Você está na página 1de 14

Opinion

IL-1 Family Cytokine Pathways Underlying


NAFLD: Towards New Treatment Strategies
Andreea-Manuela Mirea,1 Cees J. Tack,1 Triantafyllos Chavakis,2
Leo A.B. Joosten,1 and Erik J.M. Toonen1,3,*

Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease
worldwide. Pathways responsible for the activation of IL-1 family cytokines are
key in the development of NAFLD but underlying mechanisms are not fully
understood. Many studies have focused on the inflammasome–caspase-1
pathway and have shown that this pathway is an important inducer of inflam-
mation in NAFLD. However, this pathway is not solely responsible for the
activation of proinflammatory cytokines. Also, neutrophil serine proteases
(NSPs) are capable of activating cytokines and recent studies reported that
these proteases also contribute to NAFLD. These studies provided, for the first
time, evidence that this inflammasome-independent pathway is involved in
NAFLD. In our opinion, these new insights open up new approaches for thera-
peutic intervention.

Nonalcoholic Fatty Liver Disease: An Increasing Public Health Issue


Nonalcoholic fatty liver disease (NAFD) is one of the most common form of chronic liver disease
worldwide. The prevalence of NAFLD in the general population of Western countries is 17%–
46% and it is rapidly increasing in parallel with the increasing prevalence of obesity and
metabolic syndrome [1]. NAFLD can range from simple steatosis [nonalcoholic fatty liver
(NAFL)] to nonalcoholic steatohepatitis (NASH), characterized by liver inflammation and hepa-
tocyte ballooning, with or without fibrosis. Due to this wide spectrum of conditions, NAFLD and
NASH are associated with increased morbidity and mortality [2]. Given the high prevalence and
the fact that no treatment is available, a better understanding of its pathogenesis is essential to
be able to identify targets for drug development in order to overcome the epidemic.

Although the mechanisms underlying obesity-induced NAFLD are not fully understood, it is
known that the induction of inflammation and production of inflammatory cytokines such as
TNF and members of the IL-1 cytokine family (see Glossary) play a crucial role in disease
process [3,4]. Many pathogens or danger signals derived from the host are able to trigger innate 1
Department of Internal Medicine,
immune responses that contribute to liver diseases. Innate immune cells in the liver are able to Radboud University Medical Center,
sense molecules derived from pathogens, commonly known as pathogen-associated Nijmegen, The Netherlands
2
Institute for Clinical Chemistry and
molecular patterns (PAMPs) and also endogenous alarm signals known as damage- Laboratory Medicine, University Clinic
associated molecular patterns (DAMPs) through specific receptors called pattern rec- Carl-Gustav-Carus, Technische
ognition receptors (PRRs). After binding to PRRs, both PAMPs and DAMPs trigger the Universität Dresden, Dresden,
Germany
immune system to produce and activate proinflammatory cytokines. It is now established that 3
R&D Department, Hycult Biotech,
the assembly of a cytosolic protein complex called the NLRP3 inflammasome is important for Uden, The Netherlands
the activation of proinflammatory cytokines. This protein complex activates the protease
caspase-1, which cleaves pro-IL-1b and pro-IL-18, converting them to their active forms
*Correspondence:
[5]. Several studies investigating both NAFLD mouse models and patients with metabolic e.toonen@hycultbiotech.com (Erik J.M.
syndrome have shown that some inflammasomes and their downstream effects are important Toonen).

458 Trends in Molecular Medicine, May 2018, Vol. 24, No. 5 https://doi.org/10.1016/j.molmed.2018.03.005
© 2018 Elsevier Ltd. All rights reserved.
for the induction and prolongation of obesity-induced NAFLD [5,6]. However, this inflamma- Glossary
some-dependent pathway is not solely responsible for the activation of proinflammatory Alpha-1 antitrypsin (AAT): a serine
cytokines. The neutrophil serine proteases (NSPs) neutrophil elastase (NE), proteinase peptidase inhibitor produced by the
3 (PR3), and cathepsin G (CG) are also capable of processing proinflammatory cytokines into liver. Among its targets are neutrophil
serine proteases and caspase-1.
their bioactive form. NSPs are responsible for the activation of cytokines such as membrane- Anakinra: a recombinant form of IL-
bound TNF, IL-1a, IL-1b, IL-18, and IL-33 [7,8]. Recent studies investigating mouse models of 1Ra, the natural inhibitor of IL-1.
obesity-induced NAFLD showed for the first time that these inflammasome-independent Anakinra inhibits the binding of IL-1a
and IL-1b to their receptor.
mechanisms are also important for the induction of liver inflammation and progression into
Canakinumab: a human monoclonal
liver fibrosis [9–11]. In our opinion, these recent findings provide new insights into how antibody that inhibits IL-1b effects by
dysregulation of inflammatory pathways contribute to the development of NAFLD and NASH. binding it and preventing ligation to
In addition, these results open up new perspectives for therapeutic intervention. its receptor.
Caspase-1: a molecule associated
with the NLRP3 complex. Upon
Here, we will first provide an overview of the recent literature regarding the role of proinflam- inflammasome activation, the inactive
matory cytokines in the development of NAFLD and NASH. We will focus on both the underlying form called pro-caspase-1 is cleaved
inflammasome-dependent and inflammasome-independent mechanisms responsible for the and further processes IL-1b and IL-
18 to their active forms.
processing and activation of these cytokines during the development of NAFLD and NASH.
Damage-associated molecular
Although we will provide a full overview of these mechanisms, we will emphasize recent findings patterns (DAMPs): self-molecules
regarding the involvement of the less-established inflammasome-independent pathways in capable of initiating an inflammatory
NAFLD and NASH. Additionally, we discuss the potential of targeting these pathways as novel response. They can be represented
by nuclear or mitochondrial DNA,
therapeutic approaches in obesity-induced NAFLD. heat shock proteins, metabolites
such as ATP, uric acid, and
IL-1 Family Cytokines in Obesity-Induced NAFLD cholesterol crystals.
IL-1 family cytokines are one of the main drivers of inflammation in NAFLD. When activated, Dipeptidyl peptidase I (DPPI): an
enzyme that processes neutrophil
these proinflammatory cytokines are able to disrupt insulin and lipid signaling pathways, serine proteases to their active
thereby influencing insulin sensitivity and lipid metabolism [12]. The cytokines IL-1b, IL-1a, forms.
IL-18, and IL-33, all members of the IL-1 family, are most intensively studied for their role in IL-1 cytokine family: molecules
that mediate innate immune
NAFLD.
responses. They display both
proinflammatory and anti-
IL-1b has an important role in liver disease, being involved in all stages of the disease. It inflammatory functions. Among these
promotes liver steatosis, inflammation, and fibrosis by signaling through the IL-1 receptor, molecules, both IL-1 isoforms (IL-1a
and IL-1b), IL-18, and IL-33 exhibit
which is widely expressed on the different liver cell subpopulations [13]. IL-1b promotes hepatic
proinflammatory properties.
steatosis by stimulating triglycerides and cholesterol accumulation in primary liver hepatocytes Neutrophil serine proteases
and lipid droplet formation [4]. Acting on liver sinusoidal endothelial cells, IL-1b promotes liver (NSPs): antimicrobial peptide stores
inflammation by upregulating ICAM-1 (intercellular adhesion molecule 1) expression, which in the azurophilic granules of
neutrophils.
attracts neutrophils in the liver. This effect was elegantly demonstrated in an in vivo model,
NLRP3 (NOD-like receptor P3)
where wild type (WT) C57Bl/6 mice and Icam1 deficient mice were exposed to Il-1b expression inflammasome: an intracellular
by injecting a nonreplicating adenoviral vector that contained the Il-1b gene. After 4 days there protein complex formed by the
was a significant influx of neutrophils in the livers of WT mice but not in the livers of Icam1 NACHT, LRR, and PYD domains, an
adaptor molecule called ASC
deficient mice. Blocking other adhesion molecules in the WT mice did not affect neutrophil (apoptosis associated peck-like
recruitment, suggesting that IL-1b stimulates neutrophil recruitment in the liver through ICAM-1 protein containing a CARD) and pro-
[14]. In addition, IL-1b stimulates local inflammation by inducing production of IL-6, another caspase-1. NLRP3 components
proinflammatory cytokine [15]. Several mouse models of sterile inflammation in the liver have assemble upon PRR stimulation and
initiate activation of IL-1b and IL-18
shown that IL-1b, together with IL-6 and TNF, activate local immune cells and attract other through the effector protein caspase-
leucocytes to the liver, leading to a chronic inflammatory state [16,17]. Finally, IL-1b contributes 1.
to the progression from liver inflammation to liver fibrosis, as shown in different rodent models of Pathogen-associated molecular
patterns (PAMPs): constitutive
liver fibrosis. For example, in a mouse model of thioacetamine-induced liver fibrosis, Il-1r (Il-1
components of pathogens (sugar or
receptor) knockout (k.o.) mice were protected from liver fibrosis, as assessed by hematoxylin lipid molecules from the cell wall,
and eosin staining. When compared with WT mice, these mice showed lower alanine trans- nucleic acids) that are able to trigger
aminase (ALT) concentrations in plasma and lower mRNA expression of a-smooth muscle actin an immune response.
(aSMA), a marker for hepatic stellate cells (HSC) activation [18]. In another study, Il-1r k.o. mice

Trends in Molecular Medicine, May 2018, Vol. 24, No. 5 459


were fed a choline-deficient amino acid defined (CDAA) diet, a well-established diet for the Pattern recognition receptors
induction of inflammation and liver fibrosis. Il-1r k.o. mice showed, when compared with WT (PRRs): special receptors of the
innate immune system capable of
controls, lower ALT plasma concentration and lower mRNA expression of the fibrosis markers detecting PAMPs and DAMPs.
collagen type I alpha 1 (Col1a1) and collagen type IV alpha 1 (Col4a1). Also, lower mRNA
expression was observed for Timp-1 (tissue inhibitor of metalloproteinases), a molecule that
prevents degradation of the extracellular matrix and inhibits HSC apoptosis, thereby favoring
the process of fibrosis. Furthermore, HSCs were isolated from both WT and Il-1r k.o. mice and
stimulated with IL-1b. HSCs from WT mice had an increased production of TIMP-1 protein,
elevated mRNA expression of Col1a1 and Col4a1, and suppressed mRNA expression of
Bambi (BMP and activin membrane-bound inhibitor), an inhibitor of TGF-b, the main promoter
of liver fibrosis [19]. In conclusion, IL-1b promotes liver fibrosis by stimulating HSC activation,
proliferation, and survival and by inducing the production of profibrogenic factors. Altogether,
these studies indicate that IL-1b has a major role in development of NAFLD by contributing to all
the steps of the disease, ranging from simple steatosis to steatohepatitis and liver fibrosis.

Next to IL-1b, IL-1a has been investigated regarding its role in metabolic disturbances. The
induction of steatohepatitis has been assessed in Il-1a k.o. mice fed an atherogenic diet for 18
weeks [16]. The study indicated that these mice were protected from developing steatohe-
patitis when compared with WT mice that were fed the same diet. Il-1a k.o mice showed
improved liver histology and less injury and inflammation, suggested by lower serum ALT and
serum amyloid alpha concentrations. In addition, Il-1a k.o. mice had decreased expression of
inflammatory and fibrosis genes mRNA, such as Il-1b, Il-6, Tnf, P-selectin, Cxcl1, and Tgf-b
when compared with WT mice [20].

IL-18 is also implicated in NAFLD and other metabolic conditions but results between human
and animal studies are contradictory. Studies using mice deficient for Il-18 reported that these
mice develop features of the metabolic syndrome, like dyslipidemia, obesity, atherosclerosis,
and insulin resistance, thereby suggesting a protective role for this cytokine in glucose
homeostasis and lipid metabolism [21,22]. In contrast, patients with metabolic syndrome
showed a positive correlation between plasma concentration of IL-18 and the development
of atherosclerosis, insulin resistance, and type 2 diabetes, suggesting that IL-18 might promote
the development of the metabolic syndrome [23–25]. These intriguing but contradictive results
urge the need for further studies in order to establish whether high plasma concentrations of IL-
18 are indeed involved in promoting inflammation and metabolic disturbances, or whether the
increased concentrations reflect a compensatory mechanism.

Recently, a newly described member of the IL-1 family cytokines, IL-33, was also found to be
involved in the later stages of NAFLD, whereby fibrosis is prominent. Mouse models of
experimentally induced liver fibrosis showed that Il-33 mRNA expression was higher in fibrotic
mice than WT controls [26]. Mouse models of diet-induced liver steatosis and inflammation
showed that administration of IL-33 intraperitoneally worsened the liver fibrosis content by
inducing a shift of invading macrophages and T cells towards M2 macrophages and T helper2
cells, respectively, which were abundant in IL-33-treated mice and produced profibrotic
cytokines (IL-4, IL-5, IL-13) [26]. In line with these findings, studies on patients with liver fibrosis
versus healthy controls showed increased serum concentrations of IL-33 in those with liver
fibrosis, increased mRNA expression of IL-33, and increased IL-33 protein content in the
fibrotic livers, suggesting a role for this cytokine in the mechanisms of liver fibrosis [26–28].

These studies clearly show that proinflammatory cytokines are key in the onset and prolonging
of NAFLD. Impairing the action of these cytokines could be a potentially successful approach

460 Trends in Molecular Medicine, May 2018, Vol. 24, No. 5


for treating the disease. We are convinced that preventing their activation is a promising
strategy, as these activation pathways are responsible for the activation of multiple proin-
flammatory cytokines. By targeting these underlying activation pathways, multiple cytokines are
inhibited by a single drug. It is therefore of utmost importance to have a clear and comprehen-
sive overview of all the molecular mechanisms responsible for cytokine activation in NAFLD.

The Role of NSPs in Liver Steatosis and Steatohepatitis


The discovery of the NLRP3 inflammasome provided an important molecular mechanism for
the activation of cytokines and induction of inflammation (Box 1). Although important and well-
established, the NLRP3 inflammasome and its effector protein caspase-1 are not the only
mechanisms responsible for activating proinflammatory cytokines. NSPs PR3, NE, and CG are
also able to process/activate cytokines, thereby regulating inflammatory responses (Figure 1)
[35]. Next to neutrophils, macrophages are also able to produce PR3 [35].

Although it is known that NSPs are involved in the onset and prolongation of several ‘classical’
inflammatory and infectious diseases [36], their role in inflammation-associated metabolic
diseases like obesity-induced type 2 diabetes, NAFLD, and NASH is less well investigated.
Given that IL-1 family cytokines are drivers of inflammation contributing to the disease process,
it is very well possible that NSPs, as underlying mechanisms for cytokine activation, are also
involved in these metabolic diseases. So far, studies investigating the role of these enzymes
show interesting results. PR3 has been shown to play an important role in insulin resistance by
degrading insulin-like growth factor 1 (IGF-1) and insulin-like growth factor binding protein 3
(IGFBP-3) (two molecules involved in insulin signaling) in an in vitro system using L6 skeletal
muscle cells that express IGF-1 [31]. In vivo experiments from the same study showed that
administration of PR3 in WT Balb/c mice alters the glucose tolerance compared with mice that
were administered vehicle. In addition, in another study, 8 out of 36 type 2 diabetes patients
had high protein concentrations of PR3 in their urine, while no PR3 was detected in the urine
from 36 healthy volunteers: the authors concluded that PR3 may contribute to the development
of type 2 diabetes in at least a subset of patients [37].

As well as PR3, NE has been shown to play a role in metabolic conditions by promoting insulin
resistance, liver steatosis, and obesity-driven inflammation [11,38]. For example, NE activity
was shown to be increased in the serum of both obese (ob/ob) mice and WT mice fed a high-fat

Box 1. The NLRP3 Inflammasome and NAFLD


The discovery of the NLRP3 inflammasome provided an important molecular mechanism for the activation of cytokines and induction of inflammation. The NLRP3
protein complex activates the protease caspase-1, which cleaves pro-IL-1b and pro-IL-18, thereby converting them to their active forms [29]. In the liver, the NLRP3–
caspase-1 complex is predominantly expressed by Kupffer cells, resident macrophages of the liver, but other inflammatory cells and parenchymal cells are also
known to be able to express the complex [5]. During recent years, many studies have focused on caspase-1 and the NLRP3 inflammasome and their role in liver
diseases. Several mouse models have shown that the NLRP3 inflammasome is activated in liver steatosis (Figure I), promoting progression to steatohepatitis and liver
fibrosis [30]. For example, mice deficient for the Nlrp3 gene did not develop liver steatohepatitis when fed a CDAA when compared with WT controls on the same diet
[31]. In contrast, mice overexpressing Nlrp3 and fed a normal diet, developed severe liver inflammation and fibrosis when compared with WT controls [31]. These
results revealed that long signaling via the NLRP3 inflammasome leads to liver inflammatory changes and development of liver steatohepatitis and liver fibrosis [31]. In
support of these findings, studies investigating patients with NASH showed that these patients have increased mRNA expression of the NLRP3 inflammasome gene
and of the genes PYCARD (PYD and CARD domain containing; members of the inflammasome protein complex), CASP1 (caspase-1), IL-1b, and IL-18 when
compared with patients having simple steatosis [31,32].

Caspase-1 itself is also involved in mediating inflammation in NAFLD and promoting the progression to steatohepatitis and fibrosis. Knockout of caspase-1 in mice
fed a NAFLD/NASH-inducing diet, showed a protective effect regarding the development of liver steatosis, steatohepatitis, and early fibrogenesis when compared
with WT age-matched controls [33,34].

Taken together, these studies clearly indicate that inflammasome-mediated activation of proinflammatory cytokines is an important mechanism in the development of
inflammation-induced NAFLD and NASH.

Trends in Molecular Medicine, May 2018, Vol. 24, No. 5 461


DAMP

T
L
R
4 (B)
(A)
InflammaƟon

Pro-caspase-1
NLRP3
MyD88

ASC
IL-1B

IL-18

pa se-1
Cas B
NF-κB IL-1
8
IL-1

-1B
Pro-IL
-18
NF-κB

Pro-IL
DNA transcripƟon

Figure I. The Role of the NLRP3 Inflammasome in IL-1 Family Cytokine Activation. Activation of the NLRP3 inflammasome and IL-1 family cytokines is a
two-step process. In the first step (A) a PAMP, such as LPS, binds to its receptor (TLR4 for LPS) and activates via the adaptor protein MyD88 the transcription factor
NF-kB. Next, NF-kB will translocate to the nucleus and upregulate the transcription of genes encoding for the NLRP3 protein complex and the proinflammatory
cytokines IL-1b and IL-18. In the second step (B), a DAMP signal leads to association of the NLRP3 complex and activation of the effector protein caspase-1. In turn,
caspase-1 cleaves pro-IL-1b and pro-IL-18, thereby releasing their active forms. These cytokines will be excreted from the cells and promote local (and systemic)
inflammation. Abbreviations: ASC, apoptosis-like associated protein containing a CARD domain (component of the inflammasome); DAMP, damage-associated
molecular pattern; LPS, lipopolysaccharide; MyD88, myeloid differentiation primary response 88; NF-kB, nuclear factor kB; NLRP3, NOD-like receptor P3; PAMP,
pathogen-associated molecular pattern; (pro)-IL-1b, (pro) interleukin-1b; (pro)-IL-18, (pro)-interleukin 18; TLR4, toll-like receptor.

diet (HFD) [11]. Additionally, serum concentrations of the NSP inhibitor alpha-1 antitrypsin
(AAT) were severely reduced in these mice, suggesting that an imbalance between NE activity
and its regulator might lead to obesity-driven metabolic syndrome. This theory was also
confirmed by the fact that NE deficient mice and mice overexpressing human AAT, fed a
HFD, failed to develop liver steatosis, insulin resistance, and adipose tissue inflammation when
compared with WT age-matched controls on the same diet [11]. Moreover, a similar imbalance
between NE activity and AAT protein concentrations was observed in the serum of obese
subjects when compared with lean subjects [11]. In line with this data, another study showed an
increased ratio between plasma concentrations of NE and AAT (hence, an increasing dis-
balance between NE and AAT concentrations) in patients with NAFLD that correlated with
disease severity ranging from simple steatosis to NASH or liver fibrosis [10]. These results lead
to the idea that a disequilibrium between protein expression of NE and AAT might (at least
partly) be responsible for the development of obesity-related liver steatosis, insulin resistance,
and adipose tissue inflammation. Supporting this data, low AAT concentrations were associ-
ated with development of obesity, cardiovascular disease, and insulin resistance [39,40].

The studies reviewed above reported results that are in line with our own observations. We
recently investigated the development of NAFLD and insulin resistance in double k.o. mice for

462 Trends in Molecular Medicine, May 2018, Vol. 24, No. 5


(D) InflammaƟon
NETs

(B)
(C) NE
Pro-IL-1B PR3 IL-

Azurophilic Pro-IL-1B
granule CG -18

T
Phagolysosome
(E)

(A)

AAT
Pro-IL-1B IL-1B
NE PR3

Pro-IL-1B CG IL-18
Nucleus

Figure 1. The Role of Neutrophil Serine Proteases in IL-1 Family Cytokine Activation. NSPs are stored in the azurophilic granules of neutrophils. Upon
neutrophil activation, neutrophil serine proteases will be released into the cytosol (A), where they can activate proinflammatory cytokines. They can also be released into
the phagolysosome, where they have antimicrobial effects (B), or outside the cell where they can bind to the membrane (C) and activate extracellular cytokines, form
NETs (D), and migrate into the extracellular space and activate proinflammatory cytokines (E). In the extracellular matrix, AAT can inhibit NSPs, which leads to the
inhibition of cytokine activation (E). Abbreviations: AAT, alpha-1 antitrypsin; CG, cathepsin G; NE, neutrophil elastase; NET, neutrophil extracellular trap; NSP, neutrophil
serine protease; PR3, proteinase 3.

NE/PR3 and NE/CG and in WT control mice. We showed that these double k.o. mice were,
after a HFD, protected from developing insulin resistance, liver steatosis, and obesity-induced
inflammation, suggesting an important role for NSPs in NAFLD development [9].

Although we believe that the proteases NE and PR3 are at least partly responsible for the onset
and prolongation of NAFLD and NASH, other factors may also be involved. For instance, it
might be interesting to investigate the role of neutrophil extracellular traps (NETs) in NAFLD and
NASH. NETs have an important role during infection by helping neutrophils to capture and kill
pathogens. They contain proteins from azurophilic granules such as PR3, NE, CG, and
myeloperoxidase and use these proteases for disarming pathogens. Evidence suggests that
uncontrolled or excessive production of NETs is related to the exacerbation of inflammation,
thereby contributing to the development of autoimmunity, cancer, and inappropriate throm-
bosis [41]. Also, granzyme B, a serine protease expressed by natural killer cells and cytotoxic T
cells, might be an interesting candidate for further investigation for its role in NAFLD and NASH
[42]. However, at this moment, studies providing evidence that these factors are involved in
NAFLD or NASH are lacking.

Collectively, these recent findings provide new insights pointing towards alternative mecha-
nisms that are, next to inflammasomes, very important in the process of inflammatory-driven
induction of metabolic conditions.

Trends in Molecular Medicine, May 2018, Vol. 24, No. 5 463


Therapeutic Approaches in NAFLD
Given the prominent role of inflammatory cytokines in NAFLD and NASH, it is a conceivable
approach to target these cytokines to treat the disease. IL-1 signaling can be inhibited in
humans using canakinumab [43] or anakinra (Table 1). Although not studied in NAFLD,
administration of anakinra in patients with type 1 or type 2 diabetes showed an improvement of
insulin sensitivity and reduced systemic inflammation [44,45], which could be also beneficial in
NAFLD where insulin resistance and systemic inflammation contribute to progression from
simple steatosis to steatohepatitis. However, at this point it is unclear whether targeting a single
proinflammatory cytokine is sufficient to attenuate inflammation, as multiple inflammatory
signals are involved in the pathogenesis of these conditions. In this context, new insights that
both inflammasomes and NSPs are involved in the development of NAFLD and NASH are of
particular interest and offer new targets for therapeutic intervention. Targeting inflammasomes
and/or NSPs prevents the activation of multiple proinflammatory cytokines instead of only one.
This strategy is appealing, especially in the view that no treatments are available. Currently,
several compounds targeting inflammasomes or NSP pathways are being investigated as a
potential therapy for NAFLD and NASH (Table 1).

Regarding inflammasomes, several reagents inhibiting this pathway have been developed and
evaluated in animal models of liver diseases [46–52]. The efficacy of sulforaphane (SFN), an
NLRP3 inhibitor, has been investigated in a mouse model for obesity-induced NAFLD [46]. Mice
treated with SFN showed decreased caspase-1 activity, reduced IL-1b production in the liver,
and reduced steatosis. Although steatohepatitis was not assessed in this study, these results
are encouraging since SFN appears to show efficacy at least in the early stages of NAFLD [46].
These results clearly indicate that NLRP3 inhibition might be promising for treating metabolic
disorders. However, no NLRP3 inhibitor has been tested yet in a clinical setting.

For caspase-1 inhibition, most compounds described target all the caspases in the cell (pan-
caspase inhibitors), instead of caspase-1 alone [53–56]. Studies investigating these com-
pounds are more focused on caspases involved in apoptosis, another mechanism for liver
inflammation, as opposed to caspase-1, which promotes inflammation by activating proin-
flammatory cytokines. An inhibitor that is specific for caspase-1, namely Ac-YVAD, has been
investigated using low-density lipoprotein receptor k.o. mice (LDLR–/–), Leiden mice that
display features of the metabolic syndrome when fed a HFD [57]. Mice treated with this
caspase-1 inhibitor showed reduced liver steatosis and reduced liver fibrosis, as assessed
by Sirius red staining. In addition, these mice had less neutrophil infiltrates, lower ALT plasma
concentration, and reduced Tnf gene expression in the liver [57]. Taken together, these results
show that inhibiting the inflammasomes and/or caspase-1 has potential for the treatment of
NAFLD.

As discussed above, NSPs are also able to regulate inflammation, thereby contributing to liver
diseases. Therefore, these pathways might offer promising targets for therapeutic intervention
as well. An interesting candidate for inhibiting this pathway is AAT, the natural inhibitor of NSPs
(Figure 1). Next to NSPs, AAT was described as inhibiting caspase-1 in a mouse model of acute
myocardial infarction [58]. Therefore, using a compound that is able to inhibit both NSPs and
caspase-1, might be very effective for treating sterile inflammation. Several groups have
reported promising findings regarding the use of AAT as an effective and safe treatment for
inflammatory diseases. The potential role of AAT as a drug has been investigated in a mouse
model for rheumatoid arthritis. Human AAT protein therapy delayed onset, ameliorated disease
manifestations, and reduced autoimmunity in arthritic mice when compared with control mice
that were administered a saline solution [59].

464 Trends in Molecular Medicine, May 2018, Vol. 24, No. 5


Table 1. Potential Pharmacological Agents for NAFLD
Targeted Pharmacological Targeted Targeted Species Methods and results Conclusion Refs
pathway agent molecule diseases regarding
efficacy

IL-1b signaling Canakinumab IL-1b Cardiovascular Human 10 061 patients with previous myocardial Good [43]
disease infarction and hsCRP 2 mg/l were
administered placebo and three different
doses of canakinumab at a ratio 1.5:1:1:1.
Patients with canakinumab had significant
reduction of hsCRP at 48 months and IL-6 at
12 months. Patients that received 150 mg
canakinumab had a 15% reduced risk of a
secondary cardiovascular event than patients
on placebo.

Anakinra IL-1 receptor1 Type 1 and Human After receiving anakinra for 8 days, 14 type 1 Good [44]
type 2 diabetes patients had lower counts in
diabetes neutrophils and leucocytes, improved insulin
sensitivity assessed by euglycemic
hyperinsulinemic clamp with lower glucose
concentrations, lower dose of insulin used
that lasted for 4 weeks, and a decrease of
HbA1c at 4 weeks.

70 patients with type 2 diabetes were divided Good [45]


in two groups and administered either
anakinra or placebo subcutaneously for 13
weeks. Patients that received anakinra had
lower HbA1c, lower fasting plasma glucose
concentrations, lower neutrophil counts, and
improved beta-cell secretory function as
assessed by the ratio proinsulin to insulin
when compared with patients that received
placebo.

NLRP3 and Sulforaphane NLRP3 NAFLD Mice Sulforaphane prevented development of Mild [46]
caspase-1 inflammasome hepatic steatosis in a mouse model of
obesity-induced NAFLD, as assessed by
histological examination and hepatic lipid
content. Mice that received sulforaphane had
lower ALT plasma levels, lower IL-1b protein
content in the liver and lower mRNA
expression of ASC and caspase-1
(components of the NLRP3 inflammasome),
as well as reduced caspase-1 activity in the
liver when compared with mice that received
placebo. All these results suggest that
targeting NLRP3 inflammasome in the liver
could prevent development of liver steatosis.

GW50 1516 NLRP3 NAFLD Mice, Administration of GW501516 in a mouse Mild [47]
inflammasome/ human model of HFD-induced obesity with LPS
PPAR delta cells challenge significantly reduced serum ALT
and AST levels and prevented development of
hepatic steatosis when compared with
controls. However, there was no difference in
mRNA levels of caspase-1 and mRNA and
protein expression of IL-1 b in the liver, so the
observed effect was independent of NLRP3
inflammasome pathway. In human HepG2
cells challenged with proinflammatory stimuli,
administration of GW501516 significantly

Trends in Molecular Medicine, May 2018, Vol. 24, No. 5 465


Table 1. (continued)
Targeted Pharmacological Targeted Targeted Species Methods and results Conclusion Refs
pathway agent molecule diseases regarding
efficacy
reduced NLRP3 inflammasome mRNA
expression.

Isoliquiritigenin NLRP3 HFD-induced Mice, Isoliquiritigenin prevented development of Mild [48]


inflammasome obesity human hepatic steatosis in a mouse model of HFD-
cells induced obesity by reducing hepatic lipid
content. Mice that received isoliquiritigenin
had improved insulin sensitivity, reduced ALT
levels, and reduced mRNA expression of
proinflammatory cytokines and NLRP3
inflammasome components in the adipose
tissue when compared with controls. In
mouse bone marrow-derived macrophages
and in human THP-1 macrophages
challenged with LPS and ATP, administration
of isoliquiritigenin significantly decreased
active protein expression of caspase-1 and
IL-1b, suggestive for an inhibition of NLRP3
inflammasome activity.

Β- NLRP3 Inflammation Mouse Β-hydroxybutirate inhibited activation of Good [49]


hydroxybutirate inflammasome cells caspase-1 and IL-1b, assessed by western
blot analysis, by preventing ASC
oligomerization in a dose-dependent manner,
in mouse bone marrow-derived
macrophages challenged with several
proinflammatory stimuli that triggered NLRP3
inflammasome.

MCC950 NLRP3 NASH Mice Administration of MCC950 in two mouse Good [50]
inflammasome models of diet-induced NASH significantly
reduced NLRP3 components and IL-1b
mRNA expression and IL-1b, IL-6, and MCP-
1 plasma concentrations, as well as NASH
score, liver inflammation, and liver fibrosis
assessed by histological analysis. No effect of
MCC950 was observed on liver steatosis.

Glyburide NLRP3 Type 2 Cells Glyburide administration on bone marrow- Good [51]
inflammasome diabetes derived macrophages challenged with
inflammasome triggers significantly reduced
pro-caspase-1 and pro- IL-1b NLRP3
dependent activation.

Parthenolide NLRP3 Inflammation Cells Bone marrow-derived macrophages were Good [52]
inflammasome challenged with LPS and pretreated with
parthenolide and bay 11-7082 before ATP
challenge for the two-step NLRP3 activation.
Administrating these two molecules
significantly reduced pro-caspase-1 and IL-
1b NLRP3-dependent activation, as
assessed by western blot analysis.

Bay 11-7082 NLRP3 Inflammation Cells Bone marrow-derived macrophages were Good [52]
inflammasome challenged with LPS and pretreated with
parthenolide and bay 11-7082 before ATP
challenge for the two-step NLRP3 activation.
Administrating these two molecules
significantly reduced pro-caspase-1 and IL-
1b NLRP3-dependent activation as assessed

466 Trends in Molecular Medicine, May 2018, Vol. 24, No. 5


Table 1. (continued)
Targeted Pharmacological Targeted Targeted Species Methods and results Conclusion Refs
pathway agent molecule diseases regarding
efficacy
by Western Blot analysis. GoodJuliana C
et al., as assessed by western blot analysis.

Emricasan Caspase family NAFLD/NASH Mice In a mouse model of diet-induced NASH, Mild [53]
mice treated with Emricasan had significant
reduction of scores for liver apoptosis,
inflammation, and fibrosis, assessed by
histopathological analysis, when compared
with non-treated controls on the same diet.

Vx-166 Caspase family NAFLD/NASH Mice Administrating Vx-166 in two mouse models Mild [54,55]
of HFD-induced NAFLD and MCD (methyl
choline deficient)-induced NASH showed that
this compound can significantly reduce liver
apoptosis and inflammation as scored by
histopathological analysis when compared
with mice administered a placebo. However,
no effect was observed on the overall NASH
score and no effect was observed on hepatic
steatosis.

Nivocasan Mild Caspase family Rats Nivocasan prevented development of liver Good [56]
fibrosis as assessed by histopathological
analysis and reduced mRNA expression of
fibrosis markers aSMA, collagen 1a1, and
TGF-b in a rat model of thioacetamine-
induced liver fibrosis.

Ac-YVAD Caspase-1 NAFLD/NASH Mice Ac-YVAD administration significantly reduced Mild [57]
liver steatosis and fibrosis development, as
assessed by histological analysis, in LDLR–/–
mice fed a HFD when compared with
LDLR–/– mice on HFD that did not receive
treatment.

Neutrophil serine Alpha-1 NE, PR3, CG COPD, cystic Human Administration of AAT in patients with COPD Good [60]
proteases antitrypsin fibrosis or cystic fibrosis has proven efficient in
inhibiting neutrophil elastase activity in the
lungs and has shown an improvement in lung
function. AAT is used as treatment in some
cases of lung diseases due to AAT deficiency.

Dipetide vinyl DPPI Not tested Not Unknown Unknown [64]


sulfones tested

Semicarbazide- DPPI Not tested Not Unknown Unknown [65]


derived inhibitors tested

E-64c- DPPI Not tested Not Unknown Unknown [66]


hydrazide- tested
derived inhibitors

Regarding the use of AAT in NAFLD, there are a few results indicating its importance as
potential treatment. As described above, correcting an imbalance between protein expression
of AAT and NSPs might be a good therapeutic approach [11]. We have also investigated AAT
as a potential therapy for NAFLD. In our study, WT mice were treated with human AAT during
the last 10 days of a 16-weeks HFD intervention. Treatment reduced hepatic lipid content and
fasting glucose concentrations when compared with nontreatment [9]. Similar observations

Trends in Molecular Medicine, May 2018, Vol. 24, No. 5 467


were made in patient studies. High protein expression of NE was negatively correlated with the
protein expression of AAT in patients with liver steatosis and NASH [10]. In conclusion, a deficit
of AAT is responsible for insufficient inhibition of NSPs and therefore also for inflammation-
induced liver diseases. Although we do not yet know the efficacy of AAT and the possible
adverse event in NAFLD patients, it is a promising candidate because it is a natural occurring
inhibitor of both inflammasome-dependent and independent cytokine activation pathways [59].
In patients, AAT has already been shown to be a successful therapy in NSP-mediated
inflammatory diseases, such as chronic obstructive pulmonary disease (COPD) and cystic
fibrosis [60]. Besides inhibiting NSPs and caspase-1, AAT is also a potent inducer of IL-1Ra, the
natural antagonist of IL-1b [61]: another reason to consider this protein as a good candidate for
treating IL-1-mediated diseases.

Another interesting approach for inhibiting NSP pathways might be targeting of their activator
dipeptidyl peptidase I (DPPI). Studies investigating arthritis and NSP-mediated autoimmune
diseases have shown that DPPI k.o. mice do not develop full-blown inflammation when compared
with WT mice. DPPI k.o. mice had significantly decreased enzyme activity for NE, PR3, and CG and
decreased protein expression of proinflammatory cytokines such as TNF-a and IL-1b when
compared with WT controls [36,62,63]. These results strengthen the hypothesis that targeting
DPPI might also be an interesting therapeutic approach for NSP-mediated metabolic diseases. To
date, only a few DPPI inhibitors have been described but have not yet been tested in an in vivo
model for NSP-mediated inflammatory diseases (Table 1) [64–66].

Although future research is needed to provide insights into the balance between the beneficial
and harmful effects of inhibiting these pathways, we believe that these initial studies show
encouraging results regarding the inhibition of both inflammasome-dependent and indepen-
dent pathways in treating inflammatory-driven liver diseases.

Concluding Remarks
It is known that IL-1 family cytokines play a major role in the development of liver inflammation. Since
these cytokines need enzymatic processing to become active, their activators are also important in
the process of liver inflammation. In this opinion article we provide new insights, showing that next to
the classical NLRP3 inflammasome–caspase-1 cytokine activation pathway, NSPs are also potent
cytokine activators that contribute to liver disease progression. This might explain why pharmaco-
logical agents against the NLRP3–caspase-1 pathway have a rather mild effect on cytokine
inhibition and liver inflammatory events. The realization that NSPs are also involved in cytokine
activation opens new perspectives for NAFLD treatment. Additional studies are needed to further
investigate whether the NSP pathway can be targeted. Next to that, we believe it is worthwhile to
investigate suppression of both NLRP3–caspase-1 and NSP pathways to systematically inhibit
cytokine activation, thereby reducing inflammation. Comprehensive studies are needed to investi-
gate not only efficacy but also safety aspects of blocking multiple cytokines in inflammatory-induced
metabolic conditions (see Outstanding Questions). A k.o. mouse for both caspase-1 and NSPs
would provide a valuable tool for future studies investigating simultaneous suppression of these
pathways. Additionally, experiments in transgenic mice overexpressing NSPs could provide useful
information regarding the systemic consequences of overexpressing these enzymes. Next to
animal models, investigating genetic variants in candidate genes using well-characterized NAFLD
patient cohorts would provide a better understanding of NSP regulation in humans and may
highlight new targets that have therapeutic potential (see Clinician’s Corner, Box 2).

A limitation in reviewing current literature is that most studies investigated animal models of
NAFLD. At this point it is unclear how much of the results obtained in these animal studies could

468 Trends in Molecular Medicine, May 2018, Vol. 24, No. 5


be translated to the human situation. In addition, human studies are hampered by the fact that it Outstanding Questions
is difficult to obtain relevant samples, as a liver biopsy is an invasive method that has potential What are the interactions between
complications. Therefore, it is challenging to investigate inflammatory mechanisms in human NSPs and NLRP3 inflammasome–
caspase-1 pathways? Treating NAFLD
patients with NAFLD and most data in human studies is obtained by measuring markers in is difficult and downregulating one
plasma/serum. However, it is often unclear whether these plasma/serum markers reflect the pathway might directly or indirectly
local inflammatory status in the liver. upregulate the other via interactions
not yet revealed.

Despite these limitations, we believe new insights now reveal that NSPs, next to the NLRP3–
What are the consequences of the
caspase-1 pathway, also contribute to the development of NAFLD and NASH. The realization systemic inhibition of multiple proin-
that NSPs contribute to the disease process in an inflammasome-independent manner should flammatory cytokines? These proteins
move the field forward and opens up new perspectives for therapeutic interventions for the are important for the normal immune
response to infections.
treatment of NAFLD and NASH.

Box 2. Clinician’s Corner Which (experimental) drugs are able to


inhibit both caspase-1 and NSPs? Is
 Nonalcoholic fatty liver disease (NAFLD) can range from simple steatosis to liver inflammation [nonalcoholic
AAT a good candidate to target both
steatohepatitis (NASH)] and fibrosis, which affect the survival rate of patients. Currently, there is no treatment available
pathways? What are both the short-
to stop disease progression.
and long-term effects of these (experi-
 Proinflammatory cytokines play an important role in the disease progression from liver steatosis to liver inflammation
mental) drug regarding efficacy and
and fibrosis. IL-1 family cytokines are one of the major promoters of liver inflammatory events. Blocking the action of
adverse events?
these cytokines could potentially stop NAFLD progression. Cytokines need enzymatic processing in order to become
active. Therefore, blocking the pathways responsible for cytokine processing/activation might also have therapeutic
potential. During which disease stages would
 The NLRP3 inflammasome complex and its effector protein caspase-1 is one of the most studied pathways treatment have beneficial effects?
for activation of IL-1 family cytokines. Many studies have reported that this pathway is involved in the Should these pathways be targeted
development of NAFLD and other metabolic disturbances. However, this pathway is not the only mechanism during the early stages of disease or
available for cytokine activation. Neutrophil serine proteases also play an important role in cytokine activation would it also have effects during later
during NAFLD. stages? Can it be administered as pre-
 Targeting both the NLRP3–caspase-1 and neutrophil serine protease pathways might have potential as a new ventive therapy for patients at risk to
therapy for the treatment of inflammatory-induced NAFLD. develop NAFLD?

Which biomarkers are suitable for


monitoring disease progression and/
or therapy response in inflammatory-
Disclaimer Statement
induced NAFLD?
The authors declare they have no competing interests.

Acknowledgements
This work was supported by a grant of the Else-Kröner-Fresenius-Stiftung (to E.T., L.J., and T.C.). T.C. was also supported
by the European research Council (DEMETINL).

References
1. European Association for the Study of the Liver et al. (2016) 8. Armstrong, L. et al. (2009) Tumour necrosis factor-alpha proc-
EASL-EASD-EASO clinical practice guidelines for the manage- essing in interstitial lung disease: a potential role for exogenous
ment of non-alcoholic fatty liver disease. J. Hepatol. 64, 1388– proteinase-3. Clin. Exp. Immunol. 156, 336–343
1402 9. Toonen, E.J. et al. (2016) Activation of proteinase 3 contributes to
2. Calzadilla Bertot, L. and Adams, L.A. (2016) The natural course of non-alcoholic fatty liver disease (NAFLD) and insulin resistance.
non-alcoholic fatty liver disease. Int. J. Mol. Sci. 17, E774 Mol. Med. 22, 202–214
3. Kakino, S. et al. (2017) Pivotal role of TNF-alpha in the develop- 10. Zang, S. et al. (2016) Increased ratio of neutrophil elastase to
ment and progression of nonalcoholic fatty liver disease in a alpha1-antitrypsin is closely associated with liver inflammation in
murine model. Horm. Metab. Res. 50, 80–87 patients with nonalcoholic steatohepatitis. Clin. Exp. Pharmacol.
4. Negrin, K.A. et al. (2014) IL-1 signaling in obesity-induced hepatic Physiol. 43, 13–21
lipogenesis and steatosis. PLoS One 9, e107265 11. Mansuy-Aubert, V. et al. (2013) Imbalance between neutrophil
5. Szabo, G. and Petrasek, J. (2015) Inflammasome activation and elastase and its inhibitor alpha1-antitrypsin in obesity alters insulin
function in liver disease. Nat. Rev. Gastroenterol. Hepatol. 12, sensitivity, inflammation, and energy expenditure. Cell Metab. 17,
387–400 534–548

6. Mehal, W.Z. (2014) The inflammasome in liver injury and non- 12. Dinarello, C.A. (2009) Immunological and inflammatory
alcoholic fatty liver disease. Dig. Dis. 32, 507–515 functions of the interleukin-1 family. Annu. Rev. Immunol. 27,
519–550
7. Afonina, I.S. et al. (2015) Proteolytic processing of interleukin-1
family cytokines: variations on a common theme. Immunity 42, 13. Tan, Q. et al. (2016) The role of IL-1 family members and Kupffer
991–1004 cells in liver regeneration. BioMed Res. Int. 2016, 6495793

Trends in Molecular Medicine, May 2018, Vol. 24, No. 5 469


14. Patrick, A.L. et al. (2007) Hepatic leukocyte recruitment in 39. Engstrom, G. et al. (2003) Inflammation-sensitive plasma pro-
response to time-limited expression of TNF-alpha and IL-1beta. teins, diabetes, and mortality and incidence of myocardial
Am. J. Physiol. Gastrointest. Liver Physiol. 293, G663–G672 infarction and stroke: a population-based study. Diabetes 52,
15. Garlanda, C. et al. (2013) The interleukin-1 family: back to the 442–447
future. Immunity 39, 1003–1018 40. Engstrom, G. et al. (2003) Inflammation-sensitive plasma proteins
16. Kubes, P. and Mehal, W.Z. (2012) Sterile inflammation in the liver. are associated with future weight gain. Diabetes 52, 2097–2101
Gastroenterology 143, 1158–1172 41. Honda, M. and Kubes, P. (2018) Neutrophils and neutrophil
17. Tilg, H. (2010) The role of cytokines in non-alcoholic fatty liver extracellular traps in the liver and gastrointestinal system. Nat.
disease. Dig. Dis. 28, 179–185 Rev. Gastroenterol. Hepatol. Published online January 31, 2018.
http://dx.doi.org/10.1038/nrgastro.2017.183
18. Gieling, R.G. et al. (2009) Interleukin-1 participates in the pro-
gression from liver injury to fibrosis. Am. J. Physiol. Gastrointest. 42. Omoto, Y. et al. (2010) Granzyme B is a novel interleukin-18
Liver Physiol. 296, G1324–G1331 converting enzyme. J. Dermatol. Sci. 59, 129–135

19. Miura, K. et al. (2010) Toll-like receptor 9 promotes steatohepa- 43. Ridker, P.M. et al. (2017) Antiinflammatory therapy with canaki-
titis by induction of interleukin-1beta in mice. Gastroenterology numab for atherosclerotic disease. N. Engl. J. Med. 377, 1119–
139, 323–334 1131

20. Kamari, Y. et al. (2011) Lack of interleukin-1alpha or interleukin- 44. van Asseldonk, E.J. et al. (2015) One week treatment with the IL-1
1beta inhibits transformation of steatosis to steatohepatitis and receptor antagonist anakinra leads to a sustained improvement in
liver fibrosis in hypercholesterolemic mice. J. Hepatol. 55, 1086– insulin sensitivity in insulin resistant patients with type 1 diabetes
1094 mellitus. Clin. Immunol. 160, 155–162

21. Netea, M.G. et al. (2006) Deficiency of interleukin-18 in mice leads 45. Larsen, C.M. et al. (2007) Interleukin-1-receptor antagonist in
to hyperphagia, obesity and insulin resistance. Nat. Med. 12, type 2 diabetes mellitus. N. Engl. J. Med. 356, 1517–1526
650–656 46. Yang, G. et al. (2016) Erratum: A pharmacological inhibitor of
22. Yamanishi, K. et al. (2016) Interleukin-18-deficient mice develop NLRP3 inflammasome prevents non-alcoholic fatty liver disease
dyslipidemia resulting in nonalcoholic fatty liver disease and stea- in a mouse model induced by high fat diet. Sci. Rep. 6, 26218
tohepatitis. Transl. Res. 173, 101–114 47. Lee, H.J. et al. (2015) Peroxisome proliferator-activated receptor-
23. Hung, J. et al. (2005) Elevated interleukin-18 levels are associated delta agonist ameliorated inflammasome activation in nonalco-
with the metabolic syndrome independent of obesity and insulin holic fatty liver disease. World J. Gastroenterol. 21, 12787–12799
resistance. Arterioscler. Thromb. Vasc. Biol. 25, 1268–1273 48. Honda, H. et al. (2014) Isoliquiritigenin is a potent inhibitor of
24. Troseid, M. et al. (2010) The role of interleukin-18 in the metabolic NLRP3 inflammasome activation and diet-induced adipose tissue
syndrome. Cardiovasc. Diabetol. 9, 11 inflammation. J. Leukoc. Biol. 96, 1087–1100

25. Troseid, M. et al. (2010) Arterial stiffness is independently asso- 49. Youm, Y.H. et al. (2015) The ketone metabolite beta-hydroxybu-
ciated with interleukin-18 and components of the metabolic syn- tyrate blocks NLRP3 inflammasome-mediated inflammatory dis-
drome. Atherosclerosis 209, 337–339 ease. Nat. Med. 21, 263–269

26. Gao, Y. et al. (2016) IL-33 treatment attenuated diet-induced 50. Mridha, A.R. et al. (2017) NLRP3 inflammasome blockade
hepatic steatosis but aggravated hepatic fibrosis. Oncotarget reduces liver inflammation and fibrosis in experimental NASH in
7, 33649–33661 mice. J. Hepatol. 66, 1037–1046

27. Marvie, P. et al. (2010) Interleukin-33 overexpression is associ- 51. Lamkanfi, M. et al. (2009) Glyburide inhibits the cryopyrin/Nalp3
ated with liver fibrosis in mice and humans. J. Cell. Mol. Med. 14, inflammasome. J. Cell Biol. 187, 61–70
1726–1739 52. Juliana, C. et al. (2010) Anti-inflammatory compounds partheno-
28. Weiskirchen, R. and Tacke, F. (2017) Interleukin-33 in the path- lide and Bay 11-7082 are direct inhibitors of the inflammasome. J.
ogenesis of liver fibrosis: alarming ILC2 and hepatic stellate cells. Biol. Chem. 285, 9792–9802
Cell. Mol. Immunol. 14, 143–145 53. Barreyro, F.J. et al. (2015) The pan-caspase inhibitor emricasan
29. Martinon, F. et al. (2002) The inflammasome: a molecular platform (IDN-6556) decreases liver injury and fibrosis in a murine model of
triggering activation of inflammatory caspases and processing of non-alcoholic steatohepatitis. Liver Int. 35, 953–966
proIL-beta. Mol. Cell 10, 417–426 54. Witek, R.P. et al. (2009) Pan-caspase inhibitor VX-166 reduces
30. Wree, A. et al. (2014) NLRP3 inflammasome activation results in fibrosis in an animal model of nonalcoholic steatohepatitis. Hep-
hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. atology 50, 1421–1430
Hepatology 59, 898–910 55. Anstee, Q.M. et al. (2010) Impact of pan-caspase inhibition in
31. Wree, A. et al. (2014) NLRP3 inflammasome activation is required animal models of established steatosis and non-alcoholic stea-
for fibrosis development in NAFLD. J. Mol. Med. 92, 1069–1082 tohepatitis. J. Hepatol. 53, 542–550
32. Csak, T. et al. (2011) Fatty acid and endotoxin activate inflam- 56. Kim, D.Y. et al. (2013) Combined effects of an antioxidant and
masomes in mouse hepatocytes that release danger signals to caspase inhibitor on the reversal of hepatic fibrosis in rats. Apo-
stimulate immune cells. Hepatology 54, 133–144 ptosis 18, 1481–1491

33. Dixon, L.J. et al. (2013) Caspase-1 as a central regulator of high 57. Morrison, M.C. et al. (2016) Intervention with a caspase-1 inhibitor
fat diet-induced non-alcoholic steatohepatitis. PLoS One 8, reduces obesity-associated hyperinsulinemia, non-alcoholic
e56100 steatohepatitis and hepatic fibrosis in LDLR–/–. Leiden mice.
Int. J. Obes. 40, 1416–1423
34. Dixon, L.J. et al. (2012) Caspase-1-mediated regulation of fibro-
genesis in diet-induced steatohepatitis. Lab. Invest. 92, 713–723 58. Toldo, S. et al. (2011) Alpha-1 antitrypsin inhibits caspase-1 and
protects from acute myocardial ischemia-reperfusion injury. J.
35. Netea, M.G. et al. (2015) Inflammasome-independent regulation
Mol. Cell Cardiol. 51, 244–251
of IL-1-family cytokines. Annu. Rev. Immunol. 33, 49–77
59. Grimstein, C. et al. (2011) Alpha-1 antitrypsin protein and gene
36. Joosten, L.A. et al. (2009) Inflammatory arthritis in caspase 1
therapies decrease autoimmunity and delay arthritis development
gene-deficient mice: contribution of proteinase 3 to caspase 1-
in mouse model. J. Transl. Med. 9, 21
independent production of bioactive interleukin-1beta. Arthritis
Rheum. 60, 3651–3662 60. Siekmeier, R. (2010) Lung deposition of inhaled alpha-1-protein-
ase inhibitor (alpha 1-PI) - problems and experience of alpha1-PI
37. Bae, S. et al. (2012) Neutrophil proteinase 3 induces diabetes in a
inhalation therapy in patients with hereditary alpha1-PI deficiency
mouse model of glucose tolerance. Endocr. Res. 37, 35–45
and cystic fibrosis. Eur. J. Med. Res. 15 (Suppl 2), 164–174
38. Xu, X. et al. (2015) Obesity is associated with more activated
61. Joosten, L.A. et al. (2016) Alpha-1-anti-trypsin-Fc fusion protein
neutrophils in African American male youth. Int. J. Obes. 39, 26–
ameliorates gouty arthritis by reducing release and extracellular
32

470 Trends in Molecular Medicine, May 2018, Vol. 24, No. 5


processing of IL-1beta and by the induction of endogenous IL- 64. Korver, G.E. et al. (2001) Dipeptide vinyl sulfones suitable for
1Ra. Ann. Rheum. Dis. 75, 1219–1227 intracellular inhibition of dipeptidyl peptidase I. Int. Immunophar-
62. Adkison, A.M. et al. (2002) Dipeptidyl peptidase I activates macol. 1, 21–32
neutrophil-derived serine proteases and regulates the develop- 65. Bondebjerg, J. et al. (2005) Novel semicarbazide-derived inhib-
ment of acute experimental arthritis. J. Clin. Invest. 109, 363– itors of human dipeptidyl peptidase I (hDPPI). Bioorg. Med.
371 Chem. 13, 4408–4424
63. Schreiber, A. et al. (2012) Neutrophil serine proteases promote IL- 66. Radzey, H. et al. (2013) E-64c-hydrazide: a lead structure for the
1beta generation and injury in necrotizing crescentic glomerulo- development of irreversible cathepsin C inhibitors. ChemMed-
nephritis. J. Am. Soc. Nephrol. 23, 470–482 Chem 8, 1314–1321

Trends in Molecular Medicine, May 2018, Vol. 24, No. 5 471

Você também pode gostar