Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Reviews

The role of neutrophil extracellular


traps in rheumatic diseases
Falko Apel, Arturo Zychlinsky* and Elaine F. Kenny*
Abstract | Rheumatic diseases are a collection of disorders defined by the presence of
inflammation and destruction of joints and internal organs. A common feature of these diseases
is the presence of autoantibodies targeting molecules commonly expressed in neutrophils.
These preformed mediators are released by neutrophils but not by other immune cells such as
macrophages. Neutrophils, major players in the host innate immune response, initiate a cell death
mechanism termed neutrophil extracellular trap (NET) formation as a way to ensnare pathogens.
NETs are also a source of released self-​molecules found in rheumatic diseases. Subsequently ,
research on the role of NETs in the onset, progression and resolution of inflammation in rheumatic
diseases has intensified. This Review has two aims. First, it aims to highlight the mechanisms
required for the generation of NETs, the research landscape of which is rapidly changing. Second,
it aims to discuss the role of neutrophils and NETs in systemic lupus erythematosus, vasculitis
(specifically anti-​neutrophil cytoplasmic autoantibody-associated vasculitis), rheumatoid arthritis
and gout. Our goal is to clarify the field of NET research in rheumatic diseases in the hope of
improving the therapeutic approaches utilized for these diseases.

Rheumatic diseases are a set of inflammatory disor- cytoplasm and a lobulated nucleus; hence, along with
ders that affect tissues and joints1. A hallmark of these other granulocytic immune cells such as eosinophils
diseases is the generation of antibodies that recognize and basophils, they are often defined as granulocytes.
self-​molecules usually residing within cells. These Neutrophil development is well characterized (reviewed
autoantibodies are produced as a result of a break- elsewhere6). Owing to the unusual form of their nucleus
down in self-​tolerance, and they cause inflammation they are also known as polymorphonuclear cells but we
and tissue destruction in affected areas of the body2. will use the term neutrophil throughout. Neutrophils
Much research has been geared towards identifying the have a particularly short lifespan, which is a surprising
source of the self-​molecules against which these auto­ and unexplained trait; this short lifespan and their gen-
antibodies are generated. The discovery that neutrophils eral abundance mean that a healthy adult is estimated to
undergo NETosis3 (a defence and/or cell death mecha- produce one billion neutrophils per day7,8.
nism by which neutrophils release their contents) led Neutrophils produce cytokines (for example, IL-8)
to the identification of these innate immune cells as a that recruit more neutrophils or further inflammatory
potential major source of self-​molecule release in rheu- mediators, such as TNF9. They also secrete cytokines
matic disease development4. As research into both neu- that dampen the inflammatory response, presumably
trophil signalling mechanisms, including NETosis, and when the microbial threat has abated9. Neutrophils
the development and progression of rheumatic disease induce cytokine production to a lower level than other
is an ever changing landscape, we hope to clarify the myeloid cells, such as macrophages or dendritic cells;
current state of the field and aid in the advancement of however, as neutrophils are so abundant in inflamma-
our knowledge of and ability to diagnose and treat these tory sites, individual small contributions are potentially
Department of Cellular diseases. amplified9.
Microbiology, Max Planck Neutrophils receive pro-​inflammatory instructions
Institute for Infection Biology, Neutrophils from cytokines and chemokines and rapidly leave the
Berlin, Germany.
Neutrophils are the most abundant white blood cell in circulation and arrive at inflammatory sites by a care-
*e-​mail: zychlinsky@
the circulation. They develop in the bone marrow and fully orchestrated process. Upon arrival, these cells
mpiib-​berlin.mpg.de;
kenny@mpiib-​berlin.mpg.de once released into the circulation they patrol for patho- deploy their effector functions to kill microorganisms
https://doi.org/10.1038/ gens and are considered the host’s first line of immune and signal to the immune system to promote or dimin-
s41584-018-0039-z defence5. These myeloid cells have a richly granular ish the influx of cells to the site6. At the inflammatory

NATure RevIewS | RheumaToLogy volume 14 | AUGUST 2018 | 467


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Key points
proteases and recognize a variety of substrates. After
initial activation of the neutrophils, cyclin-​dependent
• Molecules released during neutrophil extracellular trap (NET) formation can often kinase 6 (CDK6) is also activated, probably permitting
become autoantigens in systemic lupus erythematosus, small vessel vasculitis, the vesiculation of the nuclear membrane14. Importantly,
rheumatoid arthritis and gout. NETs expose several enzymes such as myeloperoxidase,
• Damaged tissues and organs in these rheumatic diseases are prone to containing neutrophil elastase, cathepsin G, PR3 and LL37 as well as
NET-​derived material. histones to the extracellular space. These, and other pro-
• Neutrophils from patients with some rheumatic diseases spontaneously undergo teins, enable NETs to fulfil their function in microbial
NETosis in vitro, reflecting the pre-​activation of neutrophils during chronic defence, coagulation and immune activation. ROS are
inflammation.
also released during NETosis.
• To date, whether NET formation is deleterious to progression of all rheumatic diseases Not all NET inducers utilize this classical pathway to
is unclear.
NETosis. For example, NET production in response
to the calcium ionophore A23187 and nigericin lacks
the requirements of classical NET formation (described
site, neutrophils are further activated and can degran- elsewhere11).
ulate, phagocytose or produce cytokines. These cells Citrullination of histones via protein arginine deimi-
contain several types of granules that store defined and nase (PAD) enzymes occurs during some forms of
overlapping cargoes of enzymes and antimicrobial pep- NETosis. However, the role of citrullination is unclear
tides. Granule fusion with the plasma membrane exter- in NET production. Human neutrophils do not require
nalizes the cargo in a process termed degranulation. PADs to generate NETs, but proteins in them are often
This process is well characterized in vitro, although citrullinated in response to many stimuli11; however,
confirming it in vivo is challenging. Neutrophils are further work is needed to definitively identify the role
extraordinarily efficient phagocytes; they recognize PADs have in NETosis. These citrullinated proteins are
microorganisms directly or through receptors that rec- common in rheumatic disease, so the source of these
ognize complement or antibodies and engulf particles modified proteins is of great therapeutic interest11.
by classical phagocytosis into vacuoles. Granules then Much work on citrullination and the role of PAD4
fuse to these vacuoles and unload their contents. The has used the PAD4-deficient mouse; however, major
NADPH oxidase complex is also recruited to the surface differences seem to exist in the way human and mouse
of the vacuole, where it produces reactive oxygen spe- neutrophils induce NETosis7. For example, although
cies (ROS), for example, superoxide. The combination neutrophil elastase is required for NET formation in
of ROS, acidification and the many enzymes and anti- humans, mice deficient in neutrophil elastase can make
microbial peptides that come from the granule makes NETs15, but mice in which both neutrophil elastase
the phagolysosome an inhospitable environment for and PR3 are deleted can not14. Another major differ-
microorganisms10. ence between human and mouse neutrophils is the
total number in circulation, with neutrophils making
Neutrophil extracellular traps up 70% of white blood cells in the human circulation
Another aspect to the defensive arsenal of neutrophils is compared with only 30% in mice7. Differences also exist
neutrophil extracellular trap (NET) formation3 (Fig. 1). in the binding abilities of the Fc receptors expressed on
NETs are defined as extracellular structures that contain human and mouse neutrophils16 and in their ability to
DNA, histones and neutrophil proteins such as myelo­ react to infection. All of these factors could contribute
peroxidase, neutrophil elastase or calgranulin. During to the difference seen in NETosis induction between
NET formation, neutrophils undergo chromatin remod- the species.
elling via processing of histones by proteases, and the Neutrophils are deployed to inflammatory sites
remodelled chromatin subsequently binds antimicrobial where they release molecules that are toxic to micro-
proteins and enzymes. The modified chromatin is then organisms but also to the host17. Neutrophils are cul-
released through a process termed NETosis. NETs occur pable for inducing collateral damage in the process of
in inflammatory sites and probably have a short lifespan antimicrobial defence. It is well documented that neu-
owing to degradation by local DNases. Hence, detecting trophil proteases and ROS can destroy healthy tissue18.
and quantifying NETs is tricky (Box 1). Interestingly, extracellular histones are potent antimicro-
Several pathways result in NET release11. Classically, bials, as described more than 50 years ago by Hirsch19,
NETosis requires proteases and the generation of ROS. and can also kill host cells, and they are implicated in
A complex of proteins, called the ‘azurosome’, senses many pathologies. Furthermore, the externalization
hydrogen peroxide through myeloperoxidase, which of neutrophil proteins as well as histones and DNA, as
converts this ROS into halic acids12. Myeloperoxidase occurs in NETosis, provides a source for antigen pres-
activation leads to the permeabilization of neutro- entation in the development of different rheumatic dis-
phil granules and the dissociation of the azurosome. eases. NETs are also immune-​stimulants that exacerbate
This process results in the release of serine proteases, inflammation and activate the acquired immune system.
including neutrophil elastase, which translocate to the Furthermore, the role of NETs in thrombus formation, a
nucleus where they clip histones13. This proteolytic activ- complication of rheumatic diseases20, adds another link
ity decondenses the chromatin, which is necessary for between NETosis and autoimmunity.
NET formation. Neutrophil elastase, cathepsin G and In the following sections we focus on the role of NETs
proteinase 3 (PR3) belong to the same family of serine in the development and progression of systemic lupus

468 | AUGUST 2018 | volume 14 www.nature.com/nrrheum


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

C D mechanism might be the opsonization of NETs by


complement factors37 and their further complement-​
receptor-mediated uptake and presentation by follicular
dendritic cells to B cells38. Neutrophils themselves might
serve as antigen-​presenting cells; in mice, neutrophils
have been shown to transport antigens to the sites of
T cell activation39,40 and function as antigen-​presenting
cells to T cells41. Although none of the above-​mentioned
mechanisms have been described for SLE specifically,
they are potential explanations for how NETs could
contribute to antibody-​dependent autoimmunity.
In patients with SLE, autoantibodies are directed
to nuclear antigens, such as double-stranded DNA
(dsDNA) and chromatin, and to proteins such as his-
Fig. 1 | Neutrophil extracellular trap induction in human neutrophils. a | Unstimulated tones, myeloperoxidase, PR3 and neutrophil elastase42–47;
primary human neutrophils. b | Primary human neutrophils treated with 50 nM phorbol all these antigens are NET components. Interestingly,
myristate acetate for 3 hours. Cells were then fixed and stained for neutrophil elastase anti-​NET autoantibodies induce NET formation in
(green) and chromatin (red) with antibodies and for DNA (blue) with Hoechst. Scale bar is IFNα-​primed neutrophils30,31,48 and can perpetuate
20 µm. Image courtesy of V. Brinkmann, Max Planck Institute for Infection Biology , Germany. immune activation. Furthermore, immune complexes,
probably formed between autoantibodies and NET com-
ponents, directly contribute to the development of LN.
erythematosus (SLE), vasculitis, rheumatoid arthritis The complexes deposit along the glomerular capillary of
(RA) and gout. the kidney, inducing necrosis and kidney destruction, as
seen in vasculitis49.
Systemic lupus erythematosus
SLE is a multifactorial autoimmune disease affecting DNase1. Normally, NETs are degraded by endo­
different organs, and is most prevalent in women of nucleases27 and DNase1-like proteins in the circula-
childbearing age21. Its pathogenesis combines dysregu- tion. DNase1 activity negatively correlates with disease
lated innate and adaptive immune responses, culminat- activity and the concentration of serum markers such as
ing in a lack of tolerance to self-​antigens. The molecular antinuclear antibodies, anti-​dsDNA antibodies and anti-​
hallmarks of SLE are the formation of autoantibodies nucleosome antibodies in SLE50. Missense mutations in
(commonly antinuclear antibodies) and upregulation the genes encoding DNase1 (ref.51) and DNase1L3 (also
of type I interferon-​regulated genes. Patients with SLE known as DNase gamma52) cause familial forms of SLE,
are at increased risk of developing atherosclerosis and whereas mutations in DNases lead to lupus-like disease
cardiovascular symptoms compared with the general in mice53,54.
population22,23. Another major complication is lupus Additional DNase1-related mechanisms reduce NET
nephritis (LN), which can lead to symptoms ranging degradation in SLE. NETs contain oxidized DNA in the
from mild proteinuria to acute kidney failure24. form of 8-hydroxyguanosine, which protects nucleic
Dating back to the 1940s, it was observed that neu- acids from degradation, increasing exposure of the NETs
trophils and macrophages phagocytose nuclear mate- to autoantigens and thus triggering an autoimmune
rial opsonized by autoantibodies. This so-​called LE response55. NET-​binding proteins, such as antibodies
(lupus erythematosus) cell phenomenon was used to or complement component C1q, protect the NETs from
diagnose SLE25 and might reflect an increase in NET degradation, possibly by inhibiting DNase1 (refs27,37).
formation in these patients or reduced clearance of Additionally, sera from patients with SLE contain pro-
nuclear material. Although 40–60% of patients with teins that inhibit DNase1, including globular actin
SLE are neutropenic26, increasing evidence shows that (G-​ a ctin)56 and anti-​DNase1 antibodies57,58. Taken
neutrophils and NETs contribute to SLE pathogenesis. together, the evidence suggests that in SLE NET degra-
NETs are detectable in the skin and in the glomeruli dation is hampered, leading to prolonged exposure of
of patients with SLE27–29. In vitro experiments suggest autoantigens to the immune system.
that SLE neutrophils make NETs spontaneously28,30 and
upon stimulation with antibodies against LL37 (ref.30) Type I interferons. Type I interferons have an important
or ribonuclear particles31. role in SLE pathogenesis59–62. In vitro, NETs induce pro-
In healthy individuals, phagocytes clear apoptotic neu- duction of type I interferons in plasmacytoid dendritic
trophils32 and NETs33. This clearance is immunologically cells. The NETs are endocytosed with the help of LL37
silent and without the production of pro-​inflammatory and high mobility group B1 (HMGB1), and activate
cytokines33. However, in SLE, an increased number of Toll-​like receptor 9 (TLR9)31. In addition, both oxidized
apoptotic neutrophils and NETs is observed, while their mitochondrial DNA and genomic NET DNA stimulate
clearance by macrophages is impaired31,34,35. This lack interferons31,48,63,64. Interestingly, TLR9-deficient mice, in
of clearance provides antigens for autoantibodies recog- an MRL.FASlpr mouse model of SLE, produce increased
nizing neutrophil proteins36. How NET material reaches amounts of autoantibodies and display a severe renal
the germinal centres to contribute to antibody-​dependent phenotype65,66. NETs and their components also induce
autoimmunity is not fully understood. One potential IL-1β production by activating the inflammasome67.

NATure RevIewS | RheumaToLogy volume 14 | AUGUST 2018 | 469


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 1 | Looking for neutrophil extracellular traps


different treatment regimens, and further studies are
needed to achieve a conclusive result.
The detection of neutrophil extracellular traps (NETs) in vivo at sites of infection or injury In summary, the ineffective clearance of NETs in
is aided by DNA dyes143,144. This technique is, however, restricted to experimental animals patients with SLE exposes self-​molecules to the immune
and is limited by a lack of markers for other NET components. Hence, animal models system and contributes to the development of autoan-
in which other NET markers can be observed are needed to aid our understanding
tibodies and pro-​inflammatory cytokines, driving the
of NETosis in vivo.
NETs can be readily detected in tissue sections of both experimental animals and
pathogenesis of SLE (Fig. 2a).
pathological samples from patients. Indeed, standard paraffin sections, commonly used
in clinical pathology laboratories, can be used to detect NETs by immunofluorescence. Vasculitis
Standard technologies combining a DNA dye and commercially available antibodies The term vasculitis is used for a group of rare chronic
that detect histones and neutrophil proteins (including neutrophil elastase and diseases that present with inflammation of blood
myeloperoxidase) are useful for detecting the presence of NETs145. Furthermore, vessels81, which is sometimes accompanied by necro­
automatic microscopy methods can be used to quantify the juxtaposition of the three tizing cell death. Although the direct cause of these
elements of NETs (DNA, histones and neutrophil proteins) and determine the level of diseases is unclear, the presence of autoantibodies82 and
NET formation in a specific sample. pro-​inflammatory events (for example, infections83) have
NETs can also be detected in fluids such as serum or sputum146. Different techniques
an important role. A proportion of these vasculitides are
can be used to achieve this detection, many of which are quantitative. A useful tool that
can be implemented in most laboratories is a ‘sandwich’ enzyme-​linked immunosorbent
termed small vessel vasculitis, in which neutrophils have
assay. An antibody against histones is used to capture NETs, and a second antibody, a detrimental role and damage organs84,85.
which detects a neutrophil protein, is used to identify the presence of NETs in the Small vessel vasculitis is a relapsing-​remitting
sample. Given that only NETs contain both histones and neutrophil proteins, specific chronic autoimmune disease characterized by dam-
detection of NETs is guaranteed147. The automation capacity and precision of this age to small blood vessels. A portion of patients with
approach could be exploited in the clinic to determine NET formation in samples small vessel vasculitis present with anti-​neutrophil
from patients. cytoplasmic auto­antibody (ANCA)-associated vascu-
litis (AAV). More than 90% of these patients produce
autoantibodies against proteins located in the cytoplasm
Low-​density granulocytes. Interestingly, most patients of neutrophils85. These ANCAs are mainly generated
with SLE present a subset of neutrophil-​like cells termed against PR3 and myeloperoxidase86–88. AAV can be
low-​density granulocytes (LDGs)68 that separate with further divided into granulomatosis with polyangiitis,
the peripheral blood mononuclear cell fraction in a microscopic poly­angiitis and eosinophilic granulomato-
density gradient69,70. These cells serve as a marker of sis with polyangiitis89. In these diseases, ANCA damage
juvenile SLE71, and their occurrence correlates with is present in small-​sized and medium-​sized blood vessels
skin involvement and vasculitis in patients with SLE68. and other tissues including the lung, upper respiratory
LDGs produce increased amounts of IL-6, IL-8, TNF tract and kidney.
and type I interferons68 and spontaneously make NETs, Stimulation of neutrophils with ANCAs induces
which damage the endothelium28. Although the origin ROS production and degranulation90, and exposure of
and maturity state of these cells require further investi- neutrophils primed with TNF to ANCAs stimulates
gation, these findings suggest that LDGs are important NETosis91. Patients with AAV with anti-​myeloperoxidase
contributors to the pathogenesis of SLE. antibodies in the circulation also have evidence of NETs
in their glomeruli91. Neutrophils isolated from patients
Mouse studies. Further evidence for the role of NETs in with AAV produce NETs spontaneously92. This process,
SLE progression also comes from mouse studies. DNase1 in turn, increases the levels of NET products in the
injection ameliorates disease severity in the NZBW circulation92. Indeed, dead and dying neutrophils on
mouse model of SLE72 but not in human patients73. We the wall of damaged small blood vessels are a histological
suggest that this discrepancy results from the timing of marker of this disease85.
the initiation of treatment or the presence of DNase1 In 2010, two whole-​genome gene expression profiling
inhibitors in humans. Mice deficient in NADPH oxidase 2 studies identified the presence of a granulocyte signature
(NOX2) present with increased autoantibody produc- in blood from patients with AAV93,94. Subsequent work
tion and a severe renal phenotype74–76. Furthermore, confirmed the source of this signature to be the presence
NOX2 inhibitors mitigate pristine-​induced lupus, a dis- of LDGs (a similar finding to that in patients with SLE95).
ease model in which LDGs are also observed77. A poten- These cells spontaneously produce NETs containing
tial explanation for these seemingly paradoxical results PR3 and myeloperoxidase. The presence of LDGs in the
might be either that autoimmune-​related NETs are pro- blood of patients with AAV is associated with increased
duced independently of NOX2 or that the blockage of disease activity and a decreased response to treatment
ROS-​dependent NET formation results in another form with rituximab95.
of cell death, providing the required antigens. Mouse models of vasculitis and kidney damage sup-
The effect of PAD4 inhibition on SLE progression port the idea that preventing NETosis or neutralizing
in mouse models is still under debate. Although two the molecules released from NETs is beneficial in treat-
studies78,79 reported an ameliorated disease outcome in ing this disease. Co-​incubation of pro-​inflammatory
Cl-​amidine (a PAD inhibitor)-treated lupus-​prone mice, subcutaneously elicited neutrophils with myeloid
a study combining Cl-​amidine treatment and genetic dendritic cells (mDCs) resulted in the uptake of PR3
depletion of PAD4 in MRL.Faslpr lupus-​prone mice did and myeloperoxidase into the mDCs; immunization
not observe an effect80. This discrepancy might reflect of mice with these mDCs resulted in the generation of

470 | AUGUST 2018 | volume 14 www.nature.com/nrrheum


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

C5.' D8CUEWNKVKU
'HHGEVQPFKUGCUG
'HHGEVQPPGWVTQRJKNUCPFQT0'6U
'HHGEVWPMPQYP
-KFPG[
FGUVTWEVKQP
+OOWPG
#WVQCPVKDQFKGU EQORNGZGU

0'6UKP
6[RG+ INQOGTWNK
R&% QHMKFPG[
KPVGTHGTQPU
$EGNN #WVQCPVKDQFKGU

&0CUG

0'/21
&0#CPF.. 5RQPVCPGQWU 5RQPVCPGQWU0'6U 0'/21CPF&0#
0'6U
.&)U

F)QWV E4#

/57ET[UVCNU


0'CPF/21

#WVQCPVKDQFKGU
CICKPUVEKVTWNNKPCVGF
CPVKIGPU

0GWVTQRJKNUKPLQKPV 5[PQXKCNȱWKF

Fig. 2 | The role of neutrophil extracellular traps in rheumatic diseases. Neutrophil extracellular trap (NET)
components are detected in the circulation and joints of patients with rheumatic diseases. a | In systemic lupus
erythematosus (SLE), DNase1 fails to degrade NETs spontaneously produced by low-​density granulocytes (LDGs).
Increased exposure induces autoantibody production by autoreactive B cells. Autoantibodies form immune complexes
with their respective antigen, which accumulate in the glomeruli of the kidney , leading to kidney destruction. Furthermore,
immune complexes activate plasmacytoid dendritic cells (pDCs), potentially via Toll-​like receptor 9 (TLR9), to produce
type I interferons and drive an inflammatory feedforward loop, causing more NET formation. b | Patients with vasculitis
present with autoantibodies against NET proteins, and NETs are detected in kidney lesions. c | In rheumatoid arthritis (RA),
citrullinated NET proteins are recognized by autoantibodies, which can be utilized for diagnosis. Neutrophils are also
known to infiltrate into the affected joint. d | In gout, NETs are also found in the affected joint, where they may aggregate
and degrade cytokines. MPO, myeloperoxidase; MSU, monosodium urate, NE, neutrophil elastase.

ANCAs, anti-​dsDNA antibodies and renal damage96. to the accumulation of NET components. In conclusion,
This finding also occurred in biopsy samples from skin the role of NETs in AAV is detrimental, as diagnosis is
lesions of patients with microscopic polyangiitis96. In based on the presence of ANCAs, and NETs provide the
another mouse model, glomerulonephritis (a common source of these antigens (Fig. 2b).
feature of AAV) was reduced by blocking extracellular
histones, again suggesting the importance of NET com- Rheumatoid arthritis
ponent release in disease development97. Finally, a PAD RA is a chronic immune-​mediated systemic disease
inhibitor reduced the titre of anti-myeloperoxidase anti- with both genetic and environmental contributing fac-
bodies in a mouse model of myeloperoxidase-ANCA- tors. RA is characterized by synovial joint inflammation,
associated vasculitis98. Further evidence for the detri- bone destruction and the presence of autoantibodies.
mental role of NETs in AAV comes from a study99 in Onset of RA can be separated into two stages. First,
which a mouse deficient in both DNase1 and DNase3 there is a preclinical asymptomatic stage that can last
(also known as TREX1) had vascular occlusion owing for several years. During this stage, biomarkers, such as

NATure RevIewS | RheumaToLogy volume 14 | AUGUST 2018 | 471


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

autoantibodies against rheumatoid factor (RF, an anti- a transcription factor involved in ROS generation),
body targeting the Fc chain of immunoglobulin G (IgG) indirectly implicating NETs in RA pathogenesis119.
molecules) and citrullinated proteins, increase in circu- However, despite much evidence of citrullination in
lation. In the second stage, clinical symptoms become human RA, PAD4-deficient mice develop arthritis in the
evident100–103. serum transfer model, which suggests that citrullination
Neutrophils are implicated in RA progression and are of NET components is not required in this model120. This
the most abundant cells in the synovial fluid, where they finding again suggests that disease progression and NET
are longer lived than normal104,105. Ex vivo, neutrophils induction vary greatly between mice and humans.
from patients with RA are pre-​activated by immune Therefore, although increasing evidence suggests that
complexes such as RF, resulting in excessive ROS release, the death of neutrophils via NETosis occurs in patients
degranulation and NETosis106,107. Thus, the activation of with RA and that this activity is detrimental for RA pro-
neutrophils in the synovium is linked to the character- gression, further work is needed to clarify the source of the
istic inflammation and damage in the joint. Neutrophil citrullinated proteins and subsequent autoantigens,
granular proteins such as myeloperoxidase, neutrophil the mechanism by which they are modified and whether
elastase and cathepsin G are abundant in the synovial they are actually important in RA pathogenesis (Fig. 2c).
fluid, probably because of excessive NETosis, and these
proteins cause damage to the joint108. Felty syndrome . Felty syndrome is a complication of
As anti-​citrullinated protein antibodies (ACPAs) are RA that affects 1–3% of patients. This syndrome typ-
a diagnostic hallmark of RA, the source of the modi- ically presents with splenomegaly, neutropenia and
fied self-​proteins that serve as autoantigens is of great joint destruction121. A low absolute neutrophil count is
interest. ACPAs against vimentin, collagen II and his- central to a diagnosis of Felty syndrome. Development
tones, among others, have been identified in serum from of Felty syndrome usually occurs 10–15 years after
patients with RA107–109. The source of these ACPAs in the onset of RA, although simultaneous diagnosis can
patients with RA has been investigated110. Indeed, citrul- occur and has a calamitous outcome because patients
linated vimentin is found on NETs isolated from healthy become far more susceptible to severe infections and
donors and patients with RA108. NETs containing citrul- sepsis than those patients who do not develop the syn-
linated proteins (such as histone H3, histone H4 and drome121. Patients with Felty syndrome have abundant
vimentin) are ingested by fibroblast-like synoviocytes anti-​histone autoantibodies in circulation, the source
(FLS) in a receptor for advanced glycation end-products of which could be histones released during NETosis. In
(RAGE)–TLR9-dependent manner. This process leads Felty syndrome, neutropenia occurs because neutrophils
to the upregulation of major histocompatibility complex are sequestered in the spleen or die. The bone marrow
(MHC) class II expression by the FLS and the subse- might also fail to produce these cells122. Thus, evidence
quent presentation of NET peptides to CD4+ T cells. suggests that Felty syndrome is also a disease in which
The outcome of this inflammatory response to NETs NETs are detrimental.
by the FLS is the activation of B cells and the generation
of ACPAs110. Gout
Another neutrophil feature in RA is the presence of Gout is an acute inflammatory arthritic disease induced
LDGs that express low levels of TNF receptor 1 (TNFR1) by the accumulation of needle shaped monosodium
and TNFR2 on their surface and thus are less respon- urate (MSU) crystals. Crystal deposition in joints such
sive to TNF stimulation111. This feature could potentially as the big toe or in the kidneys initiates an inflammatory
explain why some patients with RA do not respond to event that rapidly progresses and then resolves. Gout
TNF interference treatment, as they contain a subpopu- occurs in 1–2% of the adult population in developed
lation of cells that remain TNF-unresponsive. However, countries. Gout has a higher incidence in men than
unlike LDGs in patients with SLE, LDGs in RA do not in women and was described in detail as far back as
produce NETs excessively111. ancient Egypt123–126.
The role of neutrophils in the progression and res-
Mouse models. Mouse models are used to investigate olution of the acute inflammatory flare in gout is not
the development and progression of RA. The F1 gener- well characterized127. A marked influx of neutrophils
ation of K/BxN mice, produced by crossing T cell recep- into the synovium of the affected joint occurs after acti-
tor transgenic KRN mice on a C57Bl/6 background vation of local immune cells including macrophages,
with autoimmune-​prone non-​obese diabetic (NOD) dendritic cells and synoviocytes128,129. These cells then
mice, develop severe arthritis by 3–5 weeks of age112,113. phagocytose the MSU crystals, thus producing ROS
Transfer of serum or purified IgG molecules from these and initiating inflammatory pathways. The activation
mice into mice deficient in B cells or T cells replicates of the inflammasome and release of IL-1β recruits neu-
the RA-​like phenotype113. The phenotype is also repli- trophils and additional immune cells130. These cells
cated by injection of IgG molecules into healthy mice114. produce chemokines, which in turn exacerbate neu-
Neutrophil depletion in this model of serum transfer trophil recruitment131. These neutrophils generate ROS
arthritis abrogates disease onset115,116. Furthermore, in and release of antimicrobial peptides, such as IL-1β and
a serum transfer model using a Gfi1-deficient mouse, IL-8 (ref.132).
in which neutrophil maturation is reduced, arthritis Neutrophils isolated from patients with gout spon-
does not progress117,118. Notably, serum transfer failed taneously release NETs, and serum from patients with
to induce arthritis in Nrf2-deficient mice (Nrf2 encodes gout induces NET formation in neutrophils of healthy

472 | AUGUST 2018 | volume 14 www.nature.com/nrrheum


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

donors 133. In the synovium, MSU crystals induce an anti-​C D20 monoclonal antibody that depletes
NET production, which can be visualized by detect- B cells138,139. Patients with vasculitis also utilize steroids
ing DNA, myeloperoxidase and neutrophil elastase to control inflammation, along with rituximab. Even with
in gouty tophi. Upon reaching a certain threshold, these therapies, the mortality of patients with vasculitis
NETs sequester and form aggregations of NETs or is high, mostly owing to infection, cardiovascular disease
aggNETs134, which might degrade cytokines. A study or cancer140. Patients with RA are treated with steroids
demonstrated that NADPH-​i ndependent NETosis to reduce inflammation but also receive DMARDs such
occurs in response to MSU crystals135. MSU crystals as methotrexate or infliximab, some of which target
interact with lysosomes, and chromatin decondensa- cytokines produced during flares102. Rituximab is also
tion mediated by neutrophil elastase occurs upon cell used to treat RA. Gout is a well-​understood and well-​
death. The resulting NETs could potentially sequester managed rheumatic disease. Steroid treatment and the
DNA-​coated MSU crystals in tophi135. Thus, neutro- development of modern drugs along with patient edu-
phils might also have a role in the rapid resolution of cation and lifestyle alterations have had a major effect on
inflammation in gout. patient well-​being and mortality141.
This resolution is not due to the removal of MSU Rituximab treatment in patients with autoimmune
crystals, as they remain in the affected joint after the diseases results in reduced NET production and autoan-
inflammation dissipates. Generation of aggNETs is tigen generation; hence, this treatment also targets neu-
thought to provide a backbone for the inflammation-​ trophils4. Therapies that target cytokines also reduce
silent deposits of MSU crystals that are visualized via NETosis, and PAD inhibitors have been used with some
dual-​energy CT. This disease state is referred to as success in mouse models of RA142 and SLE78,79. Regulating
chronic tophaceous gout136. However, this area of gout neutrophils, particularly NETosis and the effects of NETs,
research remains to be clarified. are approaches that need to be developed and hold great
NET aggregation has been shown to require ROS, promise (reviewed elsewhere4).
as injecting MSU crystals into the paw of NADPH-​ Much work is now focusing on targeting NETs in the
oxidase-deficient mice results in accumulation of pro-​ hope of finding a NET inhibitor to aid in halting the pro-
inflammatory cytokines and paw swelling that persists gression of these diseases. This endeavour is challenging, as
longer than the inflammatory response to MSU crystals neutrophils are fundamental to the host immune response,
in wild-​type control mice134. Furthermore, proteases in and as such, the NETs must be targeted in only specific
aggNETs degrade IL-6, TNF and macrophage inflamma- situations while leaving the patient immunologically intact.
tory protein 1α (MIP1α; also known as CCL3)134. Hence,
the accumulation of MSU crystals and NETs in the Conclusions
affected area is central to the resolution of inflammation A growing body of evidence indicates that neutrophils,
by trapping the MSU and degrading pro-​inflammatory and specifically the process of NETosis, are involved in
cytokines. Notably, a study from 2016 challenged the progression of rheumatic diseases. Many studies
the role of neutrophils in gout resolution owing to diffi­ have demonstrated the presence of NET material in
culty in reproducing the data showing that aggNETs the affected tissues of patients, and autoantibodies are
degrade cytokines137. commonly directed against neutrophil proteins that
Although we have some understanding of the role are known to be released during the process of NETosis.
of neutrophils in gout in humans, the role of neutro- Evidence also suggests that neutrophils are required for
phils in mouse models of gout remains uncertain. resolution of inflammation. Questions remain, however,
This uncertainty is yet another example of the differ- as to the mechanism of NET release in these diseases; as
ences in how human and mouse neutrophils respond such, the role of infection prior to disease onset is of
to attack and generate NETs. Neutrophils seem to be great interest for the development of therapeutics.
detrimental in the inflammatory phase of a gout attack, Another factor in the study of rheumatic diseases in
but their ability to aid in resolution requires further terms of the role of neutrophils is the inconsisten-
investigation (Fig. 2d). cies between patient data and mouse models. These
inconsistencies highlight the stark differences between
Therapeutic approaches neutrophils from humans and mice, which are rarely
Treatment of rheumatic diseases is mainly aimed at discussed. Overall, further studies into the role of NETs
reducing the inflammation of the affected area. A com- in these diseases will aid in the development of new
mon treatment is steroid-​induced immunosuppression therapeutic approaches.
via cyclophosphamide or glucocorticoids. In SLE, this
treatment is boosted with chemotherapy and rituximab, Published online 21 June 2018

1. Goldblatt, F. & O’Neill, S. G. Clinical aspects of 4. Gupta, S. & Kaplan, M. J. The role of neutrophils and to disease. Annu. Rev. Immunol. 30, 459–489
autoimmune rheumatic diseases. Lancet 382, NETosis in autoimmune and renal diseases. Nat. Rev. (2012).
797–808 (2006). Nephrol. 12, 402–413 (2016). 8. Tak, T., Tesselaar, K., Pillay, J., Borghans, J. A. M.
2. Diamond, B., Bluestone, J. & Wofsy, D. The immune 5. Kolaczkowska, E. & Kubes, P. Neutrophil recruitment & Koenderman, L. What’s your age again?
tolerance network and rheumatic disease: immune and function in health and inflammation. Nat. Rev. Determination of human neutrophil half-​lives
tolerance comes to the clinic. Arthritis Rheum. 44, Immunol. 13, 159–175 (2013). revisited. J. Leukoc. Biol. 94, 595–601
1730–1735 (2001). 6. Borregaard, N. Neutrophils, from marrow to microbes. (2013).
3. Brinkmann, V. et al. Neutrophil extracellular Immunity 33, 657–670 (2010). 9. Tecchio, C., Micheletti, A. & Cassatella, M. A.
traps kill bacteria. Science 303, 1532–1535 7. Amulic, B., Cazalet, C., Hayes, G. L., Metzler, K. D. Neutrophil-​derived cytokines: facts beyond
(2004). & Zychlinsky, A. Neutrophil function: from mechanisms expression. Front. Immunol. 5, 508 (2014).

NATure RevIewS | RheumaToLogy volume 14 | AUGUST 2018 | 473


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

10. Mantovani, A., Cassatella, M. A., Costantini, C. 35. Shao, W.-H. H. & Cohen, P. L. Disturbances of 59. Hooks, J. J. et al. Immune interferon in the circulation
& Jaillon, S. Neutrophils in the activation and apoptotic cell clearance in systemic lupus of patients with autoimmune disease. N. Engl. J. Med.
regulation of innate and adaptive immunity. Nat. Rev. erythematosus. Arthritis Res. Ther. 13, 202 (2011). 301, 5–8 (1979).
Immunol. 11, 519–531 (2011). 36. Midgley, A. & Beresford, M. W. Cellular localization of 60. Preble, O. T., Black, R. J., Friedman, R. M., Klippel, J. H.
11. Kenny, E. F. et al. Diverse stimuli engage different nuclear antigen during neutrophil apoptosis: & Vilcek, J. Systemic lupus erythematosus: presence in
neutrophil extracellular trap pathways. eLife 6, mechanism for autoantigen exposure? Lupus 20, human serum of an unusual acid-​labile leukocyte
e24437 (2017). 641–646 (2011). interferon. Science 216, 429–431 (1982).
12. Papayannopoulos, V., Metzler, K. D., Hakkim, A. 37. Leffler, J. et al. Neutrophil extracellular traps that 61. Banchereau, J. & Pascual, V. Type I interferon in
& Zychlinsky, A. Neutrophil elastase and are not degraded in systemic lupus erythematosus systemic lupus erythematosus and other autoimmune
myeloperoxidase regulate the formation of neutrophil activate complement exacerbating the disease. diseases. Immunity 25, 383–392 (2006).
extracellular traps. J. Cell Biol. 191, 677–691 (2010). J. Immunol. 188, 3522–3531 (2012). 62. Crow, M. K. Type I interferon in the pathogenesis of
13. Metzler, K. D., Goosmann, C., Lubojemska, A., 38. Fang, Y., Xu, C., Fu, Y. X., Holers, V. M. & Molina, H. lupus. J. Immunol. 192, 5459–5468 (2014).
Zychlinsky, A. & Papayannopoulos, V. A Expression of complement receptors 1 and 2 on 63. Lande, R. et al. Plasmacytoid dendritic cells sense
myeloperoxidase-​containing complex regulates follicular dendritic cells is necessary for the generation self-​DNA coupled with antimicrobial peptide. Nature
neutrophil elastase release and actin dynamics during of a strong antigen-​specific IgG response. J. Immunol. 449, 564–569 (2007).
NETosis. Cell Rep. 8, 883–896 (2014). 160, 5273–5279 (1998). 64. Caielli, S. et al. Oxidized mitochondrial nucleoids
14. Amulic, B. et al. Cell-​cycle proteins control production 39. Maletto, B. A. et al. Presence of neutrophil-​bearing released by neutrophils drive type I interferon
of neutrophil extracellular traps. Dev. Cell 43, antigen in lymphoid organs of immune mice. Blood production in human lupus. J. Exp. Med. 213,
449–462 (2017). 108, 3094–3102 (2006). 697–713 (2016).
15. Martinod, K. et al. Neutrophil elastase-​deficient mice 40. Duffy, D. et al. Neutrophils transport antigen from the 65. Lartigue, A. et al. Role of TLR9 in anti-​nucleosome and
form neutrophil extracellular traps in an experimental dermis to the bone marrow, initiating a source of anti-​DNA antibody production in lpr mutation-​induced
model of deep vein thrombosis. J. Thromb. Haemost. memory CD8+ T Cells. Immunity 37, 917–929 (2012). murine lupus. J. Immunol. 177, 1349–1354 (2006).
14, 551–558 (2016). 41. Abi Abdallah, D. S., Egan, C. E., Butcher, B. A. 66. Nickerson, K. M., Cullen, J. L., Kashgarian, M. &
16. Bruhns, P. Properties of mouse and human IgG & Denkers, E. Y. Mouse neutrophils are professional Shlomchik, M. J. Exacerbated autoimmunity in the
receptors and their contribution to disease models. antigen-​presenting cells programmed to instruct Th1 absence of TLR9 in MRL. Fas(lpr) mice depends on
Blood 119, 5640–5649 (2012). and Th17 T cell differentiation. Int. Immunol. 23, Ifnar1. J. Immunol. 190, 3889–3894 (2013).
17. Kaplan, M. J. & Radic, M. Neutrophil extracellular 317–326 (2011). 67. Kahlenberg, J. M., Carmona-​Rivera, C., Smith, C. K.
traps: double-​edged swords of innate immunity. 42. Friou, G. J. Clinical application of a test for lupus & Kaplan, M. J. Neutrophil extracellular trap-​associated
J. Immunol. 189, 2689–2695 (2012). globulin-​nucleohistone interaction using fluorescent protein activation of the NLRP3 inflammasome is
18. Bryan, N. et al. Reactive oxygen species (ROS) — a antibody. Yale J. Biol. Med. 31, 40–47 (1958). enhanced in lupus macrophages. J. Immunol. 190,
family of fate deciding molecules pivotal in 43. Fritzler, M. J. & Tan, E. M. Antibodies to histones in 1217–1226 (2013).
constructive inflammation and wound healing. drug-​induced and idiopathic lupus erythematosus. 68. Denny, M. F. et al. A distinct subset of
Eur. Cell. Mater. 24, 249–265 (2012). J. Clin. Invest. 62, 560–567 (1978). proinflammatory neutrophils isolated from patients
19. Hirsch, J. G. Bactericidal action of histone. 44. Nässberger, L., Jonsson, H., Sjöholm, A. G., Sturfelt, G. with systemic lupus erythematosus induces vascular
J. Exp. Med. 108, 925–944 (1958). & Heubner, A. Circulating anti-​elastase in systemic damage and synthesizes type I IFNs. J. Immunol. 184,
20. Lee, J. J. & Pope, J. E. A meta-​analysis of the risk of lupus erythematosus. Lancet 1, 509 (1989). 3284–3297 (2010).
venous thromboembolism in inflammatory rheumatic 45. Spronk, P. E. et al. Antineutrophil cytoplasmic 69. Hacbarth, E. & Kajdacsy-​Balla, A. Low density
diseases. Arthritis Res. Ther. 16, 435 (2014). antibodies in systemic lupus erythematosus. neutrophils in patients with systemic lupus
21. Rahman, A. & Isenberg, D. A. Systemic lupus Br. J. Rheumatol. 35, 625–631 (1996). erythematosus, rheumatoid arthritis, and acute
erythematosus. N. Engl. J. Med. 358, 929–939 46. Cervera, R. et al. Anti-​chromatin antibodies in rheumatic fever. Arthritis Rheum. 29, 1334–1342
(2008). systemic lupus erythematosus: a useful marker for (1986).
22. Wilhelm, A. J. & Major, A. S. Accelerated lupus nephropathy. Ann. Rheum. Dis. 62, 431–434 70. Bennett, L. et al. Interferon and granulopoiesis
atherosclerosis in SLE: mechanisms and prevention (2003). signatures in systemic lupus erythematosus blood.
approaches. Int. J. Clin. Rheumtol. 7, 527–539 47. Carmona-​Rivera, C., Zhao, W., Yalavarthi, S. J. Exp. Med. 197, 711–723 (2003).
(2012). & Kaplan, M. J. Neutrophil extracellular traps 71. Midgley, A. & Beresford, M. W. Increased expression
23. Stojan, G. & Petri, M. Atherosclerosis in systemic induce endothelial dysfunction in systemic lupus of low density granulocytes in juvenile-​onset systemic
lupus erythematosus. J. Cardiovasc. Pharmacol. 62, erythematosus through the activation of matrix lupus erythematosus patients correlates with disease
255–262 (2013). metalloproteinase-2. Ann. Rheum. Dis. 74, activity. Lupus 25, 407–411 (2016).
24. Anders, H.-J. J. & Vielhauer, V. Renal co-​morbidity in 1417–1424 (2015). 72. Macanovic, M. et al. The treatment of systemic
patients with rheumatic diseases. Arthritis Res. Ther. 48. Lood, C. et al. Neutrophil extracellular traps enriched lupus erythematosus (SLE) in NZB/W F1 hybrid mice;
13, 222 (2011). in oxidized mitochondrial DNA are interferogenic and studies with recombinant murine DNase and with
25. Hargraves, M. M., Richmond, H. & Morton, R. contribute to lupus-​like disease. Nat. Med. 22, dexamethasone. Clin. Exp. Immunol. 106, 243–252
Presentation of two bone marrow elements; the tart 146–153 (2016). (1996).
cell and the L. E. cell. Proc. Staff Meet. Mayo Clin. 23, 49. Hirose, O., Itabashi, M., Takei, T., Honda, K. & Nitta, K. 73. Davis, J. C. et al. Recombinant human Dnase I
25–28 (1948). Antineutrophil cytoplasmic antibody-​associated (rhDNase) in patients with lupus nephritis. Lupus 8,
26. Carli, L., Tani, C., Vagnani, S., Signorini, V. & Mosca, M. glomerulonephritis with immunoglobulin deposition. 68–76 (1999).
Leukopenia, lymphopenia, and neutropenia in systemic Clin. Exp. Nephrol. 21, 643–650 (2017). 74. Winkelstein, J. A. et al. Chronic granulomatous
lupus erythematosus: prevalence and clinical impact — 50. Skiljevic, D. et al. Serum DNase I activity in disease. Report on a national registry of 368 patients.
a systematic literature review. Semin. Arthritis Rheum. systemic lupus erythematosus: correlation with Medicine 79, 155–169 (2000).
45, 190–194 (2015). immunoserological markers, the disease activity and 75. Campbell, A. M., Kashgarian, M. & Shlomchik, M. J.
27. Hakkim, A. et al. Impairment of neutrophil organ involvement. Clin. Chem. Lab. Med. 51, NADPH oxidase inhibits the pathogenesis of systemic
extracellular trap degradation is associated with lupus 1083–1091 (2013). lupus erythematosus. Sci. Transl. Med. 4, 157ra141
nephritis. Proc. Natl Acad. Sci. USA 107, 9813–9818 51. Bodaño, A., Amarelo, J., González, A., Gómez-​Reino, J. J. (2012).
(2010). & Conde, C. Novel DNASE I mutations related to systemic 76. Jacob, C. O. et al. Haploinsufficiency of NADPH
28. Villanueva, E. et al. Netting neutrophils induce lupus erythematosus. Arthritis Rheum. 50, 4070–4071 oxidase subunit neutrophil cytosolic factor 2 is
endothelial damage, infiltrate tissues, and expose (2004). sufficient to accelerate full-​blown lupus in NZM 2328
immunostimulatory molecules in systemic lupus 52. Al-​Mayouf, S. M. et al. Loss-​of-function variant in mice. Arthritis Rheumatol. 69, 1647–1660 (2017).
erythematosus. J. Immunol. 187, 538–552 (2011). DNASE1L3 causes a familial form of systemic lupus 77. Kienhöfer, D. et al. Experimental lupus is aggravated
29. Wang, H., Li, T., Chen, S., Gu, Y. & Ye, S. Neutrophil erythematosus. Nat. Genet. 43, 1186–1188 (2011). in mouse strains with impaired induction of neutrophil
extracellular trap mitochondrial DNA and its 53. Napirei, M. et al. Features of systemic lupus extracellular traps. JCI insight 2, 92920 (2017).
autoantibody in systemic lupus erythematosus and a erythematosus in Dnase1-deficient mice. Nat. Genet. 78. Knight, J. S. et al. Peptidylarginine deiminase
proof-​of-concept trial of metformin. Arthritis 25, 177–181 (2000). inhibition is immunomodulatory and vasculoprotective
Rheumatol. 67, 3190–3200 (2015). 54. Sisirak, V. et al. Digestion of chromatin in apoptotic in murine lupus. J. Clin. Invest. 123, 2981–2993
30. Lande, R. et al. Neutrophils activate plasmacytoid cell microparticles prevents autoimmunity. Cell 166, (2013).
dendritic cells by releasing self-​DNA-peptide 88–101 (2016). 79. Knight, J. S. et al. Peptidylarginine deiminase
complexes in systemic lupus erythematosus. 55. Gehrke, N. et al. Oxidative damage of DNA confers inhibition disrupts NET formation and protects
Sci. Transl. Med. 3, 73ra19 (2011). resistance to cytosolic nuclease TREX1 degradation against kidney, skin and vascular disease in
31. Garcia-​Romo, G. S. et al. Netting neutrophils are and potentiates STING-​dependent immune sensing. lupus-​prone MRL/lpr mice. Ann. Rheum. Dis. 74,
major inducers of type I IFN production in pediatric Immunity 39, 482–495 (2013). 2199–2206 (2015).
systemic lupus erythematosus. Sci. Transl. Med. 3, 56. Frost, P. G. & Lachmann, P. J. The relationship of 80. Gordon, R. A. et al. Lupus and proliferative nephritis
73ra20 (2011). desoxyribonuclease inhibitor levels in human sera to are PAD4 independent in murine models. JCI insight
32. Arandjelovic, S. & Ravichandran, K. S. Phagocytosis the occurrence of antinuclear antibodies. Clin. Exp. 2, e92926 (2017).
of apoptotic cells in homeostasis. Nat. Immunol. 16, Immunol. 3, 447–455 (1968). 81. Elefante, E. et al. One year in review 2017: systemic
907–917 (2015). 57. Yeh, T.-M. M., Chang, H.-C. C., Liang, C.-C. C., Wu, J.-J. J. vasculitis. Clin. Exp. Rheumatol. 35 (Suppl. 1), 5–26
33. Farrera, C. & Fadeel, B. Macrophage clearance & Liu, M.-F. F. Deoxyribonuclease-​inhibitory antibodies (2017).
of neutrophil extracellular traps is a silent process. in systemic lupus erythematosus. J. Biomed. Sci. 10, 82. Kallenberg, C. G. M., Heeringa, P. & Stegeman, C. A.
J. Immunol. 191, 2647–2656 (2013). 544–551 (2003). Mechanisms of disease: pathogenesis and treatment
34. Ren, Y. et al. Increased apoptotic neutrophils and 58. Trofimenko, A. S., Gontar, I. P., Zborovsky, A. B. of ANCA-​associated vasculitides. Nat. Rev. Rheumatol.
macrophages and impaired macrophage phagocytic & Paramonova, O. V. Anti-​DNase I antibodies in 2, 661–670 (2006).
clearance of apoptotic neutrophils in systemic lupus systemic lupus erythematosus: diagnostic value and 83. Muñoz-​Grajales, C. & Pineda, J. C. Pathophysiological
erythematosus. Arthritis Rheum. 48, 2888–2897 share in the enzyme inhibition. Rheumatol. Int. 36, relationship between infections and systemic
(2003). 521–529 (2016). vasculitis. Autoimmune Dis. 2015, 1–8 (2015).

474 | AUGUST 2018 | volume 14 www.nature.com/nrrheum


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

84. Jennette, J. C. & Falk, R. J. Small-​vessel vasculitis. 106. Eggleton, P., Wang, L., Penhallow, J., Crawford, N. & 130. Martinon, F., Pétrilli, V., Mayor, A., Tardivel, A. &
N. Engl. J. Med. 337, 1512–1523 (1997). Brown, K. A. Differences in oxidative response of Nature, T.-J. Gout-​associated uric acid crystals activate
85. Söderberg, D. & Segelmark, M. Neutrophil subpopulations of neutrophils from healthy subjects and the NALP3 inflammasome. Nature 440, 237–241
extracellular traps in ANCA-​Associated vasculitis. patients with rheumatoid arthritis. Ann. Rheum. Dis. 54, (2006).
Front. Immunol. 7, 256 (2016). 916–923 (1995). 131. Ryckman, C. et al. Role of S100A8 and S100A9 in
86. Falk, R. J. & Jennette, J. C. Anti-​neutrophil 107. Corsiero, E., Pratesi, F., Prediletto, E., Bombardieri, M. neutrophil recruitment in response to monosodium
cytoplasmic autoantibodies with specificity for & Migliorini, P. NETosis as source of autoantigens urate monohydrate crystals in the air-​pouch model
myeloperoxidase in patients with systemic vasculitis in rheumatoid arthritis. Front. Immunol. 7, 485 of acute gouty arthritis. Arthritis Rheum. 48,
and idiopathic necrotizing and crescentic (2016). 2310–2320 (2003).
glomerulonephritis. N. Engl. J. Med. 318, 108. Khandpur, R. et al. NETs are a source of citrullinated 132. Popa-​Nita, O. & Naccache, P. H. Crystal-​induced
1651–1657 (1988). autoantigens and stimulate inflammatory responses in neutrophil activation. Immunol. Cell Biol. 88, 32–40
87. Niles, J. L., McCluskey, R. T., Ahmad, M. F. & rheumatoid arthritis. Sci. Transl. Med. 5, 178ra40 (2010).
Arnaout, M. A. Wegener’s granulomatosis autoantigen (2013). 133. Mitroulis, I. et al. Neutrophil extracellular trap
is a novel neutrophil serine proteinase. Blood 74, 109. Wegner, N. et al. Autoimmunity to specific citrullinated formation is associated with IL-1β and autophagy-​
1888–1893 (1989). proteins gives the first clues to the etiology of related signaling in gout. PLoS ONE 6, e29318
88. Charles, L. A., Caldas, M. L., Falk, R. J., Terrell, R. S. rheumatoid arthritis. Immunol. Rev. 233, 34–54 (2011).
& Jennette, J. C. Antibodies against granule proteins (2010). 134. Schauer, C. et al. Aggregated neutrophil
activate neutrophils in vitro. J. Leukoc. Biol. 50, 110. Carmona-​Rivera, C. et al. Synovial fibroblast-​ extracellular traps limit inflammation by degrading
539–546 (1991). neutrophil interactions promote pathogenic adaptive cytokines and chemokines. Nat. Med. 20, 511–517
89. Brogan, P. & Eleftheriou, D. Vasculitis update: immunity in rheumatoid arthritis. Sci. Immunol. 2, (2014).
pathogenesis and biomarkers. Pediatr. Nephrol. 33, eaag3358 (2017). 135. Chatfield, S. M. et al. Monosodium urate crystals
187–198 (2017). 111. Wright, H. L., Makki, F. A., Moots, R. J. & Edwards, S. W. generate nuclease-​resistant neutrophil extracellular
90. Falk, R. J., Terrell, R. S., Charles, L. A. & Jennette, J. C. Low-​density granulocytes: functionally distinct, traps via a distinct molecular pathway. J. Immunol.
Anti-​neutrophil cytoplasmic autoantibodies induce immature neutrophils in rheumatoid arthritis with 200, 1802–1816 (2018).
neutrophils to degranulate and produce oxygen altered properties and defective TNF signalling. 136. Chhana, A. & Dalbeth, N. The gouty tophus: a review.
radicals in vitro. Proc. Natl Acad. Sci. USA 87, J. Leukoc. Biol. 101, 599–611 (2017). Curr. Rheumatol. Rep. 17, 19 (2015).
4115–4119 (1990). 112. Kouskoff, V. et al. Organ-​specific disease provoked by 137. Reber, L. L., Gaudenzio, N., Starkl, P. & Galli, S. J.
91. Kessenbrock, K. et al. Netting neutrophils in systemic autoimmunity. Cell 87, 811–822 (1996). Neutrophils are not required for resolution of acute
autoimmune small-​vessel vasculitis. Nat. Med. 15, 113. Korganow, A. S. et al. From systemic T cell self-​ gouty arthritis in mice. Nat. Med. 22, 1382–1384
623–625 (2009). reactivity to organ-​specific autoimmune disease via (2016).
92. Söderberg, D. et al. Increased levels of neutrophil immunoglobulins. Immunity 10, 451–461 (1999). 138. Clain, J. M., Cartin-​Ceba, R., Fervenza, F. C.
extracellular trap remnants in the circulation of 114. Ji, H. et al. Genetic influences on the end-​stage & Specks, U. Experience with rituximab in the
patients with small vessel vasculitis, but an inverse effector phase of arthritis. J. Exp. Med. 194, treatment of antineutrophil cytoplasmic antibody
correlation to anti-​neutrophil cytoplasmic antibodies 321–330 (2001). associated vasculitis. Ther. Adv. Musculoskelet. Dis. 6,
during remission. Rheumatology 54, 2085–2094 115. Wipke, B. T. & Allen, P. M. Essential role of neutrophils 58–74 (2013).
(2015). in the initiation and progression of a murine model of 139. Mok, C. C. Current role of rituximab in systemic lupus
93. Cheadle, C. et al. Transcription of proteinase 3 and rheumatoid arthritis. J. Immunol. 167, 1601–1608 erythematosus. Int. J. Rheum. Dis. 18, 154–163
related myelopoiesis genes in peripheral blood (2001). (2015).
mononuclear cells of patients with active Wegener’s 116. Christensen, A. D., Haase, C., Cook, A. D. & Hamilton, 140. Flossmann, O. et al. Long-​term patient survival in
granulomatosis. Arthritis Rheum. 62, 1744–1754 J. A. K/BxN serum-​transfer arthritis as a model for ANCA-​associated vasculitis. Ann. Rheum. Dis. 70,
(2010). human inflammatory arthritis. Front. Immunol. 7, 213 488–494 (2011).
94. Lyons, P. A. et al. Novel expression signatures (2016). 141. Ragab, G., Elshahaly, M. & Bardin, T. Gout: an old
identified by transcriptional analysis of separated 117. Karsunky, H. et al. Inflammatory reactions and severe disease in new perspective – a review. J. Adv. Res. 8,
leucocyte subsets in systemic lupus erythematosus neutropenia in mice lacking the transcriptional 495–511 (2017).
and vasculitis. Ann. Rheum. Dis. 69, 1208–1213 repressor Gfi1. Nat. Genet. 30, 295–300 (2002). 142. Willis, V. C. et al. N-​Α-benzoyl-​N5-(2-chloro-1-
(2010). 118. Monach, P. A. et al. Neutrophils in a mouse model of iminoethyl)-L-​ornithine amide, a protein arginine
95. Grayson, P. C. et al. Neutrophil-​related gene autoantibody-​mediated arthritis: critical producers deiminase inhibitor, reduces the severity of murine
expression and low-​density granulocytes associated of Fc receptor gamma, the receptor for C5a, and collagen-​induced arthritis. J. Immunol. 186,
with disease activity and response to treatment in lymphocyte function-​associated antigen 1. Arthritis 4396–4404 (2011).
antineutrophil cytoplasmic antibody-​associated Rheum. 62, 753–764 (2010). 143. McDonald, B., Urrutia, R., Yipp, B. G., Jenne, C. N. &
vasculitis. Arthritis Rheumatol. 67, 1922–1932 119. Maicas, N. et al. Deficiency of Nrf2 accelerates the Kubes, P. Intravascular neutrophil extracellular traps
(2015). effector phase of arthritis and aggravates joint capture bacteria from the bloodstream during sepsis.
96. Sangaletti, S. et al. Neutrophil extracellular traps disease. Antioxid. Redox Signal. 15, 889–901 (2011). Cell Host Microbe 12, 324–333 (2012).
mediate transfer of cytoplasmic neutrophil antigens 120. Rohrbach, A. S., Hemmers, S., Arandjelovic, S., Corr, M. 144. Kolaczkowska, E. et al. Molecular mechanisms of NET
to myeloid dendritic cells toward ANCA induction and & Mowen, K. A. PAD4 is not essential for disease in the formation and degradation revealed by intravital
associated autoimmunity. Blood 120, 3007–3018 K/BxN murine autoantibody-​mediated model of imaging in the liver vasculature. Nat. Commun. 6,
(2012). arthritis. Arthritis Res. Ther. 14, R104 (2012). 6673 (2015).
97. Kumar, S. V. et al. Neutrophil extracellular trap-​related 121. Owlia, M. B., Newman, K. & Akhtari, M. Felty’s 145. Brinkmann, V., Abu Abed, U., Goosmann, C. &
extracellular histones cause vascular necrosis in severe syndrome, insights and updates. Open Rheumatol. J. Zychlinsky, A. Immunodetection of NETs in
GN. J. Am. Soc. Nephrol. 26, 2399–2413 (2015). 8, 129–136 (2014). paraffin-​embedded tissue. Front. Immunol. 7, 513
98. Kusunoki, Y. et al. Peptidylarginine deiminase inhibitor 122. Dwivedi, N. & Radic, M. Citrullination of autoantigens (2016).
suppresses neutrophil extracellular trap formation and implicates NETosis in the induction of autoimmunity. 146. Papayannopoulos, V., Staab, D. & Zychlinsky, A.
MPO-​ANCA production. Front. Immunol. 7, 227 Ann. Rheum. Dis. 73, 483–491 (2014). Neutrophil elastase enhances sputum solubilization
(2016). 123. Nuki, G. & Simkin, P. A. A concise history of gout and in cystic fibrosis patients receiving DNase therapy.
99. Jiménez-​Alcázar, M. et al. Host DNases prevent hyperuricemia and their treatment. Arthritis Res. Ther. PLoS ONE 6, e28526 (2011).
vascular occlusion by neutrophil extracellular traps. 8, S1 (2006). 147. Caudrillier, A. et al. Platelets induce neutrophil
Science 358, 1202–1206 (2017). 124. Malawista, S. E., de Boisfleury, A. C. & Naccache, P. H. extracellular traps in transfusion-​related acute
100. Deane, K. D. Preclinical rheumatoid arthritis Inflammatory gout: observations over a half-​century. lung injury. J. Clin. Invest. 122, 2661–2671
(autoantibodies): an updated review. Curr. Rheumatol. FASEB J. 25, 4073–4078 (2011). (2012).
Rep. 16, 419 (2014). 125. Kuo, C.-F., Grainge, M. J., Mallen, C., Zhang, W. &
101. Chaudhari, K., Rizvi, S. & Syed, B. A. Rheumatoid Doherty, M. Comorbidities in patients with gout Author contributions
arthritis: current and future trends. Nat. Rev. Drug prior to and following diagnosis: case-​control study. All authors contributed to substantial discussion of content,
Discov. 15, 305–306 (2016). Ann. Rheum. Dis. 75, 210–217 (2014). researching data, writing and reviewing and/or editing the
102. Smolen, J. S., Aletaha, D. & McInnes, I. B. Rheumatoid 126. Kuo, C.-F., Grainge, M. J., Zhang, W. & Doherty, M. manuscript before submission.
arthritis. Lancet 388, 2023–2038 (2016). Global epidemiology of gout: prevalence, incidence
103. Angelotti, F. et al. One year in review 2017: and risk factors. Nat. Rev. Rheumatol. 11, 649–662 Competing interests
pathogenesis of rheumatoid arthritis. Clin. Exp. (2015). The authors declare no competing interests.
Rheumatol. 35, 368–378 (2017). 127. Rada, B. Neutrophil extracellular traps and
104. Edwards, S. W. & Hallett, M. B. Seeing the wood microcrystals. J. Immunol. Res. 2017, 1–7 (2017). Publisher’s note
for the trees: the forgotten role of neutrophils in 128. Amaral, F. A. A. et al. NLRP3 inflammasome-​mediated Springer Nature remains neutral with regard to jurisdictional
rheumatoid arthritis. Immunol. Today 18, 320–324 neutrophil recruitment and hypernociception depend claims in published maps and institutional affiliations.
(1997). on leukotriene B(4) in a murine model of gout.
105. Ottonello, L. et al. Delayed neutrophil apoptosis Arthritis Rheum. 64, 474–484 (2012). Reviewer information
induced by synovial fluid in rheumatoid arthritis: role 129. Mitroulis, I., Kambas, K. & Ritis, K. Neutrophils, IL-1β, Nature Reviews Rheumatology thanks M. Herrmann, M. Radic
of cytokines, estrogens, and adenosine. Ann. N. Y and gout: is there a link? Semin. Immunopathol. 35, and the other anonymous reviewer(s) for their contribution to
Acad. Sci. 966, 226–231 (2002). 501–512 (2013). the peer review of this work.

NATure RevIewS | RheumaToLogy volume 14 | AUGUST 2018 | 475


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

You might also like