue3.1 poly cours et exercices
 
Plan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 pPlan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 p
 
M2 bmc2007 cours01
M2 bmc2007 cours01M2 bmc2007 cours01
M2 bmc2007 cours01
 
Feuilletage
FeuilletageFeuilletage
Feuilletage
 
Chapitre 1
Chapitre 1Chapitre 1
Chapitre 1
 
Biophy
BiophyBiophy
Biophy
 
Bioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutionsBioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutions
 
Poly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicalesPoly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicales
 
Methodes travail etudiants
Methodes travail etudiantsMethodes travail etudiants
Methodes travail etudiants
 
Atelier.etude.efficace
Atelier.etude.efficaceAtelier.etude.efficace
Atelier.etude.efficace
 
There is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_introThere is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_intro
 

Vasculitis nrrheum.2014.103

  • 1. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  AUGUST 2014  |  463 Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, 308 Brinkhous-Bullitt Building, CB#7525, Chapel Hill, NC 27599-7525, USA (J.C.J., R.J.F.). Correspondence to: J.C.J. jcj@med.unc.edu 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 T‑cell and B‑cell regulation. A putative 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. 10, 463–473 (2014); published online 8 July 2014; doi:10.1038/nrrheum.2014.103 Introduction Antineutrophil cytoplasmic autoantibodies (ANCAs) are associated with, and are the probable cause of, a distinct form of vasculitis that can affect any organ in the body. Although small vessels are the predominant target, ANCA-associated vasculitis (AAV) can affect many different types of vessels including ar­teries, arterioles, capillaries, venules and veins (Figure 1).1 Immuno­pathologically, AAV has an absence or paucity of immunoglobulin deposition in injured vessels (pauci- immune vasculitis), compared with the more extensive deposition of immunoglobulin in immune-complex- mediated small-vessel vasculitis.1 AAV can be limited to one organ at the time of presentation (for example, lung-limited disease or renal-limited disease) or involve multiple organs. Frequent target tissues are the upper and lower respiratory tract, kidneys, skin and periph- eral nerves. Onset and exacerbations induce signs and symptoms of high levels of circulating inflammatory cytokines, such as fever, arthralgia and myalgia. In addi- tion to these nonspecific systemic inflammatory mani- festations, inflammation of vessel walls causes more specific signs and symptoms of vasculitis depending on which vessels and which organs are affected, for example purpura caused by dermal venulitis; pulmonary haemorrhage caused by alveolar capillaritis; glomerulo- nephritis caused by glomerular capillaritis; peripheral neuropathy caused by epineural arteritis; and ocular inflammation caused by vasculitis in small vessels in the eye and orbit. In addition to vasculitis, some variants of ANCA-associated disease have extravascular necrotiz- ing granulomatous inflammation (granulomatosis)—­ seemingly not arising from vascular inflammation—that most often affects the upper and lower respiratory tracts but can affect any organ. On the basis of the distribution of vascular inflam- mation and the presence or absence of granulomatosis and asthma, systemic AAV is categorized as microscopic polyangiitis (MPA) if there is vasculitis but no evidence of granulomatosis or asthma, granulomatosis with poly- angiitis (GPA) if there is granulomatosis but no asthma, and eosinophilic granulomatosis with poly­angiitis (EGPA; also known as Churg–Strauss syndrome) if there is granulomatosis, asthma and blood eosinophilia.1 Organ-limited disease, including isolated pauci-immune necrotizing and crescentic glomerulonephritis, can be considered limited variants of MPA. Limited variants of GPA and EGPA can be confined to the respiratory tract. AAV can also be classified on the basis of auto­ antigen specificity. In patients with vasculitis, the two best-­documented autoantigen targets of ANCA are myelo­peroxidase (MPO)2 and proteinase 3 (PR3).3–5 Classification of AAV by antigen specificity clearly shows differences in clinical presentations and outcomes, Competing interests J.C.J. has acted as a consultant for Amicus Therapeutics, Genentech, GlaxoSmithKline and Protalix BioTherapeutics, and has undertaken research in collaboration with ChemoCentryx. R.J.F. declares no competing interests. FOCUS ON VASCULITIS © 2014 Macmillan Publishers Limited. All rights reserved
  • 2. 464  |  AUGUST 2014  |  VOLUME 10 www.nature.com/nrrheum organ system involvement, patterns of extravascular inflammation, and genetic associations including HLA associ­ations.6,7 The most informative AAV classifica- tion scheme includes both the ANCA antigen specifi­ city and the clinicopathological phenotype, for example MPO‑ANCA MPA or PR3‑ANCA MPA.1 MPO and PR3 are not the only autoantigens recog- nized by ANCAs. For instance, ANCAs specific for elastase occur, and are most often identified, in patients with drug-induced ANCA-associated disease, including disease induced by cocaine adulterated with leva­misole.8,9 A controversial autoantigen specificity is for lysosomal- associated membrane protein 2 (LAMP‑2). Kain et al.10 reported finding LAMP‑2-ANCA in ~90% of patients Key points ■■ Antineutrophil cytoplasmic autoantibodies (ANCAs) are associated with and probably cause pauci-immune systemic necrotizing small-vessel vasculitis and glomerulonephritis ■■ ANCAs specific for proteinase 3 or myeloperoxidase occur with organ- limited disease, microscopic polyangiitis, granulomatosis with polyangiitis or eosinophilic granulomatosis with polyangiitis ■■ ANCAs seem to cause vasculitis by activating circulating primed neutrophils and causing them to attach to, penetrate and damage vessel walls by undergoing respiratory burst, degranulation, NETosis, apoptosis and necrosis ■■ ANCA-induced neutrophil activation also releases factors that activate the alternative complement pathway, which establishes a destructive inflammatory amplification loop that attracts and activates more neutrophils that, in turn, further activates the complement system ■■ ANCA-associated granulomatosis might result from the same pathogenic sequence of events involving extravascular primed neutrophils and interstitial- fluid ANCAs, followed by a granulomatous reaction to wall off the resulting extravascular necrosis with pauci-immune necrotizing glomerulonephritis, including patients who were positive for MPO-ANCA or PR3-ANCA and others who were negative for both. However, Roth et al.11 were unable to confirm this high frequency of LAMP‑2-ANCA in these patients. Similarly, Kawakami et al. did not detect LAMP‑2-ANCA in patients with MPA, although they did report evidence for LAMP‑2-ANCA in some patients with cutaneous poly­ arteritis nodosa12 and IgA vasculitis (formerly known as Henoch–Schönlein purpura).13 ANCAs also occur in patients with inflammatory bowel disease, and are detected by indirect immunofluorescence assay in ~60% of patients with ulcerative colitis and ~20% of those with Crohn disease.14–16 In this setting, ANCAs only rarely have specificity for PR3, MPO or elastase: the major autoantigens are catalase, α‑enolase, lacto­transferrin and bactericidal permeability-increasing protein.14–16 ANCA, usually MPO‑ANCA, occurs in a small minor- ity of patients with systemic lupus erythe­matosus who seem to to have vasculitic features of ANCA-associated disease.17 Approximately 30% of patients with anti­ glomerular basement membrane disease have ANCA, usually MPO‑ANCA.18 An important role for ANCAs in the pathogenesis of vasculitis and glomerulonephritis is supported by clinical evidence and by in vitro and in vivo experimental data (summarized in Box 1). The leading theory proposes that circulating neutrophils and monocytes that have been primed by inflammatory stimuli display ANCA antigens at or near the cell surface, and that interaction of these antigens with ANCAs results in neutrophil activation and initiation of vascular inflammation. An extension of this theory proposes that primed extravascular neutro- phils interact with interstitial ANCAs, causing necrotiz- ing inflammation and resultant reactive granulomatous inflammation. This Review summarizes observations that support these pathogenic theories as well as puta- tive mechanisms for the origin of the pathogenic ANCA autoimmune response. Pathology of ANCA-associated disease Any valid theories about pathogenic mechanisms in ANCA-associated disease must explain the development of the observed pathologic lesions. The acute and chronic lesions described in this section provide ­guidance in ­formulating pathogenic theories. Vasculitis and glomerulonephritis In patients19–21 and experimental animal models,22 the acute vascular and extravascular lesions of ANCA- associated disease begin as neutrophil-rich necrotizing inflammation. In vessels, the earliest lesions have mar- gination and diapedesis of predominantly neutrophils with admixed monocytes (Figure 2a,b). Areas of segmen- tal vascular necrosis also develop perivascular infiltrates that initially have numerous neutrophils. Within hours, neutrophils undergo apoptosis and necrosis (Figure 2a,b). The early destruction of neutro- phils by apoptosis and necrosis during ANCA-associated glomerulonephritis and vasculitis is supported by the Medium-vessel vasculitis Polyarteritis nodosa Kawasaki disease Immune complex small-vessel vasculitis Cryoglobulinemic vasculitis IgA vasculitis (Henoch–Schönlein) Hypocomplementemic urticarial vasculitis (Anti-C1q vasculitis) ANCA-associated small-vessel vasculitis Microscopic polyangiitis Granulomatosis with polyangiitis (Wegener) Eosinophilic granulomatosis with polyangiitis (Churg–Strauss) Large-vessel vasculitis Takayasu arteritis Giant cell arteritis Anti-GBM disease Figure 1 | Diagram depicting the predominant types of vessels affected by major categories of systemic vasculitis. Note that vasculitis that is associated with ANCAs can target a broad range of vessel types including medium and small arteries, arterioles, capillaries, venules and veins. Large arteries, such as main visceral arteries, are rarely affected by AAV. Thus, the types of vessels affected by AAV overlap with other variants of systemic vasculitis, which results in many overlapping clinical signs and symptoms of disease. Abbreviations: AAV, ANCA- associated vasculitis; ANCA, antineutrophil cytoplasmic antibody; GBM, glomerular basement membrane. Reproduced with permission from John Wiley Sons, Inc. © Jennette, J. C. et al. Arthritis Rheum. 65, 1–11 (2013). REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 3. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  AUGUST 2014  |  465 frequent observation of leukocytoclasia (fragmentation of nuclei) at sites of acute fibrinoid necrosis (Figure 2d). Conspicuous leukocytoclasia with numerous nuclear fragments could result from the intensity of neutrophil infiltration or reduced clearance of neutrophil fragments, or both. Neutrophil cytoplasmic and nuclear constitu- ents, including neutrophil extracellular traps (NETs), are present at sites of inflammation and acute necro- sis.26–28 NETs comprise extracellular fibrillary material containing chromatin and granule proteins extruded by ­activated neutrophils. Necrotizing injury to vessel walls results in haemor- rhage and release of plasma proteins into the vessel walls and adjacent extravascular tissue. These proteins include coagulation factors, which are activated by thrombogenic cellular and tissue debris, and tissue factor, which results in the formation of fibrin within areas of fibrinoid necro- sis (Figure 2c,d). Fibrin formation also might be facili- tated by tissue factor present in NETs.29 In glo­meruli, foci of segmental fibroid necrosis develop adjacent cel- lular reactions (crescents) composed predominantly of monocytes, macrophages and activated epithelial cells (Figure 2d). Within days, the initial acute neutrophil-rich inflam- mation and necrosis is replaced by inflammation with a predominance of monocytes and macrophages and, eventually, T cells (Figure 2c). Vascular and extravascu- lar inflammatory infiltrates also often contain eosino- phils, which are most numerous in EGPA. In glomerular lesions of ANCA-associated glomerulonephritis, at the time of biopsy, leukocytes are predominantly mono- cytes and macrophages with admixed T cells and usually only scant neutrophils.23–25 Within glomeruli in animal models of ANCA-associated disease, the neutrophils that are predominant during the first several days of necrotizing inflammation are subsequently replaced by ­monocytes and macrophages.22 Within 1–2 weeks, vascular segments transition through the monocyte and/or macrophage-rich phase of inflammation to a phase of scarring that involves inter- stitial deposition of collagen. The extent of scarring, and the likelihood of return to normal structure and func- tion, depends on the severity and duration of the inflam- matory injury. Areas of mild segmental vascular injury can undergo complete remodelling to restore normal architecture, whereas severe vascular injury could result in irreversible total vascular occlusion. Granulomatosis Patients with GPA and EGPA have nodular extra­ vascu­lar inflammatory lesions that contain numerous macro­phages, often including multinucleated giant cells (Figure 3).19–21 In an early granulomatous pulmonary lesion, the centre is in essence a microabscess contain- ing neutrophils in varying stages of necrosis and apop- tosis, and the margin has a thin band of macrophages that are responding to the necrosis (Figure 3a). In a later 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 epithelioid macrophages (macrophages with elongated nuclei and abundant acidophilic cytoplasm, resembling epithelial cells) (Figure 3b). At this stage, neutrophils are no longer conspicuous, and the granulomatous inflammation contains progressively more lymphoid cells including effector T cells, memory T cells, B cells, dendritic cells, macrophages and plasma cells.30 At a still- later stage, the necrotic debris is replaced by scar tissue, and lymphocytes, including organized lymphoid folli- cles, become components of the chronic granulo­matous inflammation. End-stage lesions are predominantly scar tissue, often with no definitive histological features of the earlier granulomatous inflammation. Genesis of the ANCA autoimmune response The evidence suggests that multiple genetic and environ- mental factors converge to induce the onset of ANCA- associated disease, and these factors have modulatory effects on the clinical and pathological phenotype of disease. These factors differ among patients; for example, in a given patient the pivotal aetiological event could be an infection, a drug, impaired immune regulation or dysregulation of genomic expression of autoantigens, or combinations of these and other factors. Natural ANCAs Although Ehrlich described the horror of autotoxicus and Burnet theorized the elimination of ‘forbidden clones’, all healthy individuals seem to have numerous circulating autoantibodies with beneficial homeostatic functions.31 Thus, pathogenic autoimmunity might arise from dysregulation of homeostatic autoimmunity. Healthy individuals have circulating autoantibodies against MPO and PR3.32–36 In asymptomatic indi­viduals, such autoantibodies are often designated ‘natural’ or Box 1 | Evidence that ANCAs are pathogenic Clinical evidence ■■ Strong association with ANCA (MPA 90%, GPA 90%, EGPA 40%*) ■■ Partial correlation of ANCA titre with disease activity ■■ Correlation of ANCA epitope specificity with disease activity (MPO‑ANCA only) ■■ Disease induction by transplacental transfer of ANCA (one MPO‑ANCA case report) ■■ Similar disease associated with drug-induced ANCA ■■ HLA genetic associations with MPO‑ANCA and PR3‑ANCA-associated disease ■■ Response to immunosuppressive therapy that targets B cells In vitro evidence ■■ Activation of cytokine-primed neutrophils by ANCA IgG ■■ Endothelial injury by ANCA-activated neutrophils ■■ Alternative complement pathway activation by ANCA-activated neutrophils Evidence from animal models‡ ■■ Induction of pauci-immune vasculitis, glomerulonephritis and granulomatosis in mice and rats by anti-MPO IgG ■■ Prevention of murine anti-MPO IgG-induced disease by deficiency of neutrophils ■■ Prevention of murine anti-MPO IgG-induced disease by blockade of alternative complement pathway activation or blockade of C5a receptors *More than 75% of patients with EGPA and glomerulonephritis have ANCA. ‡ Convincing animal models of MPO‑ANCA-associated disease have been described, but only less-convincing animal models of PR3‑ANCA-associated disease have been described. Abbreviations: ANCA, antineutrophil cytoplasmic autoantibody; EGPA, eosinophilic granulomatosis with polyangiitis; GPA, granulomatosis with polyangiitis; MPA, microscopic polyangiitis; MPO, myeloperoxidase; PR3, proteinase 3. FOCUS ON VASCULITIS © 2014 Macmillan Publishers Limited. All rights reserved
  • 4. 466  |  AUGUST 2014  |  VOLUME 10 www.nature.com/nrrheum nonpathogenic autoantibodies. Presumably, quanti­ tative or qualitative differences determine autoantibody pathogenicity. Compared with pathogenic MPO‑ANCA, natural MPO‑ANCA has lower titres, lower avidity, less subclass diversity and less capability to activate neutro- phils in vitro.34 In autoimmune diseases that have auto­ antigens with multiple epitope specificities, the epitope specificity of natural autoantibodies can differ from that of pathogenic autoantibodies. Roth et al.35 identified more than 20 MPO epitopes that can be recognized by MPO‑ANCA in patients with ANCA-associated dis­ ease. MPO‑ANCA IgG from patients with active disease recognized the largest number of epitopes, including epitopes that were never recognized by MPO‑ANCA IgG from patients with ANCA-associated disease in remission or from healthy individuals. Healthy indi­ viduals had very low titres of MPO‑ANCA 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 using conventional clinical assays reacted with a specific MPO epitope when a highly sensitive epitope-excision method was used; this epitope was blocked from reacting with ANCA IgG in serum because of competitive binding 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 to become pathogenic. 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 PR3‑ANCA titre before diagnosis (63% versus 0%) and a greater rate of increase in titre (62% versus 0%). The rises in PR3‑ANCA titre typically occurred before the onset of elevations in serum C‑reactive protein level or signs and symptoms of GPA. These findings are consistent with the concept that endogenous or exogenous influences might modify quan- titative or qualitative characteristics of natural ANCAs to render them pathogenic. Pathogenic ANCAs Factors that could induce the development of patho- genic ANCAs include exposure to exogenous anti- gens that influence ANCA epitope specificity (such as drugs or microbes), newly expressed or modified ANCA autoantigens (such as alternatively spliced tran- scripts or antisense transcripts), immunogenic display of ANCA autoantigens (for example, on apoptotic cells or NETs), or loss of effective suppression of the ANCA autoimmune response (for example, because of defec- tive T cells, B cells or myeloid-derived suppressor cells). This pathogenic transformation could be multifactorial, with genetic, environmental and immunological events conspiring to break autoimmune homeostasis. Mapping of epitopes that are the targets of natural and pathogenic MPO‑ANCAs has demonstrated that these two classes of epitopes occur in close proximity on the 3D structure of MPO.35 This finding suggests that epitope spreading of ANCA specificity, from nonpathogenic to pathogenic epitopes, occurs as disease develops. Modulation of existing epitope specificities is known to occur during immune responses to infectious patho- gens, and several microorganisms have been implicated in the induction and modulation of ANCA-associated disease, such as Staphylococcus aureus37,38 and Ross River virus.39 A novel model for induction of the ANCA autoimmune response theorizes that the initial immune a c d b E L A A N Figure 2 | Vascular and glomerular pathology of AAV. a | Leukocytoclastic venulitis in the nasal septal mucosa of a patient with PR3‑ANCA-associated disease (magnification ×400, HE staining) demonstrates features of early AAV lesions: numerous neutrophils (arrows) are marginating along and penetrating the wall of a venule, and accumulating in the perivascular interstitium, with some undergoing apoptosis and karyorrhexis (leukocytoclasia). b | Another venule from the same specimen demonstrates neutrophils in the lumen, marginating neutrophils, endothelial cells and apoptotic bodies derived from neutrophils (magnification ×1,000, HE staining). c | Necrotizing arteritis of a small artery in the kidney of a patient with microscopic polyangiitis involves a circumferential zone of fibrinoid necrosis (arrow) and perivascular leukocytes that, at this phase, contain predominantly mononuclear leukocytes (magnification ×400, HE staining). d | A glomerulus from a patient with ANCA-associated glomerulonephritis shows segmental fibrinoid necrosis (lower arrow) and an adjacent cellular crescent composed of mononuclear leukocytes and epithelial cells (upper arrow) (magnification ×400, HE staining). Abbreviations: A, apoptotic bodies; AAV, ANCA-associated vasculitis; ANCA, antineutrophil cytoplasmic antibody; E, endothelial cells; HE, haematoxylin and eosin; L, lumen; N, marginating neutrophils; PR3, proteinase 3. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 5. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  AUGUST 2014  |  467 response is not against the autoantigen but rather a peptide that has a complementary structure relative to the auto­antigen, and that the anti-idiotypic response to this initial immune response results in antibodies that recognize the autoantigen (Figure 4).40,41 Complementary 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 an antibody response to the sense peptide.42,43 Patients positive for PR3‑ANCA have circulating antibodies reactive not only to PR3 sense peptides but also to PR3 complementary antisense peptides.40,41 In addition to antibodies that recognize complementary PR3 antigens, Yang et al.44 detected anticomplementary PR3-specific memory T cells—directed against a PR3 complementary peptide—in patients with PR3‑ANCA. The stimulus for the antibodies to complementary PR3 could have been endogenous PR3 antisense peptides45 or exogenous pep- tides that mimic the antisense peptides, such as peptides brought in by commensal or pathogenic microbes.40 Interestingly, the bioinformatics software tool BLAST (basic local alignment search tool) identifies multiple microbial peptide homologues of complementary PR3, including peptides from microbes that are known to be associated with PR3‑ANCAs such as Ross River virus and S. aureus.40,41 A genome-wide association study has indicated that different HLA specificities correlate with MPO‑ANCA and PR3‑ANCA, and a gene encoding PR3 was associ- ated with PR3‑ANCA but not MPO‑ANCA.7 These findings suggest that antigen-recognition capabilities are important in ANCA-associated disease, and that the initial antigen recognized by antigen-presenting cells is different in PR3‑ANCA-associated disease as opposed to MPO‑ANCA-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 PR3‑ANCA-associated disease (odds ratio 73.3).46 The DRB1*1501 protein binds with high affinity to both sense-PR3 peptide as well as the complementary antisense-PR3 peptide.46 This finding suggests that one factor in the induction of ANCA- associated disease could be the presence of HLA antigen-­ binding sites that recognize the autoantigen and a complementary protein. Thus, the theory of autoantigen complementarity sug- gests that a patient with a genetically determined pre- disposing antigen-recognition capability could produce pathogenic ANCA as a result of an initial appropriate immune response against a microbial mimic of a com- plementary peptide, or against an endogenously pro- duced antisense transcript, that would be transformed into an autoimmune response by an anti-idiotypic res­ ponse (Figure 4). In support of this theory, anti-idiotypic a b Figure 3 | Pathology of ANCA-associated necrotizing granulomatosis. a | The edge of an early acute pulmonary lesion in a patient with PR3‑ANCA-positive GPA is characterized by a central zone of necrosis containing numerous neutrophils in varying stages of leukocytoclasia, and by an adjacent band of monocytes and macrophages (arrows) (magnification ×400, HE staining). b | A later stage of granulomatous inflammation in the same pulmonary lesion specimen demonstrates a central zone of amorphous necrotic debris containing a few pyknotic leukocytes, and an adjacent marginal zone of epithelioid macrophages (arrows) (magnification ×400, HE staining). Abbreviations: ANCA, antineutrophil cytoplasmic antibody; GPA, granulomatosis with polyangiitis; HE, haematoxylin and eosin; PR3, proteinase 3. Endogenous source Exogenous source Microbes Neutrophil Anti-idiotypic antibody (autoantibody) Mimic of antisense peptide Antisense transcription Sense transcription Sense DNA strand Antisense DNA strand ANCA antigen peptide Antisense peptide T cell Idiotypic antibody Idiotope mimicking the autoantigen Figure 4 | Diagram of the induction of an ANCA-mediated autoimmune response by an initial immune response to a peptide that is complementary to an autoantigen peptide. This complementary peptide immunogen could arise from antisense transcription of the antisense strand of the autoantigen gene, or could be a mimic of an antisense peptide that is produced by a symbiotic or pathogenic microbe. The anticomplementary peptide antibody idiotopes would engender an anti- idiotypic antibody response that crossreacts with the autoantigen epitopes that are complementary to the initial immunogenic peptide. Abbreviation: ANCA, antineutrophil cytoplasmic antibody. FOCUS ON VASCULITIS © 2014 Macmillan Publishers Limited. All rights reserved
  • 6. 468  |  AUGUST 2014  |  VOLUME 10 www.nature.com/nrrheum antibodies raised against autoantibodies have been shown in multiple animal models to induce anti-­anti- idiotypic antibodies that possess characteristics of the initial ­autoantibodies and cause autoimmune disease.47 Induction of pathogenic ANCAs by microbial molecu- lar mimicry of autoantigens has been proposed as a cause for ANCAs with specificity for LAMP‑2.10 The human LAMP‑2 epitope has complete homology with the bacte- rial adhesin FimH, and human LAMP‑2-ANCA cross- reacts with this microbial protein. Kain et al.10 reported that rats immunized with FimH develop antibodies to rat and human LAMP‑2, and develop pauci-immune necrotizing glomerulonephritis that resembles human ANCA-associated glomerulonephritis. This study pro- poses that infection with fimbriated bac­teria can cause an immune response against FimH that crossreacts with human LAMP‑2 and mediates ANCA-associated disease. However, Roth et al.11 were not able to ­reproduce the findings from this ­experimental animal model. Another exogenous stimulus that can induce a patho­genic ANCA immune response is drugs, includ- ing hydralazine, minocycline, propylthiouracil and levamisole-­adulterated cocaine.8,9 The mecha­nisms by which drugs induce ANCAs could shed light on patho­ genic mechanisms that do not involve drugs. Hydralazine- induced ANCA-associated disease is ac­companied by dysregulation of ANCA-antigen expression by neutro- phils.8 Hydralazine is a DNA methylation inhibi­tor that might reverse epigenetic silencing of PR3 and MPO, resulting in increased expression of both autoantigens in neutrophils. This increased autoantigen expression in drug-induced disease is similar to a phenomenon observed in patients with ANCA-associated disease who have abnormal epigenetically deregulated overexpres- sion of MPO and PR3 in neutrophils.48,49 Increased auto­ antigen expression in neutrophils could either facilitate a loss of tolerance to these proteins, or enhance the activa- tion of neutrophils by ANCAs, or both. Organized lym- phoid configuration in areas of granulomatosis that bring together antigen-presenting cells with T cells and B cells,30 as well as effective presentation of neutrophil antigens on apoptotic neutrophils50,51 and NETs,27 could also augment ANCA autoimmune responses. Regulation of the ANCA immune response The pathogenic ANCA autoimmune response seems to be enabled by impaired T‑cell and B‑cell suppression, and possibly by enhanced B‑cell stimulation by ANCA- activated neutrophils. Patients with ANCA-associated disease have dysfunction of regulatory T (TREG ) cells that could contribute to loss of tolerance and emergence of a pathogenic ANCA response.52–54 TREG cells are charac- terized by high expression of CD25 and FOXP3. Even though patients with active GPA have increased numbers of CD4+ CD25+ T cells, the numbers of FOXP3+ cells are decreased.52 The suppressive function of TREG cells from patients with ANCA-associated disease has been reported to be markedly decreased in active disease and more normal during remission.53 Free et al.54 described an increase in a proinflammatory suppression-­resistant effector CD4+ T‑cell subset in patients with active ANCA-associated disease, in comparison with patients in remission and with healthy individuals. Although circulating TREG cell numbers were increased in active dis­ease, these cells had decreased suppressive function because of expression of a FOXP3 isoform that lacks exon 2.54 A study by Wilde et al.55 showed that increased expression of the negative co-stimulator programmed cell death protein 1 (PD‑1) on circulating T cells in patients with GPA might oppose T‑cell activation; how­ ever, T cells in renal lesions in these patients mostly lacked PD‑1 expression. B-cell regulation might also influence production of pathogenic ANCAs.56,57 B cells that have high expression of CD5 and produce IL‑10 have regulatory capabilities. Patients with active ANCA-associated disease have a reduced percentage of circulating CD5+ B cells whereas most patients in remission have a normal percentage.52 In addition, the degree of normalization of circulating CD5+ B cells after therapy with rituximab correlates with more-persistent remission.56 ANCA-activated neutrophils might augment the production of ANCAs by stimulating B cells. Acti­ vated neutrophils release ligands for B‑cell-activating fac­tor (BAFF; also known as TNF ligand superfamily mem­ber 13B or B lymphocyte stimulator), which stimu- lates B‑cell proliferation and inhibits apoptosis.58 Patients with ANCA-associated disease have serum levels of BAFF that are elevated during active disease and decline during remission.59–62 Thus, B‑cell-stimulating factors released by ANCA- activated neutrophils along with permissive T‑cell and B‑cell regulation seem to facilitate the production of pathogenic ANCAs. Pathogenesis of vascular inflammation Neutrophil and monocyte activation by ANCAs In vitro studies have shown that both MPO‑ANCA and PR3‑ANCA IgG can activate neutrophils that have been primed by proinflammatory stimuli, such as TNF,63 bac- terial lipopolysaccharide64 or the complement anaphyla- toxin C5a.65 Priming causes neutrophils to release ANCA target antigens (for example, MPO and PR3) at the cell surface and into the microenvironment that can then interact with ANCAs. Full neutrophil activation results from engagement of constitutively expressed Fcγ recep- tors (FcγRs) by immune complexes containing ANCA and antigen66,67 and from binding of F(ab')2 fragments of ANCA to antigen expressed on neutrophil surfaces.68,69 Phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK) and the extracellular signal-regulated kinase (ERK) are involved in neutrophil priming and activation, and ­blockade of MAPK prevents this priming and activation.70–72 ANCA-activated neutrophils generate a respiratory burst with production of toxic oxygen radicals, release des­tructive enzymes through degranulation, and extrude NETs that have proinflammatory properties.27,28,63–74 These ANCA-activated cells have been shown to cause injury to and death of cultured monolayers of endo­thelial REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 7. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  AUGUST 2014  |  469 cells.75–78 Release of NETs (a process referred to as NETosis) by ANCA-activated neutrophils27,28 contributes to endothelial injury and death.79 In considering pathogenic pathways in ANCA- associated disease, it is important to note that monocytes as well as neutrophils contain ANCA antigens (including PR3 and MPO) and can be activated in vitro by engage- ment of FcγRs by ANCA IgG.80,81 ANCA-activated monocytes release proinflammatory cytokines, includ- ing monocyte chemoattractant protein‑1 (MCP‑1, also known as CCL2)80 and IL‑8.81 IL‑8 attracts and activates neutrophils and thus could amplify neutrophil-­mediated injury. MCP‑1 attracts monocytes and macrophages and could participate in the transition of ANCA-induced vascular injury from predominantly neutrophil-rich inflammation to predominantly monocyte and/or macrophage-rich inflammation, including granulo­ matous inflammation. Monocytes are conspicuous at sites of ANCA vascular inflammation,24 especially after the first day or two, and there is evidence for sys- temic activation of monocytes in patients with active ANCA-associated disease.82 Induction of necrotizing vascular inflammation Pathogenic mechanisms of ANCA-induced vascular inflammation must be in accord with observed lesions in tissue specimens from patients with ANCA-associated disease or in animal models. Earlier in this Review, we 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/ 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 inflammation, including glomerulonephritis, in patients’ biopsies15,26,83 and in experimental animal models of ANCA-associated disease.84 In vitro studies have dem- onstrated that ANCA-induced neutrophil activation releases into the microenvironment ANCA-antigens that can bind to nearby structures to act as targets for in situ immune-complex formation.76 However, the amount of immunoglobulin deposited at sites of inflammation in ANCA-associated disease typically is far less than is seen with conventional forms of immune-complex-mediated vascular inflammation.1 This observation supports the concept that the vascular inflammation of ANCA- associated disease is caused primarily by direct activation of neutrophils and monocytes by ANCA, with somewhat incidental deposition of immune complexes containing ANCA and ANCA antigen at the sites of injury. The most compelling evidence that ANCAs are pathogenic and the most informative experiments that elucidate the underlying pathogenic mechanisms come from animal models of ANCA-associated disease.85–87 Multiple convincing models of MPO‑ANCA-associated dis­ease have been described and confirmed.84–87 How­ ever, no convincing models of PR3-ANCA-associated disease have been widely accepted, although several have been proposed.88–91 Mouse models of AAV and ANCA-associated glo- merulonephritis induced by anti-MPO antibodies have been used most extensively to elucidate pathogenic mechanisms.64,65,71,84,92–98 Disease can be induced by injecting mouse anti-MPO IgG into immunocompetent or immunodeficient mice,84 injecting splenocytes con- taining anti-MPO B cells into immunodeficient mice,84 or transplanting bone marrow that contains MPO- positive myeloid cells into MPO-knockout mice.93 In these models, a pathogenic level of anti-MPO antibodies induces necrotizing and crescentic glomerulonephritis in all mice of susceptible strains, and systemic necrotiz- ing small-vessel vasculitis and granulomatous inflam- mation in some, but not all, animals. The inflammatory lesions in these mouse models closely mimic human ­ANCA-associated disease. Several conclusions can be drawn from experiments using mouse models of MPO‑ANCA-associated disease: C5a C5a receptor Cytokine Cytokine receptor Neutrophil Apoptotic body Macrophage Monocyte Unprimed neutrophil Priming Activation Alternative pathway activation NETosis Acute inflammation Chronic inflammation and scarring ANCA antigen ANCA Fc receptor T cell Fibroblast Fibrin Collagen Epithelial cell Figure 5 | Putative sequence of pathogenic events in ANCA-mediated vasculitis. Circulating neutrophils are primed for activation by ANCA by inflammatory cytokines or C5a derived from complement activation (note that monocytes can be similarly primed and activated but are not illustrated). Primed neutrophils release ANCA antigens at the cell surface and into the microenvironment, where they interact 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 activation. ANCA-activated neutrophils release factors that activate the alternative complement pathway, generating C5a. C5a and ANCA create an inflammatory amplification loop, with C5a attracting and priming more neutrophils for activation by ANCA, which causes, in turn, further activation of the alternative complement pathway and production of more C5a. ANCA-activated neutrophils marginate and penetrate vessel walls and undergo respiratory burst, degranulation, NETosis, apoptosis and necrosis. Disruption of endothelium allows plasma to spill into vascular and perivascular tissue where activation of the coagulation cascade produces the fibrin strands of fibroid necrosis. The innate inflammatory response orchestrates the conversion of the initial acute neutrophil-rich inflammation into mononuclear leukocyte-rich inflammation and subsequent collagen deposition and fibrosis. Abbreviation: ANCA, antineutrophil cytoplasmic antibody. FOCUS ON VASCULITIS © 2014 Macmillan Publishers Limited. All rights reserved
  • 8. 470  |  AUGUST 2014  |  VOLUME 10 www.nature.com/nrrheum antibody to MPO alone (in the absence of functional T cells) is sufficient to cause acute disease;84 bone-­ marrow-derived cells are sufficient and necessary to induce disease;92 neutrophils are required;92 genetic back- ground determines the susceptibility to and severity of dis­­ease;94 circulating proinflammatory factors exacer­bate dis­ease;64 and activation of the alternative complement path­way is required to induce vascular inflammation.96 Studies of these mouse models also showed that disease can be prevented or ameliorated by hydrolysis of anti- MPO IgG glycans, which affects FcγR–antibody inter- actions;95 by inhibition of p38 MAPK, which prevents priming of neutrophils by inflammatory stimuli;71 and by blockade of C597 or C5a receptor,65,98 which prevents activation of the alternative complement pathway. Evidence from animal models of the involvement of alternative complement pathway activation in the patho- genesis of ANCA-associated disease prompted studies to identify supportive evidence in human disease. Exami­ nation of glomerular and vascular inflammatory lesions in biopsy specimens from patients with ANCA-associated dis­ease revealed markers of alternative complement path­ way activation including factor B and properdin, as well 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 ANCA-associated disease, including elevated plasma levels of C3a, C5a, soluble C5b‑9, 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 scenario can be proposed for the interaction of patho- genic ANCAs with neutrophils that have been primed by a synergistic proinflammatory condition (Figure 5). 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. FcγR engagement by ANCAs bound to ANCA antigens as well as F(ab')2 binding to ANCA antigens expressed on neutrophil surfaces cause neu- trophil activation. ANCA-activated neutrophils release factors that activate the alternative complement pathway with generation of C5a. Neutrophils, C5a and ANCAs create an inflammatory amplification loop, with C5a attracting and priming more neutrophils for activation by ANCAs, which in turn causes further activation of the alternative complement pathway and consequent production of more C5a. ANCA-activated neutrophils marginate and penetrate vessel walls, and undergo res- piratory burst, degranulation, NETosis, apoptosis and necrosis. Disruption of endo­thelium enables plasma to spill into vascular and perivascular tissue where acti- vation of the coagulation cascade produces the fibrin strands of fibroid necrosis. Modulated by the severity and reversibility of the acute injury, the innate inflammatory response orchestrates the conversion of the acute inflam- mation into ­mononuclear-leukocyte-rich inflammation and subsequent fibrosis. Pathogenesis of granulomatosis There has been substantial controversy over the nature of the granulomatosis seen in GPA and EGPA. The term ‘granulomatosis’ has been used to describe the multiple foci of extravascular inflammation that were described initially by Klinger102 and Wegener103,104 in GPA, and by Churg and Strauss in EGPA.105 Controversy over the pathogenesis of this granulomatous inflammation derives from the fact that it can arise from at least two distinct pathways—one initiated by a T‑cell mediated (type IV) immune response specific for PR3 or MPO, and another by an antigen-independent innate response of macro­ phages to something in the tissue that needs to be Unprimed neutrophils Activation Microabscess formation Granulomatosis Priming C5a C5a receptor Cytokine Cytokine receptor Neutrophil Macrophage Monocyte ANCA antigen ANCA Fc receptor T cell Multinucleated giant cell Fibrin Figure 6 | Putative sequence of pathogenic events in ANCA-mediated necrotizing granulomatosis. An inflammatory prodrome, such as an infectious or allergic inflammatory respiratory tract disease, positions increased numbers of primed neutrophils in extravascular interstitial tissue. ANCA immunoglobulin in the interstitial fluid would activate primed neutrophils and initiate an inflammatory amplification loop that would attract and activate more neutrophils, resulting in the formation of a necrotizing microabscess. This acute inflammation and necrosis would initiate an innate inflammatory response that would wall off the necrotic zone with granulomatous inflammation containing a predominance of monocytes and macrophages with admixed lymphocytes. Abbreviation: ANCA, antineutrophil cytoplasmic antibody. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 9. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  AUGUST 2014  |  471 walled off.106 The latter pathway is an exaggerated version of the innate monocyte and macrophage response that closely follows any acute inflammatory event. Although many investigators have proposed that ANCA-associated granulomatosis is caused by an antigen-specific type IV adaptive immune response, we favour the theory that the granulomatosis is an innate inflammatory response to acute extravascular inflammation that is initiated by ANCA-induced neutrophil activation. The concept that the granulomatosis of GPA is a res­ponse to acute neutrophil-mediated necrosis is not new. For example, based on careful histopathological examination of lesions in patients with GPA and EGPA, Fienberg and his associates concluded that “micro­ necrosis, usually with neutrophils (microabscesses), constitutes the early phase in the development of the pathognomonic organized palisading granuloma”.21 Lungs of patients with active GPA and EGPA often have a range of granulomatous lesions, from acute lesions with central microabscesses (Figure 3a) or subacute lesions with central necrosis (Figure 3b) to chronic lesions with a central zone of scarring and chronic inflammation.20 Chronic lesions contain substantial accumulations of lymphocytes and dendritic cells, including T cells and B cells with specificity for ANCA antigens that could be a source for autoantibody production.108 On the basis of this apparent sequence of pathologi- cal events, and the knowledge that ANCA IgG activates primed neutrophils, even in vitro, a reasonable hypothesis is that if an inflammatory condition (perhaps infection in GPA and allergy in EGPA) positions primed neutrophils in the extravascular tissue (for example, the submucosa of the respiratory tract), these primed neutrophils would be activated by ANCA IgG in the interstitial fluid of a patient with circulating ANCAs. ANCA-induced neutro- phil activation, possibly fuelled by activation of the alter- native complement path­way, would induce microabscess formation that, in turn, would engender a granulomatous reaction to wall off the zone of necrosis (Figure 6). Conclusions Pauci-immune necrotizing small-vessel vasculitis and extra­vascular granulomatosis are associated with, and might be caused by, ANCAs. The distinctive, extremely des­tructive necrotizing vascular inflammation of AAV seems to be fuelled by an amplification loop in which ANCA-activated neutrophils activate the alterna- tive complement pathway, which, in turn, leads to the attraction and activation of more neutrophils. Promising therapies are those that can target one or multiple com- ponents of this vicious cycle (for example ANCAs, ANCA antigens, neutrophil activation and complement activation). The pathogenesis of ANCA-associated necrotizing extra­vascular granulomatous inflammation is not known, but might be fuelled by the same inflam- matory amplification loop occurring in the extravas- cular compartment, in­volving primed neutrophils and ­interstitial ANCAs. Review criteria This Review is based primarily on full-text, peer-reviewed, English-language medical literature from the 1980s into 2014 reporting on vasculitis and glomerulonephritis caused by antineutrophil cytoplasmic autoantibodies, as well as the authors’ knowledge of the field acquired over more than 30 years of involvement in research on glomerulonephritis and vasculitis. 1. Jennette, J. C. et al. Revised International Chapel Hill Consensus Conference nomenclature of vasculitides. Arthritis Rheum. 65, 1–11 (2013). 2. Falk, R. J. Jennette, J. C. Anti-neutrophil cytoplasmic autoantibodies with specificity for myeloperoxidase in patients with systemic vasculitis and idiopathic necrotizing and crescentic glomerulonephritis. N. Engl. J. Med. 318, 1651–1657 (1988). 3. Goldschmeding, R. et al. Wegener’s granulomatosis autoantibodies identify a novel diisopropylfluorophosphate-binding protein in the lysosomes of normal human neutrophils. J. Clin. Invest. 4, 1577–1579 (1988). 4. Niles, J. L., McCluskey, T., Ahmad, M. F. Amin Arnaout, M. A. Wegener’s granulomatosis autoantigen is a novel neutrophil serine proteinase. Blood 74, 1888–1893 (1989). 5. Jennette, J. C., Hoidal, J. H. Falk, R. J. Specificity of anti-neutrophil cytoplasmic autoantibodies for proteinase 3. Blood 75, 2263–2264 (1990). 6. Lionaki, S. et al. Classification of antineutrophil cytoplasmic autoantibody vasculitides: the role of antineutrophil cytoplasmic autoantibody specificity for myeloperoxidase or proteinase 3 in disease recognition and prognosis. Arthritis Rheum. 64, 3452–3462 (2012). 7. Lyons, P. A. et al. Genetically distinct subsets within ANCA-associated vasculitis. N. Engl. J. Med. 367, 214–223 (2012). 8. Pendergraft, W. F. 3rd Niles, J. L. Trojan horses: drug culprits associated with antineutrophil cytoplasmic autoantibody (ANCA) vasculitis. Curr. Opin. Rheumatol. 26, 42–49 (2014). 9. Graf, J. Rheumatic manifestations of cocaine use. Curr. Opin. Rheumatol. 25, 50–55 (2013). 10. Kain, R. et al. Molecular mimicry in pauci- immune focal necrotizing glomerulonephritis. Nat. Med. 14, 1088–1096 (2008). 11. Roth, A. J. et al. Anti‑LAMP‑2 antibodies are not prevalent in patients with antineutrophil cytoplasmic autoantibody glomerulonephritis. J. Am. Soc. Nephrol. 23, 545–555 (2012). 12. Kawakami, T., Ishizu, A., Arimura,Y. Soma, Y. Serum anti‑lysosomal‑associated membrane protein‑2 antibody levels in cutaneous polyarteritis nodosa. Acta Derm.Venereol. 93, 70–73 (2013). 13. Kawakami, T., Takeuchi, S., Arimura, Y. Soma, Y. Elevated antilysosomal-associated membrane protein‑2 antibody levels in patients with adult Henoch-Schönlein purpura. Br. J. Dermatol. 166, 1206–1212 (2012). 14. Roozendaal, C. et al. Does analysis of the antigenic specificities of anti-neutrophil cytoplasmic antibodies contribute to their clinical significance in the inflammatory bowel diseases? Scand. J. Gastroenterol. 34, 1123–1131 (1999). 15. Ooi, C. J., Lim, B. L., Cheong, W. K., Ling, A. E. Ng, H. S. Antineutrophil cytoplasmic antibodies (ANCAs) in patients with inflammatory bowel disease show no correlation with proteinase 3, lactoferrin, myeloperoxidase, elastase, cathepsin G and lysozyme: a Singapore study. Ann. Acad. Med. Singapore 29, 704–707 (2000). 16. Mahler, M. et al. PR3-ANCA: a promising biomarker for ulcerative colitis with extensive disease. Clin. Chim.Acta. 424, 267–273 (2013). 17. Hervier, B. et al. Systemic lupus erythematosus associated with ANCA-associated vasculitis: an overlapping syndrome? Rheumatol. Int. 32, 3285–3290 (2012). 18. Levy, J. B., Hammad, T., Coulthart, A., Dougan, T. Pusey, C. D. Clinical features and outcome of patients with both ANCA and anti-GBM antibodies. Kidney Int. 66, 1535–1540 (2004). 19. Jennette, J. C. Thomas, D. B. in Heptinstall’s Pathology of the Kidney 6th edn (eds Jennette, J. C. et al.) 643–674 (Lippincott Williams Wilkins, 2007). 20. Jennette, J. C. Nomenclature and classification of vasculitis: lessons learned from granulomatosis with polyangiitis (Wegner’s granulomatosis). Clin. Exp. Immunol. 164 (Suppl. 1), 7–10 (2011). 21. Mark, E. J., Matsubara, O., Tan-Liu, N. S. Fienberg, R. The pulmonary biopsy in the early diagnosis of Wegener’s (pathergic) granulomatosis: a study based on 35 open lung biopsies. Hum. Pathol. 19, 1065–1071 (1988). 22. Jennette, J. C., Xiao, H., Falk, R. Gasim, A. M. Experimental models of vasculitis and FOCUS ON VASCULITIS © 2014 Macmillan Publishers Limited. All rights reserved
  • 10. 472  |  AUGUST 2014  |  VOLUME 10 www.nature.com/nrrheum glomerulonephritis induced by antineutrophil cytoplasmic autoantibodies. Contrib. Nephrol. 169, 211–220 (2011). 23. Rastaldi, M. P. et al. Glomerular monocyte- macrophage features in ANCA-positive renal vasculitis and cryoglobulinemic nephritis. J.Am. Soc. Nephrol. 11, 2036–2043 (2000). 24. Weidner, S., Carl, M., Riess, R. Rupprecht, H. D. Histologic analysis of renal leukocyte infiltration in antineutrophil cytoplasmic antibody-associated vasculitis: importance of monocyte and neutrophil infiltration in tissue damage. Arthritis Rheum. 50, 3651–3657 (2004). 25. Cunningham, M. A. et al. Prominence of cell- mediated immunity effectors in “pauci-immune” glomerulonephritis. J.Am. Soc. Nephrol. 10, 499–506 (1999). 26. Bajema, I. M., Hagen, E. C., de Heer, E., van der Woude, F. J. Bruijn, J. A. Colocalization of ANCA-antigens and fibrinoid necrosis in ANCA-associated vasculitis. Kidney Int. 60, 2025–2030 (2001). 27. Kessenbrock, K. et al. Netting neutrophils in autoimmune small-vessel vasculitis. Nat. Med. 15, 623–625 (2009). 28. Abreu-Velez, A. M., Smith, J. G. Jr Howard, M. S. Presence of neutrophil extracellular traps and antineutrophil cytoplasmic antibodies associated with vasculitides. North Am. J. Med. Sci. 1, 309–313 (2009). 29. Kambas, K. et al. Tissue factor expression in neutrophil extracellular traps and neutrophil derived microparticles in antineutrophil cytoplasmic antibody associated vasculitis may promote thromboinflammation and the thrombophilic state associated with the disease. Ann. Rheum. Dis. http://dx.doi.org/10.1136/ annrheumdis-2013-203430 (2013). 30. Lamprecht, P. Gross, W. L. Current knowledge on cellular interactions in the WG‑granuloma. Clin. Exp. Rheumatol. 25 (Suppl. 44), S49–S51 (2007). 31. Jennette, J. C. Falk, R. J. The rise and fall of horror autotoxicus and forbidden clones. Kidney Int. 78, 533–535 (2010). 32. Cui, Z., Zhao, M. H., Segelmark, M. Hellmark, T. Natural autoantibodies to myeloperoxidase, proteinase 3, and the glomerular basement membrane are present in normal individuals. Kidney Int. 78, 590–597 (2010). 33. Olson, S. W. et al. Asymptomatic autoantibodies associate with future anti-glomerular basement membrane disease. J.Am. Soc. Nephrol. 22, 1946–1952 (2011). 34. Xu, P. C., Cui, Z., Chen, M., Hellmark, T. Zhao, M. H. Comparison of characteristics of natural autoantibodies against myeloperoxidase and anti-myeloperoxidase autoantibodies from patients with microscopic polyangiitis. Rheumatology (Oxford) 50, 1236–1243 (2011). 35. Roth, A. J. et al. ANCA epitope specificity determines pathogenicity, detectability and clinical predictive value. J. Clin. Invest. 123, 1773–1783 (2013). 36. Olson, S. W. et al. Relation between asymptomatic proteinase 3 antibodies and future granulomatosis with polyangiitis. Clin. J. Am. Soc. Nephrol. 8, 1312–1318 (2013). 37. Popa, E. R., Stegeman, C. A., Kallenberg, C. G.  Tervaert, J. W. Staphylococcus aureus and Wegener’s granulomatosis. Arthritis Res. 4, 77–79 (2002). 38. Laudien, M. et al. Nasal carriage of Staphylococcus aureus and endonasal activity in Wegener’s granulomatosis as compared to rheumatoid arthritis and chronic rhinosinusitis with nasal polyps. Clin. Exp. Rheumatol. 28 (Suppl. 57), 51–55 (2010). 39. Davies, D. J., Moran, J. E., Niall, J. F. Ryan, G. B. Segmental necrotizing glomerulonephritis with antineutrophil antibody: possible arbovirus aetiology? Br. Med. J. (Clin. Res. Ed.) 285, 606 (1982). 40. Pendergraft, W. F. et al. Autoimmunity is triggered by cPR-3(105–201), a protein complementary to the autoantigen proteinase 3. Nat. Med. 10, 72–79 (2004). 41. Preston, G. A., Pendergraft, W. F. 3rd Falk, R. J. New insights that link microbes with the generation of antineutrophil cytoplasmic autoantibodies: the theory of autoantigen complementarity. Curr. Opin. Nephrol. Hypertens. 14, 217–222 (2005). 42. Heal, J. R., Roberts, G. W., Raynes, J. G., Bhakoo, A. Miller, A. D. Specific interactions between sense and complementary peptides: the basis for the proteomic code. Chembiochem. 3, 136–151 (2002). 43. Mekler, L. B. On the specific mutual interaction of amino acid residues of polypeptide chains and amino acid residues with codons. Oncology 27, 286–288 (1973). 44. Yang, J. J. et al. ANCA patients have T cells responsive to complementary PR‑3 autoantigen. Kidney Int. 74, 1159–1169 (2008). 45. Badhwar, A. K., Simmons, J., Jennette, J. C., Falk, R. J. Preston, G. A. Characterization of an antisense transcript from the proteinase‑3 gene locus in patients with ANCA-vasculitis utilizing strand-specific RT‑PCR and 5'RACE [abstract]. J. Am. Soc. Nephrol. 19, 188A (2008). 46. Cao, Y. et al. DRB1*15 allele is a risk factor for PR3-ANCA disease in African Americans. J.Am. Soc. Nephrol. 22, 1161–1167 (2011). 47. Shoenfeld, Y. Idiotypic induction of autoimmunity: a new aspect of the idiotypic network. FASEB J. 8, 1296–1301 (1994). 48. Yang, J. J. et al. Circumvention of normal constraints on granule protein gene expression in peripheral blood neutrophils and monocytes of patients with antineutrophil cytoplasmic autoantibody-associated glomerulonephritis. J. Am. Soc. Nephrol. 15, 2103–2114 (2004). 49. Ciavatta, D. J. et al. Epigenetic basis for aberrant upregulation of autoantigen genes in humans with ANCA vasculitis. J. Clin. Invest. 120, 3209–3219 (2010). 50. Durant, S. et al. Apoptosis-induced proteinase 3 membrane expression is independent from degranulation. J. Leukoc. Biol. 75, 87–98 (2004). 51. Kantari, C. et al. Proteinase 3, the Wegener autoantigen, is externalized during neutrophil apoptosis: evidence for a functional association with phospholipid scramblase 1 and interference with macrophage phagocytosis. Blood 110, 4086–4095 (2007). 52. Morgan, M. D. et al. Patients with Wegener’s granulomatosis demonstrate a relative deficiency and functional impairment of T‑regulatory cells. Immunology 130, 64–73 (2010). 53. Rimbert, M. et al. Decreased numbers of blood dendritic cells and defective function of regulatory T cells in antineutrophil cytoplasmic antibody-associated vasculitis. PLoS ONE 6, e18734 (2011). 54. Free, M. E. et al. Patients with antineutrophil cytoplasmic antibody-associated vasculitis have defective TREG cell function exacerbated by the presence of a suppression-resistant effector cell population. Arthritis Rheum. 65, 1922–1933 (2013). 55. Wilde, B et al. Aberrant expression of the negative costimulator PD‑1 on T cells in granulomatosis with polyangiitis. Rheumatology (Oxford) 7, 1188–1197 (2012). 56. Bunch, D. O. et al. Decreased CD5+ B cells in active ANCA vasculitis and relapse after rituximab. Clin. J. Am. Soc. Nephrol. 8, 382–391 (2013). 57. Wilde B. et al. Regulatory B cells in ANCA- associated vasculitis. Ann. Rheum. Dis. 72, 1416–1419 (2013). 58. Scapini, P., Bazzoni, F. Cassatella, M. A. Regulation of B‑cell‑activating factor (BAFF)/B lymphocyte stimulator (BLyS) expression in human neutrophils. Immunol. Letters 116, 1–6 (2008). 59. Krumbholz, M. et al. BAFF is elevated in serum of patients with Wegener’s granulomatosis. J. Autoimmun. 25, 298–302 (2005). 60. Sanders, J. S., Huitma, M. G., Kallenberg, C. G. Stegeman, C. A. Plasma levels of soluble interleukin 2 receptor, soluble CD30, interleukin 10 and B cell activator of the tumour necrosis factor family during follow-up in vasculitis associated with proteinase 3‑antineutrophil cytoplasmic antibodies: associations with disease activity and relapse. Ann. Rheum. Dis. 65, 1484–1489 (2006). 61. Nagai, M. et al. Serum levels of BAFF and APRIL in myeloperoxidase anti-neutrophil cytoplasmic autoantibody-associated renal vasculitis: association with disease activity. Nephron Clin. Pract. 118, c339–c345 (2011). 62. Bader, L., Koldingsnes, W. Nossent, J. B‑lymphocyte activating factor levels are increased in patients with Wegener’s granulomatosis and inversely correlated with ANCA titer. Clin. Rheumatol. 29, 1031–1035 (2010). 63. Falk, R. J., Terrell, R. S., Charles, L. A. Jennette, J. C. Anti-neutrophil cytoplasmic autoantibodies induce neutrophils to degranulate and produce oxygen radicals in vitro. Proc. Natl Acad. Sci. USA 87, 4115–4119 (1990). 64. Huugen, D. et al. Aggravation of anti- myeloperoxidase antibody induced glomerulonephritis by bacterial lipopolysaccharide: role of tumor necrosis factor α. Am. J. Pathol. 167, 47–58 (2005). 65. Schreiber, A. et al. C5a receptor mediates neutrophil activation and ANCA-induced glomerulonephritis. J. Am. Soc. Nephrol. 20, 289–298 (2009). 66. Porges, A. J. et al. Anti-neutrophil cytoplasmic antibodies engage and activate human neutrophils via Fcγ RIIa. J. Immunol. 153, 1271–1280 (1994). 67. Kocher, M., Siegel, M. E., Edberg, J. C. Kimberly, R. P. Cross-linking of Fcγ receptor IIa and Fcγ receptor IIIb induces different proadhesive phenotypes on human neutrophils. J. Immunol. 159, 3940–3948 (1997). 68. Kettritz, R., Jennette, J. C. Falk, R. J. Cross- linking of ANCA-antigens stimulates superoxide release by human neutrophils. J. Am. Soc. Nephrol. 8, 386–394 (1997). 69. Williams, J. M. et al. Activation of the Gi heterotrimeric G protein by ANCA IgG F(ab')2 fragments is necessary but not sufficient to stimulate the recruitment of those downstream mediators used by intact ANCA IgG. J. Am. Soc. Nephrol. 14, 661–669 (2003). 70. Kettritz, R., Schreiber, A., Luft, F. C. Haller, H. Role of mitogen-activated protein kinases in activation of human neutrophils by antineutrophil cytoplasmic antibodies. J.Am. Soc. Nephrol. 12, 37–46 (2001). REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 11. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  AUGUST 2014  |  473 71. van der Veen, B. S. et al. Effects of p38 mitogen- activated protein kinase inhibition on anti- neutrophil cytoplasmic autoantibody pathogenicity in vitro and in vivo. Ann. Rheum. Dis. 70, 356–365 (2011). 72. Hao, J., Meng, L. Q., Xu, P. C., Chen, M. Zhao, M. H. p38MAPK, ERK and PI3K signaling pathways are involved in C5a-primed neutrophils for ANCA-mediated activation. PLoS ONE 7, e38317 (2012). 73. Charles, L. A., Caldas, M. L., Falk, R. J., Terrell, R. S. Jennette, J. C. Antibodies against granule proteins activate neutrophils in vitro. J. Leukoc. Biol. 50, 539–546 (1991). 74. Csernok, E., Ernst, M., Schmitt, W., Bainton, D. F. Gross, W. L. Activated neutrophils express proteinase 3 on their plasma membrane in vitro and in vivo. Clin. Exp. Immunol. 95, 244–250 (1994). 75. Ewert, B. H., Jennette, J. C. Falk, R. J. Anti-myeloperoxidase antibodies stimulate neutrophils to damage human endothelial cells. Kidney Int. 41, 375–383 (1992). 76. Savage, C. O., Gaskin, G., Pusey, C. D. Pearson, J. D. Myeloperoxidase binds to vascular endothelial cells, is recognized by ANCA and can enhance complement dependent cytotoxicity. Adv. Exp. Med. Biol. 336, 121–123 (1993). 77. Ewert, B. H., Becker, M. E., Jennette, J. C. Falk, R. J. Antimyeloperoxidase antibodies induce neutrophil adherence to cultured human endothelial cells. Ren. Fail. 17, 125–133 (1995). 78. Lu, X., Garfield, A., Rainger, G. E., Savage, C. O.  Nash, G. B. Mediation of endothelial cell damage by serine proteases, but not superoxide released from antineutrophil cytoplasmic antibody-stimulated neutrophils. Arthritis Rheum. 54, 1619–1628 (2006). 79. Gupta, A. K. et al. Activated endothelial cells induce neutrophil extracellular traps and are susceptible to NETosis-mediated cell death. FEBS Letters 584, 3193–3197 (2010). 80. Casselman, B. L., Kilgore, K. S., Miller, B. F.  Warren, J. S. Antibodies to neutrophil cytoplasmic antigens induce monocyte chemoattractant protein‑1 secretion from human monocytes. J. Lab. Clin. Med. 126, 495–502 (1995). 81. Ralston, D. R., Marsh, C. B., Lowe, M. P. Wewers, M. D. Antineutrophil cytoplasmic antibodies induce monocyte IL‑8 release. Role of surface proteinase‑3, α1-antitrypsin, and Fcγ receptors. J. Clin. Invest. 100, 1416–1424 (1997). 82. Muller Kobold, A. C., Kallenberg, C. G. Tervaert, J. W. Monocyte activation in patients with Wegener’s granulomatosis. Ann. Rheum. Dis. 58, 237–245 (1999). 83. Arimura, Y. et al. The role of myeloperoxidase and myeloperoxidase-antineutrophil cytoplasmic antibodies (MPO-ANCAs) in the pathogenesis of human MPO‑ANCA‑associated glomerulonephritis. Clin. Exp. Nephrol. 17, 634–637 (2013). 84. Xiao, H. et al. Antineutrophil cytoplasmic autoantibodies specific for myeloperoxidase cause glomerulonephritis and vasculitis in mice. J. Clin. Invest. 110, 955–963 (2002). 85. Jennette, J. C., Xiao, H., Falk, R. Gasim, A. M. Experimental models of vasculitis and glomerulonephritis induced by antineutrophil cytoplasmic autoantibodies. Contrib. Nephrol. 169, 211–220 (2011). 86. van Timmeren, M. M. Heeringa, P. Pathogenesis of ANCA-associated vasculitis: recent insights from animal models. Curr. Opin. Rheumatol. 24, 8–14 (2012). 87. Salama, A. D. Little, M. A. Animal models of antineutrophil cytoplasm antibody-associated vasculitis. Curr. Opin. Rheumatol. 24, 1–7 (2012). 88. van der Geld, Y. M. et al. Rats and mice immunized with chimeric human/mouse proteinase 3 produce autoantibodies to mouse Pr3 and rat granulocytes. Ann. Rheum. Dis. 66, 1679–1682 (2007). 89. Pfister, H. et al. Antineutrophil cytoplasmic autoantibodies against the murine homolog of proteinase 3 (Wegener autoantigen) are pathogenic in vivo. Blood 104, 1411–1418 (2004). 90. Primo, V. C. et al. Anti-PR3 immune responses induce segmental and necrotizing glomerulonephritis. Clin. Exp. Immunol. 159, 327–337 (2010). 91. Little, M. A. et al. Anti-proteinase 3 anti-neutrophil cytoplasm autoantibodies recapitulate systemic vasculitis in mice with a humanized immune system. PLoS ONE 7, e28626 (2012). 92. Xiao, H. et al. The role of neutrophils in the induction of glomerulonephritis by anti- myeloperoxidase antibodies. Am. J. Pathol. 167, 39–45 (2005). 93. Schreiber, A., Xiao, H., Falk, R. J. Jennette, J. C. Bone marrow-derived cells are sufficient and necessary targets to mediate glomerulonephritis and vasculitis induced by anti-myeloperoxidase antibodies. J. Am. Soc. Nephrol. 17, 3355–3364 (2006). 94. Xiao, H. et al. Genetically determined severity of anti-myeloperoxidase glomerulonephritis. Am. J. Pathol. 8, 139–160 (2013). 95. van Timmeren, M. M. et al. IgG glycan hydrolysis attenuates ANCA-mediated glomerulonephritis. J. Am. Soc. Nephrol. 21, 1103–1114 (2010). 96. Xiao, H., Schreiber, A., Heeringa, P., Falk, R. J. Jennette, J. C. Alternative complement pathway in the pathogenesis of disease mediated by antineutrophil cytoplasmic autoantibodies. Am. J. Pathol. 170, 52–64 (2007). 97. Huugen, D. et al. Inhibition of complement factor C5 protects against anti-myeloperoxidase antibody-mediated glomerulonephritis in mice. Kidney Int. 71, 646–654 (2007). 98. Xiao, H. et al. C5a receptor (CD88) blockade protects against MPO-ANCA glomerulonephritis. J. Am. Soc. Nephrol. 25, 225–231 (2014). 99. Xing, G. Q. et al. Complement activation is involved in renal damage in human antineutrophil cytoplasmic autoantibody associated pauci-immune vasculitis. J. Clin. Immunol. 29, 282–291 (2009). 100. Yuan, J. et al. C5a and its receptors in human anti-neutrophil cytoplasmic antibody (ANCA)- associated vasculitis. Arthritis Res.Ther. 14, R140 (2012). 101. Gou, S. J., Yuan, J., Chen, M., Yu, F. Zhao, M. H. Circulating complement activation in patients with anti-neutrophil cytoplasmic antibody-associated vasculitis. Kidney Int. 83, 129–137 (2012). 102. Klinger, H. Grenzformen der periarteriitis nodosa [German]. Frankf. Z. Pathol. 42, 455–480 (1931). 103. Wegener, F. Über generalisierte septische Gefässerkrankungen [German]. Verh. Deut. Pathol. Ges. 29, 202–210 (1936). 104. Wegener, F. Über eine eigenartige rhinogene Granulomatose mit besonderer Beteiligung des Arteriensystems und der Nieren [German]. Beitr. Pathol.Anat. 102, 30–68 (1939). 105. Churg, J. Strauss, L. Allergic granulomatosis, allergic antiitis, and periarteritis nodosa. Am. J. Pathol. 27, 277–294 (1951). 106. Petersen, H. J. Smith, A. M. The role of the innate immune system in granulomatous disorders. Front. Immunol. 4, 120 (2013). 107. Voswinkel, J., Müller, A. Lamprecht, P. Is PR3-ANCA formation initiated in Wegener’s granulomatosis lesions? Granulomas as potential lymphoid tissue maintaining autoantibody production. Ann. NY Acad. Sci. 1051, 12–19 (2005). Acknowledgements The authors are supported by a grant from the NIH National Institute of Diabetes and Digestive and Kidney Diseases (P01-DK058335‑11). Author contributions J.C.J. researched data for the article, and both authors contributed equally to discussions of content, writing and review/editing of the manuscript before submission. FOCUS ON VASCULITIS © 2014 Macmillan Publishers Limited. All rights reserved