You are on page 1of 9

REVIEWS

Inflammation in gout: mechanisms


and therapeutic targets
Alexander K. So1 and Fabio Martinon2
Abstract | The acute symptoms of gout are triggered by the inflammatory response to
monosodium urate crystals, mediated principally by macrophages and neutrophils. Innate
immune pathways are of key importance in the pathogenesis of gout, in particular the activation
of the NLRP3 inflammasome, which leads to the release of IL‑1β and other pro-inflammatory
cytokines. The orchestration of this pro-inflammatory cascade involves multiple intracellular and
extracellular receptors and enzymes interacting with environmental influences that modulate
the inflammatory state. Furthermore, the resolution of inflammation in gout is becoming better
understood. This Review highlights recent advances in our understanding of both positive and
negative regulatory pathways, as well as the genetic and environmental factors that modulate
the inflammatory response. Some of these pathways can be manipulated and present novel
therapeutic opportunities for the treatment of acute gout attacks.

Gout is now the most common cause of inflammatory can be present without an inflammatory response. These
Autoinflammatory disease
Inflammatory diseases not due arthritis, and its epidemiology worldwide points to an observations suggest that regulatory mechanisms exists
to infections or injuries, mostly increase in incidence and prevalence in both developed that modify the acute inflammatory response, and a
caused by malfunction in the and developing countries1. Gout is caused by hyper­ thorough understanding of pro-­inflammatory as well
innate immune system. uricaemia (serum urate levels >7 mg/l (420 μmol/l)) lead­ as anti-inflammatory pathways could help to develop
Inflammasome
ing to the formation and deposition of monosodium urate new strategies for the treatment of gout. In this Review,
A multiprotein cytoplasmic (MSU) crystals. Clinically, the disease is characterized by we discuss advances over the past decade in the field of
complex that activates one or acute episodes of joint inflammation, usually affecting a gout inflammation research and emerging therapeutic
more inflammatory caspases, single joint, interspersed with symptom-free periods of strategies to manage acute gout attacks.
such as caspase‑1, leading
variable duration. If untreated, gout typically progresses
to the processing and secretion
of the pro-inflammatory to the formation of urate deposits (tophi) in soft tissues, Uric acid-mediated inflammation
cytokines IL‑1β and IL‑18, and recurrent attacks of arthritis affecting multiple joints and IL‑1β–mediated inflammation is a key aspect of gouty
the processing and activation progressive joint destruction. Other complications include inflammation. In gout, IL‑1β production is mediated
of factors triggering pyroptosis renal deposits of uric acid that can provoke renal failure by MSU crystals triggering the inflammasome, a multi­
such as gasdermin D.
and the formation of renal stones. These and other clinical molecular complex whose dysregulation is central to
features have been reviewed elsewhere2. many pathological inflammatory conditions.
Gout is now regarded as a prototypical inflamma­
tory disease driven by activation of the innate immune Inflammasome activation by MSU crystals
1
Service of Rheumatology, system. Gout has also been termed an autoinflammatory MSU crystals trigger an inflammatory response from
Centre Hospitalier disease3; however, this classification is misleading as, macrophages. The crystals are first taken up by macro­
Universitaire Vaudois and
University of Lausanne,
unlike hereditary autoinflammatory diseases, the acute phages and promote the assembly and activation of the
Avenue Pierre Decker 4, trigger of gout is MSU crystals. Uric acid itself is an endo­ NLRP3 inflammasome4. Inflammasomes are cytosolic
1011 Lausanne, Switzerland. genous and ubiquitous metabolite that is not considered multiprotein complexes that can initiate inflammatory
2
Department of to be pro-inflammatory, and MSU crystal formation is responses5,6.
Biochemistry, University of
required to provoke clinically observed inflammation. Inflammasomes assemble when cytosolic pattern-­
Lausanne, 155 Chemin des
Boveresses, 1066 Epalinges, The study of the underlying mechanisms of gouty recognition receptors (PRRs) such as NLRP3 sense acti­
Switzerland. inflammation has led to remarkable insights into the con­ vating signals that have reached the cytosol of the cell.
Correspondence to A.K.S.  trol of the inflammasome and pro-inflammatory cytokine This signalling leads to the oligomerization of the PRR
alexanderkai-lik.so@chuv.ch release. Nevertheless, we must bear in mind some of the and its recruitment to a complex of adaptor proteins and
doi:10.1038/nrrheum.2017.155 other distinguishing features of gout: firstly, that the attack effector enzymes (FIG. 1). NLRP3 inflammasomes are
Published online 28 Sep 2017 is usually self-limiting and, secondly, that MSU crystals formed by the recruitment of the adaptor protein ASC

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points Mechanisms of inflammasome engagement


Despite the fact that activation of IL‑1β production and
• Inflammatory cytokines, in particular IL‑1β, are the key mediators of gouty the role of the NLRP3 inflammasome in gout is well-­
inflammation described, the upstream pathway that links MSU crystals
• The NLRP3 inflammasome is the major pathway by which MSU crystals trigger to NLRP3 activation is poorly understood. Inflammasome
the cellular inflammatory response engagement can be dissected into two prerequisite steps:
• Multiple regulatory pathways modulate the activity of the inflammasome priming and activation. Reliance on two signals is a key
and the release of IL‑1β; this may explain in part the clinical origins of gouty feature of most inflammasomes and increases the speci­
inflammation ficity of the response while avoiding inappropriate firing
• Diet influences hyperuricaemia as well as the inflammatory state of macrophages of the pathway. In gout, the nature of signal 1 is unclear but
in gout might rely on the activation of Toll-like receptors (TLRs)14.
• Nutrients can modulate inflammasome activity and IL‑1β release and participate Signal 2 is provided by the interaction of MSU crystals
in the regulation of pro-inflammatory as well as anti-inflammatory pathways with inflammasome-competent cells15.
in gout
• The resolution of gouty inflammation is regulated at the cellular level as well as at the Signal 1 of inflammasome engagement primes cells for
level of activation of the inflammasome; these pathways provide promising new
inflammasome assembly. Priming of the cell, also known
avenues for therapeutic intervention
as signal 116, controls the expression of all components
required for the assembly and activation of the inflam­
masome and contributes to the expression of the pre­
and subsequent recruitment of caspase‑1. Following cursor proteins that are the substrates of inflammatory
initial oligomerization within the inflammasome, caspases. This inflammasome-competent stage, which is
ASC monomers can further auto-assemble into high-­ achieved as a result of an inflammatory milieu, can orig­
molecular-weight oligomers. This process, referred to as inate by the engagement of innate immune receptors on
‘prion-like’ polymerization, amplifies the signals sensed the cell surface or as part of an auto-amplification loop
by the PRR and engages virtually all ASC molecules into via IL‑1β itself.
one active cellular complex 7. Recruitment and oligo­ Signalling pathways mediated by cell-surface recep­
merization of caspase‑1 by this structure leads to the tors coordinate the innate immune response. The best
activation and proteolytic processing of its substrates. known of these receptors are the TLR family, of which
Caspase‑1 activates the pro-inflammatory cytokines TLR2 and TLR4 in particular have been implicated in
IL‑1β and IL‑18 by cleaving their respective pre­ gouty inflammation. Work published in 2005 showed
cursor proteins, pro‑IL‑1β and pro‑IL‑18. In gout, that mice deficient in TLR2 or TLR4 have an impaired
inflammasome-­mediated IL‑1β‑release triggers an impor­ neutrophil response to MSU crystals in the air-pouch
tant inflammatory response, with vasodilatation and rapid model of gout14. A direct interaction between MSU crys­
recruitment of neutrophils to the site of crystal deposition, tals and the TLRs was postulated, as macrophages defi­
and thereby drives acute inflammatory episodes8. cient for these receptors did not take up MSU crystals as
Additional caspase‑1 substrates, such as gasdermins, efficiently as wild-type macrophages; however, no clear
are emerging as downstream targets of inflammasome evidence demonstrated that MSU crystals could directly
engagement 9. Gasdermins promote cell death upon activate TLRs. Subsequent data suggested that TLRs
the activation of inflammatory caspases10,11. Caspase‑1 regulate gouty inflammation by recognition of ligands
and caspase‑11 can cleave gasdermin D to release its that prime monocytes and macro­phages to produce
N‑terminal portion that then polymerizes at the plasma pro‑IL‑1β. Proteins S100A8 and S100A9 are endo­
membrane, forming cytotoxic pores. These pores alter genous ligands of TLR4 and are secreted upon activa­
cellular integrity and result in cell death by pyroptosis, tion of phagocytes. Patients with gout and mice injected
which is mediated by inflammatory caspases and differs with MSU crystals produced high levels of S100A8
from cell death mediated by apoptotic caspases in that and S100A9, and genetic deletion of S100A9 reduced
it results in the release of the cytosolic contents of cells, the response to MSU crystals in mice17. Another TLR
including a plethora of pro-inflammatory mediators ligand that might have a role in macrophage priming in
and danger signals. Pyroptosis can therefore amplify gout is long-chain free fatty acids (FFAs). In a murine
the inflammatory response and facilitate the release of model of gout, arthritis was only observed when mice
cytokines, including IL‑1β. Whether this pathway con­ were injected with both long-chain FFAs and MSU
tributes to inflammation in gout remains to be estab­ crystals, whereas injection of MSU alone or long-chain
lished. Uric acid crystals have also been proposed to FFAs alone was not sufficient to elicit inflammation.
trigger necroptosis, another pro-inflammatory type Furthermore, it was demonstrated that TLR2 is the
of cell death that is mediated by the activation of the receptor that mediated the effects of long-chain FFAs
Pyroptosis
Inflammatory form of cell receptor-interacting serine/threonine-protein kinase 3 on macrophages18. The mechanisms by which TLRs can
death mediated by (RIPK3) and mixed lineage kinase domain-like protein regulate inflammation have been reviewed elsewhere19.
inflammatory caspases such as (MLKL) pathways12. Interestingly, disruption of cellular Other factors such as granulocyte-macrophage
caspase‑1 and caspase‑11 that integrity during necroptosis can elicit NLRP3 inflamma­ colony-­stimulating factor (GM‑CSF) and the comple­
results in the extracellular
release of cellular content,
some assembly 13. Thus, necroptosis and pyroptosis ment protein C5a have been proposed to affect cell prim­
including inflammatory might cooperate to amplify the release of inflammatory ing in gout. In monocytes from GM‑CSF-neutralized
mediators and danger signals. mediators in gout. mice, decreased levels of IL‑1β were observed following

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


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Signal 1 Signal 2

TLR
Potassium efflux IL-1β

Plasma
membrane MSU crystals Pyroptotic pore

Pyroptosis
Inflammasome K+
activation

ROS
Mitochondria

N-terminal
Inflammasome Nek7 Gasdermin D cleavage product
components
NF-κB

Caspase-1
activation

Pro-IL-1β IL-1β
Nucleus
Inflammasome

Figure 1 | NLRP3 inflammasome activation by monosodium urate crystals. The NLRP3 inflammasome Nature Reviews | Rheumatology
must be primed
before activation. Priming (signal 1) is mediated by NF‑κB–activating pathways, such as those activated by a member of the
Toll-like receptor (TLR) family. This signalling cascade induces the expression of functional inflammasome components such
as NLRP3. Monosdium urate (MSU) crystals provide signal 2, triggering the assembly of the inflammasome. The interaction
of MSU crystals with the plasma membrane promotes a cellular response that is still poorly understood but includes
hallmarks of NLRP3 activation, including potassium efflux through ion channels and mitochondrial perturbations leading to
the production and release of mitochondrial reactive oxygen species (ROS) into the cytosol. NLRP3‑activating factors such
as the mammalian NIMA-related Ser/Thr (Nek) kinase Nek7 are then engaged, promoting NLRP3 oligomerization and
inflammasome assembly. The adaptor protein ASC is recruited to the inflammasome and nucleates into prion-like filaments.
Caspase‑1 is recruited by ASC and oligomerizes along the ASC filaments, leading to the autoproteolytic activation of
caspase‑1. Active caspase‑1 then promotes the proteolytic cleavage and maturation of pro‑IL‑1β into biologically active
IL‑1β. Caspase‑1 also promotes the cleavage of gasdermin D to generate an N‑terminal cleavage product that oligomerizes
at the plasma membrane, causing the formation of pyroptotic pores. These pores disrupt the integrity of the cellular plasma
membrane, and might contribute to the release of inflammatory mediators including IL‑1β.

ex vivo stimulation with MSU crystals. These mono­ Perturbation of cellular ionic balances, in particular
cytes also exhibited decreased expression of NLRP3 and potassium efflux and calcium influx, is a characteristic
pro‑IL‑1β20. Conversely, treatment with C5a increased feature of NLRP3 inducers22,23. This ionic perturbation
the expression of IL‑1β and IL‑18 and exacerbated MSU is necessary for the generation of mitochondrial reactive
crystal-mediated peritonitis in a mouse model of gout20,21. oxygen species (ROS) upstream of NLRP3 inflamma­
Although priming is necessary for inflammasome some assembly. ROS production is also an essential step
assembly, this step is nonspecific and can result from for inflammasome formation, and is increased by MSU-
various conditions and signals that promote an under­ mediated leukotriene B4 (REF. 24) and might contribute
lining inflammatory response. Priming provides an envi­ to engage Nek7, a member of the family of mammalian
ronment for inflammasome engagement, but on its own NIMA-related Ser/Thr (Nek) kinases. Nek7 directly binds
is not sufficient to trigger the inflammasome pathway. NLRP3 and could be the common NLRP3‑activating
ligand25–27. How Nek7 interacts with NLRP3 and the
Signal 2 catalyses inflammasome assembly. A second mechanisms by which MSU crystals promote the ionic
signal, signal 2, is required for inflammasome activation. changes that ultimately engage the NLRP3‑activating cas­
This signal is more specific than signal 1 and directly cade are important unanswered questions.
drives post-transcriptional and translational aggregation
and polymerization of inflammasome components. The Mediators of the inflammatory response
mechanisms by which MSU crystals trigger signal 2 to IL‑1β is a key cytokine in gout
promote NLRP3 activation are still poorly understood. IL‑1β is a cytokine that acts on multiple cell types to elicit
However, several steps commonly found upstream of inflammatory responses28. Promotion of vaso­dilatation
NLRP3 activation are involved. by IL‑1β leads to the recruitment of monocytes and

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

neutrophils to sites of tissue insults, a response that IL‑1β is mainly produced by innate immune cells and
is crucial in combating infection and restoring tissue signals to target cells by binding to IL‑1 receptor type 1
homeostasis. However, sustained IL‑1β secretion can (IL‑1R1). Once activated, IL‑1R1 and its co‑receptor
result in the production of matrix-degrading enzymes IL‑1 receptor accessory protein (IL‑1RAcP) recruit a
that break down cartilage and bone29. At the systemic signalling complex that shares components with TLR
level, IL‑1β elicits a fever response by acting directly on signalling pathways, leading to the activation of pro-­
the hypothalamic temperature-regulation centre30. inflammatory transcription factors including nuclear
factor‑κB (NF‑κB) as well as p38 c‑Jun N‑terminal
kinase (JNK) (FIG. 2). These transcription factors in turn
Inflamed joint promote the transcriptional upregulation of chemokines
Uric acid and pro-inflammatory mediators that orchestrate the
deposition IL‑1‑mediated inflammatory response.
Although inflammasome activation is the best char­
acterized mechanism leading to IL‑1β maturation in
gout, it should be noted that, in addition to inflammatory
caspases, other proteases can contribute to IL‑1 matura­
tion31. In the absence of the inflammasome, neutrophils
can process pro‑IL‑1β by the activity of neutrophil-­
MSU crystals
derived serine proteases such as proteinase‑3 (PR3,
also known as myeloblastin), neutrophil elastase and
cathepsin G31,32. Other serine proteases can also process
IL‑1β33. Some metallo­proteinases and granzyme A have
also been proposed to trigger the proteolytic activation
of IL‑1β34,35. Whether these pathways function to amplify
Innate immune cells the inflammatory reaction, for example in tissues with
• Macrophages robust neutrophil recruitment, or as back‑up mechanisms
• Monocytes that maintain IL‑1β production in conditions in which
• Neutrophils
inflammasome proteins are absent or inhibited, is unclear.
Although it is now widely accepted that IL‑1β is a
IL-1β
pivotal cytokine in acute gout 15, a role for IL‑1α cannot
IL-1β IL-1R1 IL-1β IL-1RAcP Plasma be ruled out. IL‑1α is released during crystal-induced
membrane
inflammation, and mice with deletion of the Il1b gene
IL-1R-responsive cells are still capable of mounting a neutrophil response36.
• Endothelial cells
• Synoviocytes MyD88 The clinical relevance of IL‑1 is supported by data from
IRAK1/IRAK2/IRAK4 a number of different studies of IL‑1 inhibition37. The clin­
P TRAF6 ical experience with different IL‑1 inhibitors, including
the anti‑IL‑1β monoclocal antibody canakinumab and the
IKKγ synthetic IL‑1 receptor antagonist (IL‑1Ra) anakinra, in
P IKKα IKKβ P the treatment of gout is detailed in a later section.
IκB
P degradation Other contributing cytokines
p65 IκBβ P IL‑8, also known as CXC-chemokine ligand 8, is a
p50
macrophage-­secreted chemokine that acts principally
• Cytokines
• Chemokines on neutrophils. In a study published in 2015, data from
• Cytokines three different cohorts of patients with gout showed that
↑ Inflammatory p65 p50 • Chemokines circulating levels of IL‑8 were increased during the acute
cascades phase of gout and, interestingly, also during the inter­
↑ Neutrophil influx Nucleus critical phase of gout (that is, the interval between flares);
high circulating levels of IL‑8 also predicted concomitant
Figure 2 | IL‑1 signalling links inflammasome activation with inflammatory diabetes mellitus in patients with gout. By contrast, other
cascades. Monosodium urate (MSU) crystals are detected by innate
Nature immune
Reviews cells such
| Rheumatology comorbidities that are commonly seen in gout (such as
as macrophages, monocytes or neutrophils that respond and produce active IL‑1 β. cardiovascular disease and chronic kidney disease) were
IL‑1β signals through the IL‑1 receptor complex, composed of IL‑1 receptor type 1 not significantly associated with high IL‑8 levels38. The
(IL‑1R1) and its cofactor IL‑1 receptor accessory protein (IL‑1RAcP), leading to mechanisms underlying these observations have not yet
recruitment of the adaptor protein MyD88. The expression of IL‑1R1 is widespread, on been elucidated, but suggest that neutrophil recruitment
leukocytes as well as on endothelial and synovial cells. IL‑1R1 expression results in the and neutrophil activation are key inflammatory pathways.
recruitment of effector proteins such as the IL‑1 receptor-associated kinases (IRAKs)
(mostly IRAK4) and TNF receptor-associated factor 6 (TRAF6). This protein recruitment
triggers the IκB kinase (IKK) complex and leads to the phosphorylation and degradation Soluble uric acid
of the NF‑κB inhibitor IκB. Activation of NF‑κB turns on the transcription of cytokines and In gout, it is the formation of MSU crystals that trig­
neutrophil-recruiting chemokines, which will amplify the response and initiate a complex gers acute inflammation, but data show that hyperuri­
inflammatory cascade. caemia can also modulate the inflammatory response.

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


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Hyperuricaemia in the absence of MSU crystals is able Genetics of gouty inflammation


to skew the leukocyte response towards an inflammatory Genetic studies, including genome-wide association
pattern through epigenetic modifications such as histone studies (GWAS), have identified dozens of suscepti­
methylation39. In patients with hyperuricaemia, this effect bility loci associated with hyperuricaemia and gout 48.
was shown by the enhanced production of IL‑1β and These loci mostly influence uric acid levels by affecting
IL‑6, and the concomitant reduction of IL‑1Ra release39. pathways such as renal and gut excretion of uric acid.
Although hyperuricaemia might influence oxidative
Dietary factors and the microbiome stress and thereby have some effect on inflammatory path­
Clinical observations indicate that dietary factors have ways, whether polymorphisms associated with hyper-
a major role in gout, and the link between patterns of uricaemia and gout directly modulate inflammatory
food consumption and hyperuricaemia and gout has responses, beyond promoting uric acid crystallization,
long been established40. Moreover, the role of particular remains to be demonstrated.
foods in triggering an acute attack has been raised, and Two studies demonstrated an association between
findings from an Internet-based survey suggest that a gout and functional variants in the gene encoding
purine-rich diet increases the risk of an acute attack of caspase recruitment domain-containing protein 8
gout fivefold41. However, which components of food are (CARD8)49,50. CARD8 is a potential negative regulator
responsible and how they can lead to an attack remains of the NLRP3 inflammasome; therefore, it is possible
to be determined. As mentioned earlier, long-chain that these polymorphisms might increase inflammas­
(C18) FFAs might have a role in priming macrophages ome activity and thereby contribute to the intensity or
to release IL‑1β upon phagocytosis of MSU crystals18. duration of NLRP3 engagement in gouty episodes. In
The gut microbiome and how this affects inflamma­ addition, there is genetic evidence for a role for TLR4
tion has been increasingly studied in the past few years. in gout. Employing a candidate-gene approach, two
Using a mouse model of gout, Viera et al. showed that studies, one performed in a Han Chinese population
germ-free mice showed attenuated MSU crystal-induced and the other in patients of European ancestry, found
inflammation and that this effect was reproduced by anti­ an association between the same genetic polymorphism
biotic treatment. They also showed that these effects were of the TLR4 gene (rs2149356) and gout 51,52. These poly­
mediated by acetate, a short-chain FFA that is released by morphisms might affect the priming phase of inflam­
gut bacteria. Acetate acts via the macrophage receptor FFA masome engagement or might have a broader effect on
receptor 2 (also known as GPR43) to modulate inflam­ the inflammatory responses in these patients.
masome activation and IL‑1β production. Restoring the Another study linked gout incidence with a poly­
normal gut flora in germ-free mice, or the administration morphism within the PPARGC1B gene, encoding
of acetate, restored the inflammatory effects42. The same peroxi-some proliferator-activated receptor γ (PPARγ) co-
group further showed that a high-fibre diet can attenuate ­activator 1β53, which increased NLRP3 and IL‑1β expres­
arthritis severity in the same murine model of gout43. In sion. Because PPARγ co-activator 1β functions as a
this study, the increased production of acetate and other regulator of PPARγ, a master regulator of metabolism,
short-chain fatty acids resulting from the high-fibre diet this genetic evidence might link metabolic deregulation
was proposed to regulate the resolution of inflammation, with gouty inflammation.
possibly by affecting neutrophils43. These studies suggest
that environmental factors such as microbial metabolites Resolution of inflammation
can differentially regulate the cell types and pathways Monocytes and macrophages are the major cellular
involved in gouty inflammation. Changes in the micro­ sources of IL‑1, but at the site of inflammation neu­
biome in patients with gout have not been extensively trophils predominate. The major pro-inflammatory
studied; however, in one report, the bacterial flora of role of the neutrophil and the mechanisms of interac­
patients with gout was different from that of healthy indi­ tion between MSU crystals and the neutrophil have
viduals and similar to that of patients with type 2 diabetes been reviewed elsewhere54. Interestingly, neutrophils
mellitus44. These findings need to be reproduced in larger probably also have a major role in the resolution of
cohorts before we can draw clear conclusions on how the acute gout, through the formation of neutrophil extra­
microbiome modulates inflammation in gout. cellular traps (NETs). This process is favoured at high
Although interest in using diet to modulate hyper­ concentrations of neutrophils in experimental settings
uricaemia is high, its overall effect is modest 45. However, (>10 × 106 cells per ml), and results in the formation of
dietary factors might influence gouty inflammation. cellular aggregates that contain cellular debris and DNA,
Clinical data from a study published in 2016 suggest as well as neutrophil proteases released into the NETs55.
that higher dietary consumption of omega‑3 fatty acids NET formation is dependent on the generation of ROS,
is associated with a lower frequency of acute gout flares46. and molecules that regulate necroptosis via the RIPK3
Experimentally, omega‑3 fatty acids (eicosapentaenoic pathway have also been implicated56. In the absence of
acid and docosahexaenoic acid) can inhibit NLRP3 RIPK3, NET formation was inhibited completely56. Once
inflammasome activation via a pathway that involves formed, cellular aggregates containing NETs can rapidly
β‑arrestin‑2 and the G‑protein coupled receptors FFA degrade a wide range of pro-inflammatory cytokines
receptor 4 (GPR120) and FFA receptor 1 (GPR40)47. and, in experimental models, the inhibition of NET for­
These findings require confirmation by intervention mation results in severe and persistent gouty inflamma­
trials using omega‑3 fatty acids in patients with gout. tion, whereas joint inflammation spontaneously resolves

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

after 3 days when NET formation is not impaired55. Anti-inflammatory cytokines also contribute to the
These results show that neutrophils have a dual role in resolution of the acute inflammatory process. In animal
gouty inflammation: in the initial phase, when inflam­ models, the addition of exogenous transforming growth
mation is amplified by recruited neutrophils, and in the factor β1 (TGFβ1) reduced experimental inflammation59;
resolution of inflammation (FIG. 3). Currently, no thera­ in patients with gouty arthritis, TGFβ1, IL‑10 and IL‑1Ra
pies are available that can modulate NET formation or were increased in the synovial fluid and were associated
their activity. with the spontaneous resolution of acute gouty arthri­
The metabolic regulatory enzyme AMP-activated tis60. Other factors might also contribute to the timely
kinase (AMPK) contributes to the control of gout by resolution of inflammation in gout. For example, the pro­
its modulatory effects on IL‑1β and growth-regulated tein annexin A1, a potential inhibitor of phospholipase
α protein (also known as CXCL1) secretion by macro­ A2, decreases inflammation and promotes resolution in
phages, and might serve as a sensor that links metabolic mouse models of gout61.
changes to the inflammatory response in gout. AMPK Comprehensive studies interrogating host and environ-
levels are regulated by multiple factors including exer­ mental factors influencing inflammation in humans have
cise and hypoxia, and affect cellular metabolism of shown that seasonal variation in α1‑antitrypsin (AAT)
lipids and glucose57. The addition of a pharmaco­logical concentration affects cytokine production in patients
AMPK activator inhibited MSU crystal-­m ediated with gout 62. AAT, a member of the serpin superfamily
inflammation both in macrophages and in mice 58. that inhibits many serine proteases, was found to inhibit
Moreover, mice deficient for the α‑chain of AMPK had IL‑1β production upon treatment with MSU crystals in
reduced inflammation compared with wild-type mice. mice63. Circulating levels of AAT are at their highest
AMPK levels were also increased by administration in February and their lowest in the summer months. By
of low-dose colchicine in macrophages treated with contrast, retrospective studies have shown that gout peaks
MSU crystals, suggesting that the effects of colchicine in the spring and summer 64, when AAT concentration is
on AMPK might contribute to its ability to inhibit the at the lowest and IL‑1β production is at the highest. These
secretion of IL‑1β58. findings suggest that AAT is a negative regulator of gouty
inflammation62. Understanding how this seasonally reg­
ulated factor influences inflammation is an exciting new
area of research. In particular, it would be interesting
Short-chain FFA to understand how AAT influences the duration and
intensity of gouty episodes.
MSU crystals
FFAR2 Targeting inflammatory pathways
Treating gout requires two complementary approaches:
one aimed at lowering levels of uric acid and the other
• XO aimed at reducing inflammation. NSAIDs, colchicine and
• ROS
• Histone glucocorticoids are commonly used and are effective in
methylation NETosis ROS relieving the pain and inflammation of an acute attack;
however, insights into the biology of inflammation open
the way to new therapeutic strategies (FIG. 4).
Inflammasome
Proteases Modulators of the NLRP3 inflammasome
Macrophage Neutrophil As the NLRP3 inflammasome is a key component in
the response to MSU crystals, strategies that impede
its activation or affect its activity could reduce gouty
TGFβ • IL-1β inflammation. Interestingly, colchicine blocks MSU
• IL-1α Degradation
• IL-8 crystal-mediated NLRP3 activity in macrophages15,
• IL-6 Pro-inflammatory effects probably by inhibiting microtubule-driven rearrange­
• TNF
ment of mitochondria following the engagement of
NLRP3 with MSU crystals65.
Other molecules that target key steps in NLRP3 assem­
bly affect inflammation. β‑Hydroxybutyrate suppresses
Figure 3 | Checks and balances of gouty inflammation.NatureMultiple regulatory
Reviews pathways
| Rheumatology inflammasome activation in response to MSU crystals66.
influence the acute inflammatory response to monosodium urate (MSU) crystals. This ketone body is produced in the liver of mammals
The interaction between macrophages and neutrophils is important in the regulation of during nutrient deprivation. Hence, starvation attenu­
the acute inflammatory response. Modulators of NLRP3 inflammasome activation, ates caspase‑1 activation and IL‑1β secretion in mouse
including acetate, omega‑3 fatty acids and antioxidants, can dampen IL‑1β‑release. High
models of caloric restriction66. Similarly, a ketogenic diet
concentrations of neutrophils will also favour the formation of neutrophil extracellular
trap (NET) aggregates and NETosis, which contain proteases capable of degrading protects rats from MSU crystal-mediated gouty flares67.
inflammatory cytokines. Finally, the release of transforming growth factor β (TGFβ) by Mechanistically, β‑hydroxybutyrate has been proposed
macrophages also acts as a brake on the inflammatory response. FFA, free fatty acid; to inhibit potassium efflux upstream of NLRP3 and to
FFAR2, FFA receptor 2 (also known as G-protein coupled receptor 43); ROS, reactive directly affect inflammasome assembly 66. However, a
oxygen species; XO, xanthine oxidase. possible effect on priming has also been suggested67.

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


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Multiple studies in gout highlight the beneficial effects release of IL‑1β in a human cell line70; further studies are
of compounds that inhibit ROS production and decrease required to interrogate possible application of this drug
oxidative stress. Epigallocatechin gallate, a potent anti­ in gout. Xanthine oxidase inhibitors used in patients to
oxidant polyphenol found in green tea, inhibits neutro­ decrease serum urate levels also directly affect mito­
phil infiltration and IL‑1β secretion in a mouse model chondrial ROS production, thereby inhibiting MSU
of MSU crystal-mediated peritonitis68. Morin, a natural crystal-mediated inflammasome activation71.
flavonol, impairs MSU crystal-induced inflammation in Inhibiting ROS production or potassium efflux are
mouse macrophages69.The gastroprotective drug rebami­ rather nonspecific strategies that could have undesirable
pide suppresses the MSU crystal-mediated activation and effects. Identification of more specific NLRP3 inhibi­
tors could, therefore, present more suitable therapeutic
options for gout. For example, MCC950 (also known as
CP‑456,773 or CRID3) has emerged as a potential drug
Potassium of interest. This drug is a diarylsulfonylurea-­containing
Blockers of potassium efflux efflux
compound that was initially identified as an inhibitor
Liver product: of extracellular ATP-mediated maturation of IL‑1β72.
This discovery preceded the initial description of the
β-hydroxybutyrate inflammasome and the identification of NLRP3 as
the main sensor of extracellular ATP signals. Subsequently
K+ it was demonstrated that MCC950 specifically inhib­
its the NLRP3 inflammasome 73. This drug blocks
Antioxidants targeting mitochondrial ROS NLRP3‑induced ASC oligomerization in mouse and
Green tea molecule:
human macrophages without affecting the activation of
ROS
NLRP1, AIM2 or NLRC4 inflammasomes. The effects
Epigallocatechin gallate of several NLRP3 activators, including MSU crystals, were
ROS inhibited by MCC95073,74, indicating that the drug might
ROS directly act on a conserved NLRP3‑activating mechanism;
however, exactly how this drug affects NLRP3 activation
is not yet clear.
NLRP3 inflammasome inhibitors
Plant molecule: Inhibitors of IL‑1β maturation
Colchicine Considerable effort has been made to develop specific
caspase‑1 inhibitors. VX‑765, an orally available pro-
Small molecule: drug, is the best studied of these inhibitors. This drug is
MCC950 rapidly hydrolysed by plasma and liver esterases into a
potent and selective inhibitor of caspase‑175. In a mouse
model of collagen-induced arthritis, VX‑765 ameliorated
IL-1β processing inhibitors the severity and progression of disease76. The caspase‑1
Orally available drug: inhibitor pralnacasan also decreases IL‑1β production
VX-765 Caspase-1
and cartilage damage in mice with streptococcal cell wall
(SCW)-induced arthritis, a model of chronic destructive
Recombinant protein: Pro-IL-1β IL-1β joint inflammation77. However, the effects of caspase‑1
Serine inhibitors in gout have not been explored. Of particular
AAT–Fc proteases
importance will be the specificity of the inhibitor for
caspase‑1, as caspases share a highly conserved catalytic
IL-1β inhibitors
core and off-target inhibition of apoptotic caspases could
cause undesirable consequences.
Antibody:
Canakinumab IL-1β Consistent with the findings indicating that AAT
inhibits inflammation in gout 62, a recombinant human
AAT–Fc fusion protein was found to be effective in a
IL-1R inhibitors mouse model of gouty arthritis63. AAT can modulate
Recombinant protein: inflammation at multiple levels, and whether the effects
observed in the gout model are directly caused by the
Anakinra inhibition of IL‑1β processing is unclear. Nonetheless,
these data indicate that AAT–Fc, and possibly other
serine protease inhibitors, could be of interest for the
treatment of gout attacks.
Figure 4 | Therapeutic targets in gouty inflammation. Pathways leading to IL‑1 signalling
Nature Reviews
can be inhibited at many different steps. Examples of compounds and drugs| Rheumatology
that have
been proposed to affect the various steps are shown. Whereas strategies that target IL‑1 inhibitors
potassium efflux or mitochondrial reactive oxygen species (ROS) production are quite The evidence for the clinical efficacy of IL‑1 inhibition in
unspecific, inhibitors of NLRP3 inflammasome assembly such as colchicine or inhibitors of acute gout has been reviewed elsewhere37. Two IL‑1 inhib­
IL‑1β and IL‑1 receptor (IL‑1R) have already been proven to be effective in gout. itors, canakinumab and anakinra, are currently available

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Available IL‑1 blocking drugs of patients, their use is restricted to patients who have
‘difficult to treat’ disease and their safety with respect to
Drug name Mode of Terminal half Molecular Route of infectious complications needs to be considered when
action life weight administration
they are prescribed.
Anakinra IL‑1 receptor 4–6 hours 17.3 kDa Subcutaneous
antagonist
Conclusions
Canakinumab Human 26 days 145 kDa Subcutaneous
The past 12 years have shown considerable progress in
anti‑IL‑1β our understanding of the mechanisms of inflammation
monoclonal
antibody and the role of innate immune sensors in gout; however,
several questions remain unanswered, with three areas
being of particular interest. Firstly, the detailed specific
for clinical use, but only canakinumab is indicated for mechanism by which NLRP3 is activated upon exposure
acute gout in Europe. The two drugs have different to MSU crystals is still unclear. One emerging possibility
mechanisms of action: canakinumab is a specific inhib­ is that NLRP3 acts as a guardian of cellular integrity by
itor of IL‑1β, and anakinra inhibits the binding of both detecting perturbations triggered when innate immune
IL‑1α and IL‑1β to IL‑1R1. The properties of these drugs cells attempt to engulf large particulates79. This concept
are listed in TABLE 1. would suggest that the size, and possibly the chemical
IL‑1 inhibition can relieve the acute symptoms of nature and shape, of those particulates (including MSU
gout in patients who have not responded to conventional crystals) could affect immune responses. Secondly,
treatments or in whom the use of NSAIDs, colchicine caspase‑1‑independent mechanisms of IL‑1 production,
or glucocorticoidssteroids are contraindicated. Although including which proteases are engaged and what triggers
IL‑1 inhibition is not recommended as a first-line anti-­ their activation, are still poorly understood. In this con­
inflammatory treatment, the results from clinical trials text it would be important to understand at what stage of
of canakinumab and cohort studies of patients who have the response these pathways contribute to the inflamma­
received anakinra as treatment for acute flares showed tory phenotype and what are the cell types orchestrating
a rapid onset of pain relief, with responses observed in this inflammasome-independent response. Finally, the
nearly all treated patients37. Patients who had subsequent mechanisms that initiate gouty attacks in patients who
flares responded equally well when treated again with have persistent MSU crystal deposits are a key area of
canakinumab78 and, in our personal experience, ana­ interest. Whether specific initiating mechanisms contrib­
kinra is also effective when given for recurrent flares. ute to triggering the inflammatory reaction, possibly by
In patients with severe renal and cardiac impairment, acting on priming signalling, or whether a decrease in the
a clinical situation that commonly makes the choice of negative regulation of NLRP3 engagement promotes the
acute therapy difficult, the use of IL‑1 inhibition has inflammatory cascade, remains unclear. A better under­
not been formally assessed in clinical trials; however, no standing of these processes might identify potential spe­
severe adverse effects have been reported in case series37. cific therapeutic strategies that will make the prevention
In the clinical trials involving canakinumab, a significant and management of gout more effective and specific. The
reduction of gout flares was seen for up to 6 months, with study of this old disease has provided us with a greater
canakinumab treatment also reducing gout flares at the understanding of inflammatory pathways; solving the
start of urate-lowering therapy; however, the drug is not remaining questions still bears enormous potential for
registered to reduce gout flares on starting therapy. As new discoveries of pathways and treatments that might
anti‑IL‑1 drugs have not been tested in large numbers have implications in several inflammatory diseases.

1. Kuo, C. F., Grainge, M. J., Zhang, W. & Doherty, M. 9. Aglietti, R. A. & Dueber, E. C. Recent insights into the 16. Burns, K., Martinon, F. & Tschopp, J. New insights into
Global epidemiology of gout: prevalence, incidence and molecular mechanisms underlying pyroptosis and the mechanism of IL‑1β maturation. Curr. Opin.
risk factors. Nat. Rev. Rheumatol. 11, 649–662 (2015). gasdermin family functions. Trends Immunol. 38, Immunol. 15, 26–30 (2003).
2. Dalbeth, N., Merriman, T. R. & Stamp, L. K. Gout. 261–271 (2017). 17. Holzinger, D. et al. Myeloid-related proteins 8 and 14
Lancet 388, 2039–2052 (2016). 10. Abhishek, A. et al. In vivo detection of monosodium contribute to monosodium urate monohydrate crystal-
3. McGonagle, D. & McDermott, M. F. A proposed urate crystal deposits by Raman spectroscopy — a pilot induced inflammation in gout. Arthritis Rheumatol.
classification of the immunological diseases. PLoS Med. study. Rheumatology (Oxford) 55, 379–380 (2016). 66, 1327–1339 (2014).
3, e297 (2006). 11. Kayagaki, N. et al. Caspase‑11 cleaves gasdermin D 18. Joosten, L. A. et al. Engagement of fatty acids with
4. Martinon, F. & Glimcher, L. H. Gout: new insights into an for non-canonical inflammasome signalling. Nature Toll-like receptor 2 drives interleukin‑1beta
old disease. J. Clin. Invest. 116, 2073–2075 (2006). 526, 666–671 (2015). production via the ASC/caspase 1 pathway in
5. Broz, P. & Dixit, V. M. Inflammasomes: mechanism of 12. Mulay, S. R. et al. Cytotoxicity of crystals involves monosodium urate monohydrate crystal-induced
assembly, regulation and signalling. Nat. Rev. Immunol. RIPK3‑MLKL-mediated necroptosis. Nat. Commun. 7, gouty arthritis. Arthritis Rheum. 62, 3237–3248
16, 407–420 (2016). 10274 (2016). (2010).
6. Martinon, F., Burns, K. & Tschopp, J. The 13. Vince, J. E. et al. Inhibitor of apoptosis proteins limit 19. Joosten, L. A., Abdollahi-Roodsaz, S., Dinarello, C. A.,
inflammasome: a molecular platform triggering RIP3 kinase-dependent interleukin‑1 activation. O’Neill, L. & Netea, M. G. Toll-like receptors and
activation of inflammatory caspases and Immunity 36, 215–227 (2012). chronic inflammation in rheumatic diseases: new
processing of pro-IL‑β. Mol. Cell 10, 417–426 (2002). 14. Liu-Bryan, R., Scott, P., Sydlaske, A., Rose, D. M. & developments. Nat. Rev. Rheumatol. 12, 344–357
7. Cai, X. et al. Prion-like polymerization underlies signal Terkeltaub, R. Innate immunity conferred by Toll- (2016).
transduction in antiviral immune defense and like receptors 2 and 4 and myeloid differentiation 20. An, L. L. et al. Complement C5a potentiates uric acid
inflammasome activation. Cell 156, 1207–1222 factor 88 expression is pivotal to monosodium crystal-induced IL‑1β production. Eur. J. Immunol. 44,
(2014). urate monohydrate crystal-induced inflammation. 3669–3679 (2014).
8. Chen, C. J. et al. MyD88‑dependent IL‑1 receptor Arthritis Rheum. 52, 2936–2946 (2005). 21. Khameneh, H. J. et al. C5a regulates IL‑1β production
signaling is essential for gouty inflammation 15. Martinon, F., Petrilli, V., Mayor, A., Tardivel, A. & and leukocyte recruitment in a murine model of
stimulated by monosodium urate crystals. Tschopp, J. Gout-associated uric acid crystals activate the monosodium urate crystal-induced peritonitis. Front.
J. Clin. Invest. 116, 2262–2271 (2006). NALP3 inflammasome. Nature 440, 237–241 (2006). Pharmacol. 8, 10 (2017).

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


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

22. Petrilli, V. et al. Activation of the NALP3 44. Guo, Z. et al. Intestinal microbiota distinguish gout 66. Youm, Y. H. et al. The ketone metabolite
inflammasome is triggered by low intracellular patients from healthy humans. Sci. Rep. 6, 20602 β‑hydroxybutyrate blocks NLRP3 inflammasome-
potassium concentration. Cell Death Differ. 14, (2016). mediated inflammatory disease. Nat. Med. 21,
1583–1589 (2007). 45. Ragab, G., Elshahaly, M. & Bardin, T. Gout: an old 263–269 (2015).
23. Yaron, J. R. et al. K+ regulates Ca2+ to drive disease in new perspective — a review. J. Adv. Res. 8, 67. Goldberg, E. L. et al. β‑Hydroxybutyrate deactivates
inflammasome signaling: dynamic visualization of ion 495–511 (2017). neutrophil NLRP3 inflammasome to relieve gout
flux in live cells. Cell Death Dis. 6, e1954 (2015). 46. Abhishek, A., Valdes, A. M. & Doherty, M. Low flares. Cell Rep. 18, 2077–2087 (2017).
24. Amaral, F. A. et al. NLRP3 inflammasome-mediated omega‑3 fatty acid levels associate with frequent gout 68. Jhang, J. J., Lu, C. C. & Yen, G. C. Epigallocatechin
neutrophil recruitment and hypernociception depend attacks: a case control study. Ann. Rheum. Dis. 75, gallate inhibits urate crystals-induced peritoneal
on leukotriene B4 in a murine model of gout. Arthritis 784–785 (2016). inflammation in C57BL/6 mice. Mol. Nutr. Food Res.
Rheum. 64, 474–484 (2012). 47. Yan, Y. et al. Omega‑3 fatty acids prevent 60, 2297–2303 (2016).
25. He, Y., Zeng, M. Y., Yang, D., Motro, B. & Nunez, G. inflammation and metabolic disorder through 69. Dhanasekar, C., Kalaiselvan, S. & Rasool, M. Morin,
NEK7 is an essential mediator of NLRP3 activation inhibition of NLRP3 inflammasome activation. a bioflavonoid suppresses monosodium urate crystal-
downstream of potassium efflux. Nature 530, Immunity 38, 1154–1163 (2013). induced inflammatory immune response in RAW
354–357 (2016). 48. Merriman, T. R. An update on the genetic architecture 264.7 macrophages through the inhibition of
26. Schmid-Burgk, J. L. et al. A genome-wide CRISPR of hyperuricemia and gout. Arthritis Res. Ther. 17, 98 inflammatory mediators, intracellular ROS levels and
(Clustered Regularly Interspaced Short Palindromic (2015). NF‑κB activation. PLoS ONE 10, e0145093 (2015).
Repeats) screen identifies NEK7 as an essential 49. McKinney, C. et al. Multiplicative interaction of 70. Kim, S. K., Choe, J. Y. & Park, K. Y. Rebamipide
component of NLRP3 inflammasome activation. functional inflammasome genetic variants in suppresses monosodium urate crystal-induced
J. Biol. Chem. 291, 103–109 (2016). determining the risk of gout. Arthritis Res. Ther. 17, interleukin‑1β production through regulation of
27. Shi, H. et al. NLRP3 activation and mitosis are mutually 288 (2015). oxidative stress and caspase‑1 in THP‑1 cells.
exclusive events coordinated by NEK7, a new 50. Lee, Y. H. & Bae, S. C. Association between functional Inflammation 39, 473–482 (2016).
inflammasome component. Nat. Immunol. 17, 250–258 NLRP3 polymorphisms and susceptibility to 71. Ives, A. et al. Xanthine oxidoreductase regulates
(2016). autoimmune and inflammatory diseases: macrophage IL‑1β secretion upon NLRP3 inflammasome
28. Dinarello, C. A. Immunological and inflammatory a meta-analysis. Lupus 25, 1558–1566 (2016). activation. Nat. Commun. 6, 6555 (2015).
functions of the interleukin‑1 family. Annu. Rev. 51. Qing, Y. F. et al. Association of TLR4 gene rs2149356 72. Perregaux, D. G. et al. Identification and characterization
Immunol. 27, 519–550 (2009). polymorphism with primary gouty arthritis in a case- of a novel class of interleukin‑1 post-translational
29. Schlesinger, N. & Thiele, R. G. The pathogenesis of control study. PLoS ONE 8, e64845 (2013). processing inhibitors. J. Pharmacol. Exp. Ther. 299,
bone erosions in gouty arthritis. Ann. Rheum. Dis. 69, 52. Rasheed, H. et al. The Toll-like receptor 4 (TLR4) variant 187–197 (2001).
1907–1912 (2010). rs2149356 and risk of gout in European and Polynesian 73. Coll, R. C. et al. A small-molecule inhibitor of the NLRP3
30. Dinarello, C. A. Infection, fever, and exogenous and sample sets. PLoS ONE 11, e0147939 (2016). inflammasome for the treatment of inflammatory
endogenous pyrogens: some concepts have changed. 53. Chang, W. C. et al. Genetic variants of PPAR‑γ diseases. Nat. Med. 21, 248–255 (2015).
J. Endotoxin Res. 10, 201–222 (2004). coactivator 1B augment NLRP3‑mediated 74. Primiano, M. J. et al. Efficacy and pharmacology of
31. Netea, M. G., van de Veerdonk, F. L., van der inflammation in gouty arthritis. Rheumatology the NLRP3 inflammasome inhibitor CP‑456,773
Meer, J. W., Dinarello, C. A. & Joosten, L. A. (Oxford) 56, 457–466 (2017). (CRID3) in murine models of dermal and pulmonary
Inflammasome-independent regulation of IL‑1‑family 54. Popa-Nita, O. & Naccache, P. H. Crystal-induced inflammation. J. Immunol. 197, 2421–2433
cytokines. Annu. Rev. Immunol. 33, 49–77 (2015). neutrophil activation. Immunol. Cell Biol. 88, 32–40 (2016).
32. Sugawara, S. et al. Neutrophil proteinase 3‑mediated (2010). 75. Wannamaker, W. et al. (S)-1-((S)-2-{[1-
induction of bioactive IL‑18 secretion by human oral 55. Schauer, C. et al. Aggregated neutrophil extracellular (4‑amino‑3‑chloro-phenyl)-methanoyl]-amino}-
epithelial cells. J. Immunol. 167, 6568–6575 (2001). traps limit inflammation by degrading cytokines and 3,3‑dimethyl-butanoy l)-pyrrolidine‑2‑carboxylic acid
33. Mizutani, H., Schechter, N., Lazarus, G., Black, R. A. & chemokines. Nat. Med. 20, 511–517 (2014). ((2R,3S)-2‑ethoxy‑5‑oxo-tetrahydro-furan‑3‑yl)-amide
Kupper, T. S. Rapid and specific conversion of 56. Desai, J. et al. PMA and crystal-induced neutrophil (VX‑765), an orally available selective interleukin (IL)-
precursor interleukin 1 beta (IL‑1β) to an active IL‑1 extracellular trap formation involves RIPK1–RIPK3– converting enzyme/caspase‑1 inhibitor, exhibits
species by human mast cell chymase. J. Exp. Med. MLKL signaling. Eur. J. Immunol. 46, 223–229 potent anti-inflammatory activities by inhibiting the
174, 821–825 (1991). (2016). release of IL‑1β and IL‑18. J. Pharmacol. Exp. Ther.
34. Echtermeyer, F. et al. Syndecan‑4 regulates 57. Carling, D. AMPK signalling in health and disease. 321, 509–516 (2007).
ADAMTS‑5 activation and cartilage breakdown in Curr. Opin. Cell Biol. 45, 31–37 (2017). 76. Zhang, Y. & Zheng, Y. Effects and mechanisms of
osteoarthritis. Nat. Med. 15, 1072–1076 (2009). 58. Wang, Y., Viollet, B., Terkeltaub, R. & Liu-Bryan, R. potent caspase‑1 inhibitor VX765 treatment on
35. Irmler, M. et al. Granzyme A is an interleukin AMP-activated protein kinase suppresses urate collagen-induced arthritis in mice. Clin. Exp.
1β‑converting enzyme. J. Exp. Med. 181, 1917–1922 crystal-induced inflammation and transduces Rheumatol. 34, 111–118 (2016).
(1995). colchicine effects in macrophages. Ann. Rheum. Dis. 77. Joosten, L. A. et al. Inflammatory arthritis in caspase
36. Pazar, B. et al. Basic calcium phosphate crystals 75, 286–294 (2016). 1 gene-deficient mice: contribution of proteinase 3 to
induce monocyte/macrophage IL‑1β secretion through 59. Liote, F. et al. Inhibition and prevention of caspase 1‑independent production of bioactive
the NLRP3 inflammasome in vitro. J. Immunol. 186, monosodium urate monohydrate crystal-induced interleukin‑1β. Arthritis Rheum. 60, 3651–3662
2495–2502 (2011). acute inflammation in vivo by transforming growth (2009).
37. Dumusc, A. & So, A. Interleukin‑1 as a therapeutic factor β1. Arthritis Rheum. 39, 1192–1198 78. Alten, R. et al. Efficacy of canakinumab on
target in gout. Curr. Opin. Rheumatol. 27, 156–163 (1996). re‑treatment in gouty arthritis patients with limited
(2015). 60. Chen, Y. H. et al. Spontaneous resolution of acute treatment options: 24‑week results from β‑RELIEVED
38. Kienhorst, L. B. et al. Gout is a chronic inflammatory gouty arthritis is associated with rapid induction of the and β‑RELIEVED‑II [abstract]. Arthritis Rheum. 63
disease in which high levels of interleukin‑8 (CXCL8), anti-inflammatory factors TGFβ1, IL‑10 and soluble (Suppl.), S402 (2011).
myeloid-related protein 8/myeloid-related protein 14 TNF receptors and the intracellular cytokine negative 79. Hornung, V. et al. Silica crystals and aluminum salts
complex, and an altered proteome are associated with regulators CIS and SOCS3. Ann. Rheum. Dis. 70, activate the NALP3 inflammasome through
diabetes mellitus and cardiovascular disease. Arthritis 1655–1663 (2011). phagosomal destabilization. Nat. Immunol. 9,
Rheumatol. 67, 3303–3313 (2015). 61. Galvao, I. et al. Annexin A1 promotes timely 847–856 (2008).
39. Crisan, T. O. et al. Soluble uric acid primes TLR- resolution of inflammation in murine gout.
induced proinflammatory cytokine production by Eur. J. Immunol. 47, 585–596 (2017). Acknowledgements
human primary cells via inhibition of IL‑1Ra. 62. Ter Horst, R. et al. Host and environmental factors The work of F.M.is supported by a grant from the Swiss
Ann. Rheum. Dis. 75, 755–762 (2016). influencing individual human cytokine responses. National Science Foundation (310030_173152).
40. Choi, H. K., Atkinson, K., Karlson, E. W., Willett, W. & Cell 167, 1111–1124.e13 (2016).
Curhan, G. Purine-rich foods, dairy and protein intake, 63. Joosten, L. A. et al. Alpha‑1‑anti-trypsin‑Fc fusion Author contributions
and the risk of gout in men. N. Engl. J. Med. 350, protein ameliorates gouty arthritis by reducing release Both authors researched data and made a substantial contri-
1093–1103 (2004). and extracellular processing of IL‑1β and by the bution to discussion of the content and writing the article,
41. Zhang, Y. et al. Purine-rich foods intake and recurrent induction of endogenous IL‑1Ra. Ann. Rheum. Dis. 75, and reviewed or edited the manuscript before submission.
gout attacks. Ann. Rheum. Dis. 71, 1448–1453 (2012). 1219–1227 (2016).
42. Vieira, A. T. et al. A role for gut microbiota and the 64. Elliot, A. J., Cross, K. W. & Fleming, D. M. Seasonality Competing interests statement
metabolite-sensing receptor GPR43 in a murine and trends in the incidence and prevalence of gout in A.K.S. declares that he has acted as a consultant for
model of gout. Arthritis Rheumatol. 67, 1646–1656 England and Wales 1994–2007. Ann. Rheum. Dis. AstraZeneca, Menarini and Novartis. F.M. declares no com-
(2015). 68, 1728–1733 (2009). peting interests.
43. Vieira, A. T. et al. Dietary fiber and the short-chain 65. Misawa, T. et al. Microtubule-driven spatial
fatty acid acetate promote resolution of neutrophilic arrangement of mitochondria promotes activation of Publisher’s note
inflammation in a model of gout in mice. J. Leukocyte the NLRP3 inflammasome. Nat. Immunol. 14, Springer Nature remains neutral with regard to jurisdictional
Biol. 101, 275–284 (2017). 454–460 (2013). claims in published maps and institutional affiliations.

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like