You are on page 1of 9

Cell Metabolism

Review

The Inflammasome Puts Obesity in the Danger Zone


Rinke Stienstra,1,2,3 Cees J. Tack,1 Thirumala-Devi Kanneganti,4 Leo A.B. Joosten,1,2 and Mihai G. Netea1,2,*
1Department of Medicine
2Nijmegen Institute for Infection, Inflammation and Immunity (N4I)
Radboud University Nijmegen Medical Centre, Nijmegen 6525 GA, The Netherlands
3Nutrition, Metabolism and Genomics Group, Wageningen University, Wageningen 6703 HD, The Netherlands
4Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA

*Correspondence: m.netea@aig.umcn.nl
DOI 10.1016/j.cmet.2011.10.011

Obesity-induced inflammation is an important contributor to the induction of insulin resistance. Recently, the
cytokine interleukin-1b (IL-1b) has emerged as a prominent instigator of the proinflammatory response in
obesity. Several studies over the last year have subsequently deciphered the molecular mechanisms respon-
sible for IL-1b activation in adipose tissue, liver, and macrophages and demonstrated a central role of the
processing enzyme caspase-1 and of the protein complex leading to its activation called the inflammasome.
These data suggest that activation of the inflammasome represents a crucial step in the road from obesity to
insulin resistance and type 2 diabetes.

Introduction promoting activation of inflammasome-dependent caspase-1,


Accumulating evidence links inflammation to metabolic changes and highlight new findings regarding the mechanisms involved
associated with obesity and type 2 diabetes (Donath and Shoe- in the processing of IL-1b during the progression of obesity
lson, 2011). While several studies suggest that expanding and insulin resistance. In light of the growing interest to block
adipose tissue is an important first step in driving the enhanced low-grade inflammation in obese and insulin-resistant subjects,
state of inflammation, the underlying molecular mechanisms this review will particularly focus on the potential of the inflamma-
modulating this process are less clear. A wide variety of immune some as a therapeutic target in the treatment of obesity-induced
cells, including macrophages, monocytes, T cells, and b cells, insulin resistance.
have been shown to infiltrate the adipose tissue and affect its
homeostasis by releasing inflammatory cytokines (Anderson Pathogenic Role of IL-1b in the Development of Obesity,
et al., 2010). Adipocytes themselves are also capable of Insulin Resistance, and Diabetes
releasing inflammatory mediators and contribute to the inflam- IL-1b elicits potent proinflammatory actions through its binding
matory response (McGillicuddy et al., 2011; Stienstra et al., to the IL-1 receptor that in turn recruits the IL-1 receptor acces-
2010; Meijer et al., 2011). In addition to adipose tissue, inflamma- sory protein. This receptor complex signals through the myeloid
tion in liver and pancreatic islets is also evident in obese individ- differentiation factor 88 (MyD88) adaptor protein that spurs on
uals and participates in the pathogenesis of type 2 diabetes (Gre- IL-1 receptor-activated kinases (IRAK1 to 4). This leads to activa-
gor and Hotamisligil, 2011). One of the proinflammatory tion of several protein kinases, including mitogen-activated
cytokines mediating obesity-induced inflammation is interleukin protein kinase 8 (JNK), mitogen-activated protein kinase 1
(IL)-1b, which is processed by caspase-1, a cysteine protease (ERK), p38 MAPK, and inhibitor of kappaB kinase (IKK), that
regulated by a protein complex called the inflammasome. initiate the transcription factors nuclear factor kB (NF-kB) and
Although growing evidence points to a crucial role for IL-1b in activator protein 1 (AP1) to stimulate inflammatory gene expres-
mediating the development of insulin resistance, it should be sion (Dinarello, 2011) (see Figure 1). The proinflammatory actions
stressed that the inflammatory response driving the develop- of IL-1b are recognized as an important contributor to the devel-
ment of insulin resistance probably is comprised of a combina- opment of both type 1 and type 2 diabetes (Donath and Shoe-
tion of proinflammatory cytokines that jointly effectuate type 2 lson, 2011; Mandrup-Poulsen et al., 2010). While IL-1b has toxic
diabetes progression. For example, involvement of TNFa in effects on pancreatic b cells in the process of autoimmune dia-
obesity-associated insulin resistance has been frequently re- betes (Mandrup-Poulsen et al., 1986), the cytokine also appears
ported. Since biological processes are often multifactorial, to be involved in b cell deterioration related to glucotoxicity in
involvement of other cytokines like TNFa is plausible. type 2 diabetes (Donath et al., 2003), with both processes
Although detrimental effects of IL-1b on b cell function have leading to defective insulin production. By activation of Fas-trig-
been well documented, the proinflammatory effects of IL-1b that gered apoptosis, which involves the transcription factor NF-kB,
mediate the development of tissue dysfunction and peripheral IL-1b mediates b cell dysfunction. More peripherally, IL-1b
insulin resistance have only recently received more interest. While directly inhibits insulin signaling pathways by reducing tyrosine
several lines of evidence have shown involvement of IL-1b in the phosphorylation of insulin receptor substrate 1 (IRS1) and nega-
development of obesity-associated insulin resistance peripherally, tive regulation of IRS1 gene expression levels, thus inducing
the quantitative contribution remains to be defined in more detail. a state of insulin resistance (Jager et al., 2007). In animal studies,
In the present review we will discuss recently identified meta- the lack of IL-1b or its receptor protects against the development
bolic triggers that may function as potential danger signals, of adipose tissue inflammation and insulin resistance upon

10 Cell Metabolism 15, January 4, 2012 ª2012 Elsevier Inc.


Cell Metabolism

Review

Figure 1. IL-1b Signaling Pathway: Overview of the Intracellular Signaling Pathways Activated by Binding of IL-1b to the IL-1 Receptor
Upon activation of the IL-1 receptor complex, IL-1b induces recruitment of the myeloid differentiation primary response gene 88 (MyD88), which in turn promotes
activation of the interleukin-1 receptor kinase (IRAK) cascade. Via tumor necrosis factor-associated factor 6 (TRAF6) and the c-Jun N terminus (JNK), p38
mitogen-activated protein (p38 MAPK), and the inhibitor of nuclear factor b (IKK) kinases, the IkB cofactor is degraded, which subsequently promotes the nuclear
translocation of NF-kB and AP-1. Both transcription factors have the capacity to induce proinflammatory gene expression of various cytokines and chemokines
that modulate the inflammatory response.

high-fat diet feeding (Wen et al., 2011; McGillicuddy et al., 2011). sensitivity in patients diagnosed with rheumatoid arthritis (Gon-
This protective effect may partially be mediated by the absence zalez-Gay et al., 2010; Huvers et al., 2007).
of IL-1b-controlled chemokine synthesis (Dinarello, 2011), which Until recently, the triggers and molecular switches controlling
governs the influx of various immune cells into adipose tissue, IL-1b production during the development of obesity and insulin
thus promoting inflammation. resistance have remained largely unknown. In as much as
In line with the proposed pathogenic role of IL-1b, increased IL-1b elicits a vigorous inflammatory response, activation must
circulating levels of this cytokine accompanied by elevated be tightly controlled and requires processing from an inactive
levels of IL-6 positively correlate with the development of type procytokine into the biologically active form by proteolytic
2 diabetes in humans (Spranger et al., 2003). Conversely, enzymes. Processing of cytokines of the IL-1 family, such as
blockade of excessive IL-1 signaling in subjects with type 2 dia- IL-1b and IL-18, is mainly mediated by the cysteine protease
betes improves glycemic control and b cell function while caspase-1, which in turn is activated by a protein platform
reducing markers of systemic inflammation (Larsen et al., termed the inflammasome.
2007). Although clinical studies have revealed that inhibition of
IL-1 signaling improves glucose tolerance, data pointing to an The Inflammasome
improvement of insulin sensitivity upon anti-IL-1 treatment are The inflammasome is an important part of our innate immune
scarce. However, clinical studies using salsalate, an unspecific system that responds to danger signals that are sensed by
anti-inflammatory agent, have demonstrated a significant a number of different intracellular NOD-like receptors (NLRs).
improvement in insulin sensitivity (Goldfine et al., 2008). It should Different inflammasomes have been identified, including NLR
be noted that highly targeted anticytokine treatment ap- family pyrin domain-containing 1 (NLRP1), NLR family pyrin
proaches, such as treatment with the anti-TNF antibody (inflixi- domain-containing 3 (NLRP3), NLR family pyrin domain-con-
mab), have been found to be successful in improving insulin taining 6 (NLRP6), AIM2 (absent in melanoma 2), and IPAF

Cell Metabolism 15, January 4, 2012 ª2012 Elsevier Inc. 11


Cell Metabolism

Review

(IL-1b-converting enzyme protease-activating factor), which the deleterious effects of high-fat diet feeding. For example,
each have the ability to respond to a wide variety of microbial caspase-1 / mice have a decreased influx of macrophages
products or endogenous danger signals (Dunne, 2011). Hitherto, into adipose tissue upon high-fat diet feeding (Stienstra
NLRP3 is the most extensively studied inflammasome that, upon et al., 2011). Similarly, ASC / (Stienstra et al., 2011) and
its activation, forms a complex with its adaptor molecule PYD NLRP3 / animals (Vandanmagsar et al., 2011) were protected
and CARD domain containing protein (ASC) and thereby facili- against the development of hepatosteatosis instigated by high-
tates caspase-1-dependent processing of pro-IL-1b into its fat diet feeding. Importantly, HFD-fed mice lacking caspase-1
active form. Normally, this proinflammatory response provides were characterized by a robust improvement in insulin sensitivity
protection for the host by inducing an acute phase response and were rescued from the development of obesity as compared
(Dinarello, 2011). to wild-type mice (Stienstra et al., 2011). Notably, part of the
Recent studies have identified the inflammasome as an impor- protective effects of the absence of caspase-1 may be accom-
tant contributor to the development of insulin resistance by plished by the reduction in weight gain as compared to the
mediation of processing and release of IL-1b in various tissues wild-type animals fed the high-fat diet. A similar protection
and cell types during the development of obesity. In addition to against insulin resistance was observed in HFD-fed animals
IL-1b, caspase-1 is also able to process and activate IL-18 and lacking ASC or NLRP3 (Wen et al., 2011). However, different
IL-33 (Arend et al., 2008). In contrast to IL-1b, IL-18 ameliorates phenotypical responses were observed between the various in-
development of obesity and insulin resistance (Netea et al., flammasome knockout models upon high-fat diet feeding.
2006). Interestingly, caspase-1 deficient mice lacking both Whereas the absence of NLRP3 only provided protection against
IL-1b and IL-18 are characterized by an improvement in insulin high-fat diet-induced insulin resistance and fatty liver disease
resistance (Stienstra et al., 2010), suggesting that the insulin- after prolonged exposure to the diet for up to 9 months in one
desensitizing effects of IL-1b override IL-18 action. The fact model (Vandanmagsar et al., 2011), shorter periods of high-fat
that expression and circulating levels of IL-18 are easily detect- diet feeding also appear to activate NLRP3 (Wen et al., 2011).
able in healthy subjects, as compared to IL-1b, suggests that this The absence of caspase-1 and ASC conferred protection
cytokine executes different functions, among which are the against the development of insulin resistance after 12 or
opposing effects on insulin resistance. 16 weeks of diet intervention. These results imply that NLRP3
activation may require secondary danger signals that are only
NLRP3-Mediated Caspase-1 Activity in the Driver’s Seat apparent after prolonged high-fat diet feeding and may include
Several lines of evidence suggest that activation of inflamma- lipotoxic metabolites like sphingolipids. Hypothetically, differ-
some-mediated caspase-1 is one of the culprits behind the ences in the composition of the diets that were used to induce
enhanced inflammatory state characteristic of obesity and has obesity may have altered the supply of NLRP3 danger signals.
center stage in the pathogenesis of type 2 diabetes by acting Alternatively, other inflammasomes next to NLRP3 may also be
at two different levels. involved in the development of obesity-induced insulin resis-
First, caspase-1 appears to instigate defective insulin secre- tance. Furthermore, differences in expression levels between
tion by promoting pancreatic dysfunction. Pancreatic b cells it- distinct adipose tissue cell populations may explain any pheno-
self are capable of producing IL-1b (Böni-Schnetzler et al., typical variance. While caspase-1 is highly expressed in both
2008; Maedler et al., 2002) through mechanisms involving the adipocytes and nonadipoycte cells, the inflammasome
NLRP3 inflammasome (Zhou et al., 2010). Additionally, members NLRP3 and ASC are predominantly expressed in non-
enhanced macrophages’ infiltration of the pancreas observed adipocyte cells that are part of the adipose tissue. Although the
in human type 2 diabetic patients and high-fat diet (HFD)-fed lower expression levels do not rule out any function of ASC and
mice (Ehses et al., 2007) may further potentiate IL-1b production. NLRP3 in adipocytes, it appears that crosstalk between both cell
The IL-1b-driven inflammation of the islets is proposed as the types determines the secretion levels of IL-1b by adipose tissue.
central mediator of glucose-, lipid-, and amyloid-induced b cell However, it should be emphasized that isolated adipocytes do
failure leading to defective insulin secretion (Masters et al., have the potential to produce IL-1b (Koenen et al., 2011b).
2011; Mandrup-Poulsen, 2010) and ultimately b cell death, two Hence, caspase-1 activation in adipocytes may rely on non-
processes distinctive for the pathogenesis of type 2 diabetes. NLRP3 inflammasome members or is controlled independently
Second, activation of caspase-1 can alter the function of of the inflammasome.
peripheral tissues, including adipose tissue and liver, both criti- Additionally, important tissue-specific effects of various in-
cally involved in maintaining glucose homeostasis. Adipose flammasomes have recently been described, such as the regula-
tissue of animals fed a high-fat diet to induce obesity and insulin tion of IL-18 production in intestinal epithelial cells by the NLRP6
resistance is characterized by enhanced gene expression and inflammasome (Elinav et al., 2011). This opens up the intriguing
increased protein levels of caspase-1 (Stienstra et al., 2010; Van- and yet unexplored possibility of specific inflammasome compo-
danmagsar et al., 2011). Similarly, feeding mice a methionine- nents present in adipocytes, hepatocytes, macrophages, or
choline-deficient diet, which promotes the development of pancreatic b cells that may differentially affect the development
nonalcoholic steatohepatitis (NASH), has been shown to of insulin resistance. Alternatively, differential regulation of
promote NLRP3-dependent caspase-1 activation within hepato- similar inflammasome subtypes may also occur and mediate
cytes (Csak et al., 2011). The elevated activity of caspase-1 leads tissue-specific effects.
to increased processing of IL-1b and promotes a proinflamma- There is supportive evidence for the involvement of the in-
tory environment that drives tissue dysfunction. Indeed, the flammasome in obesity-induced inflammation in humans. For
absence of caspase-1 provides protection for the host against example, caspase-1 activity levels are more pronounced in the

12 Cell Metabolism 15, January 4, 2012 ª2012 Elsevier Inc.


Cell Metabolism

Review

visceral fat depot, which is a strong determinant of insulin resis- example, palmitate acts as a ligand for TLR4 and induces IL-6
tance, as compared to subcutaneous adipose tissue of mildly mRNA expression through activation of the transcription factor
obese subjects (Koenen et al., 2011b). In liver, expression levels NF-kB (Shi et al., 2006). Moreover, treatment of human THP-1
of NLRP3, caspase-1, and ASC are elevated in patients suffering cells with palmitate has been shown to induce IL-1b mRNA
from NASH compared to healthy controls (Csak et al., 2011). expression (Håversen et al., 2009) although some doubt has
Conversely, substantial weight loss in obese subjects with type been cast on whether saturated fatty acids can induce TLR
2 diabetes was shown to reduce expression levels of IL-1b and signaling (Erridge and Samani, 2009). However, elevated levels
NLRP3 in adipose tissue and positively correlated with changes of palmitate may provide the first signal needed for IL-1b produc-
in fasting plasma glucose levels (Vandanmagsar et al., 2011). tion though induction of IL-1b gene transcription, although it
Overall, these results argue that activation of IL-1b by the in- has to be kept in mind that levels of all fatty acids are elevated
flammasome plays a central role in the pathogenesis of in type 2 diabetes and may alter the effect of palmitate on
obesity-induced insulin resistance. Since activation of cas- IL-1b production.
pase-1 leading to the cleavage and secretion of IL-1b is under In addition, recent work demonstrates that palmitate also has
tight control of the inflammasome, specific danger signals the potential to directly activate the NLRP3-inflammasome in
should exist that arise during the development of obesity and macrophages and thus provide the second signal needed for
that are sensed by intracellular NLRs. Indeed, several studies IL-1b release (Wen et al., 2011). Via detailed mechanistic
have identified metabolic danger signals that can efficiently acti- in vitro studies, it was shown that palmitate negatively affects
vate the inflammasome, leading to caspase-1-driven cleavage of the activation of AMP-activated protein kinase (AMPK), an
IL-1b in a variety of cell types, both from nonmyeloid and myeloid energy-sensing kinase that regulates multiple biochemical path-
origin (Vandanmagsar et al., 2011; Wen et al., 2011; Csak et al., ways in all eukaryotes, including lipid and glucose metabolism
2011; Zhou et al., 2010; Masters et al., 2010). and apoptosis (Steinberg and Kemp, 2009). The reduction in
AMPK phosphorylation leads to defective autophagy, a process
Potential Metabolic Danger Signals that Activate IL-1b known to regulate the clearance of dysfunctional mitochondria.
Recent studies have revealed that activation of IL-1b requires The authors propose that the subsequent intracellular accumula-
two signals (Gong et al., 2010; Joosten et al., 2010; Hornung tion of mitochondrial-derived reactive oxygen species (ROS)
et al., 2009). Whereas the first signal ‘‘primes’’ the cell and acts represents the trigger activating the inflammasome. Indeed,
as an inducer of IL-1b and NLR mRNA transcription, the second ROS have long been proposed to activate the inflammasome,
signal induces conformational changes in the inflammasome although some controversy exists. For example, cells isolated
that instigates caspase-1 activation. Classically, the first signal from patients diagnosed with chronic granulomatous disease
is provided by invading pathogens driving IL-1b mRNA transcrip- (CGD) have defective NADPH activity and thus cannot generate
tion through pattern recognition receptors such as toll-like NADPH-dependent ROS (van de Veerdonk et al., 2010; Meissner
receptors (TLRs) (Lamkanfi et al., 2008). The second signal is et al., 2010). Upon stimulation, monocytes from CGD patients
of a more heterogeneous nature, as it can be represented by produced similar or even increased amounts of IL-1b as
microbial components such as microbial DNA (Muruve et al., compared to cells from unaffected subjects, indicating
2008), cell-wall components, and toxins (Ali et al., 2011), but NADPH-independent activation of the inflammasome. In this
also by endogenous ligands such as adenosine triphosphate respect, enhanced production of ROS by mitochondria, a hall-
(ATP) or uric acid (Mariathasan et al., 2006). The precise unifying mark of insufficient mitochondrial function that is strongly asso-
mechanism through which all these ligands induce caspase-1 ciated with type 2 diabetes (Jin and Patti, 2009), may be an alter-
activation is not known, although a rapid K+ efflux from cells native source driving NLRP3 inflammasome activation
seems to be a common denominator that triggers activation of (Tschopp, 2011).
the inflammasome (Lamkanfi et al., 2008). Notably, not all cell Other pathways that mediate palmitate-induced inflamma-
types need sequential hits to induce IL-1b production. For some activation can also be envisaged. Exposure of cells to
example, monocytes only require the first signal to induce in- palmitate induces intracellular accumulation of ceramide, which
flammasome-dependent IL-1b release, due to the constitutive is produced by a ubiquitous biosynthetic pathway initiated by the
activation of caspase-1 in this cell type (Netea et al., 2009). condensation of palmitoyl-coenzyme A (CoA) and serine.
Moreover, IL-1b can also be processed by caspase-1-indepen- Prevention of de novo ceramide synthesis limited the develop-
dent cleavage through neutrophil-derived serine proteases ment of insulin resistance evoked by treatment of cells with satu-
(Joosten et al., 2009), yet no information is available concerning rated fatty acids (Chavez et al., 2003). Given the fact that ceram-
the mechanism that controls this process. ide has recently been identified as a potent activator of the
Very recently, fatty acids, glucose, uric acid, and islet amyloid inflammasome in macrophages (Vandanmagsar et al., 2011),
polypeptide (IAPP) have been put forward as metabolic danger palmitate may fuel ceramide biosynthesis, thereby providing
signals that possess the capacity to activate the inflammasome a continuous supply of danger signals for NLRP3 inflammasome
and stimulate IL-1b production (Figure 2). Below we will discuss activation.
where exactly these signals might act to promote IL-1b release. The palmitate-driven activation of the inflammasome is not
Fatty Acids limited to macrophages, as nonmyeloid cell types including
Elevated circulating levels of free fatty acids are one of the hall- hepatocytes have recently been shown to be a target of palmi-
marks of type 2 diabetes (Krebs and Roden, 2005). Interestingly, tate-driven activation of the NLRP3-inflammasome as well.
it has been suggested that saturated fatty acids bind and acti- Though palmitic acid has been nominated as the meta-
vate members of the TLR-family in vitro (Lee et al., 2001). For bolic activator that links obesity-induced insulin resistance to

Cell Metabolism 15, January 4, 2012 ª2012 Elsevier Inc. 13


Cell Metabolism

Review

Figure 2. Activation of the Inflammasome


An overview of the metabolic triggers that have been shown to provide the first signal (induction of transcription) or the second signal (inflammasome activation)
leading to the production and release of IL-1b. The first signal that is aimed at inducing IL-1b transcription and subsequent production of pro-IL-1b can be
provided by glucose, palmitate, uric acid, or LPS. In order to facilitate processing of pro-IL-1b into its active form, a second signal is essential to facilitate in-
flammasome activation and caspase-1-dependent cleavage of pro-IL-1b. NLRP3-dependent activation of caspase-1 can be triggered by palmitate, ceramide,
glucose, uric acid, reactive oxygen species (ROS), or amyloid. Promotion of this cascade occurs in a variety of tissues, including the pancreas, liver, and adipose
tissue and may subsequently contribute to tissue dysfunction and the development of insulin resistance.

activation of innate immunity, this may not imply that all fatty acids Glucose
cause activation of the inflammasome. Since unsaturated fatty It has been known for almost a decade that high glucose levels
acids have anti-inflammatory effects (Browning, 2003) and lack promote IL-1b mRNA transcription (Shanmugam et al., 2003).
the ability to activate TLR4 or evoke ceramide synthesis (Chavez Glucose has been shown to activate PKC-alpha and, via phos-
et al., 2003), one would predict that highly unsaturated species phorylation of p38 MAPK and ERK1/2, lead to NF-kB activation
prevent inflammasome activation. Accordingly, it is important to and subsequent IL-1b transcription in monocytes (Dasu et al.,
characterize the inflammasome-activating potential of various 2007), hence priming cells for inflammasome activation.
fatty acids differing in length and degree of unsaturation. Additional pathways may also participate in high glucose-
A potential confounder in most of the experiments pinpointing mediated activation of IL-1b release by providing the second
ceramide or palmitate as activators of the inflammasome (and signal that is required to activate the inflammasome. Indeed,
other metabolic danger signals) may be the fact that cells were glucose has been identified as a direct stimulator of caspase-1
primed with LPS (Wen et al., 2011; Vandanmagsar et al., activity (Vincent and Mohr, 2007). Recent evidence has sug-
2011). Notably, both palmitate and ceramide did not induce in- gested that the NLRP3 inflammasome is activated in response
flammasome activation and subsequent cleavage and secretion to high levels of glucose (Zhou et al., 2010). The mechanism of
of IL-1b without prior exposure of macrophages to LPS. Since it action involves thioredoxin-interacting protein (TXNIP), a protein
has been reported that serum endotoxin levels are elevated in that acts as an endogenous inhibitor of the ROS scavenging
obese animals due to a disruption in intestinal integrity (Cani protein thioredoxin (Minn et al., 2005; Parikh et al., 2007). Inter-
et al., 2008), LPS may serve as a first signal in obese individuals. estingly, TXNIP levels are elevated in subjects with type 2 dia-
However, other priming signals may exist in vivo that synergize betes mellitus (Parikh et al., 2007), and its expression is robustly
with ceramide or palmitate to induce both proIL-1b transcription induced by glucose (Stoltzman et al., 2008). Zhou et al. (2010)
and inflammasome activation, and these include other fatty have shown that, upon activation, TXNIP is able to directly
acids, glucose, or adipocytokines. interact with NLRP3 in a ROS-dependent manner, leading to

14 Cell Metabolism 15, January 4, 2012 ª2012 Elsevier Inc.


Cell Metabolism

Review

activation of caspase-1 and processing of IL-1b in pancreatic Although more work is needed, uric acid is an important candi-
islets. However, these observations could not be reproduced date that may modulate inflammasome activation during the
in bone-marrow-derived macrophages lacking TXNIP and development of obesity and insulin resistance.
exposed to high glucose levels (Masters et al., 2010). Neverthe- Islet Amyloid Polypeptide
less, treatment of the cells with 2-deoxy-D-glucose, which Islet amyloid polypeptide (IAPP), also known as amylin, is a regu-
blocks metabolic processing, prevents IL-1b processing and latory peptide of 37 amino acids that is produced by pancreatic
release, implying that adequate glucose metabolism is indis- b cells and inhibits insulin and glucagon secretion. The ability of
pensable for inflammasome function (Masters et al., 2010). amylin to form protein aggregates that cluster in pancreatic islets
It is reasonable to expect that the effects of high levels of as amyloid deposits is believed to be of critical importance for
glucose on caspase-1-mediated IL-1b production differ among the loss of b cell mass and type 2 diabetes progression (Wester-
various cell types. Indeed, although adipose tissue ex vivo mark et al., 2011). Recent evidence has uncovered that IAPP has
responds to high levels of glucose by inducing TXNIP and the ability to energize the NLRP3 inflammasome, subsequently
producing higher levels of IL-1b, TXNIP ablation in adipocytes promoting IL-1b production by the pancreas (Masters et al.,
did not affect caspase-1 activity levels (Koenen et al., 2011a). 2010). In contrast to glucose and palmitate, IAPP lacks the ability
However, knockdown of TXNIP did reduce IL-1b mRNA levels to drive IL-1b mRNA expression, yet robustly promotes NLRP3-
within adipocytes, indicating that TXNIP regulates IL-1b gene mediated caspase-1 activation, partly by a disturbance in the
expression levels during the presence of high levels of glucose. phagocytic machinery causing oxidative stress.
As TXNIP itself has the capacity to induce chromatin modifica- Interestingly, expression of IAPP by the pancreatic b cell line
tion (Perrone et al., 2009), it is conceivable that TXNIP may regu- MIN6 cells is activated by palmitate (Westermark et al., 2011),
late IL-1b promoter availability. suggesting that fatty acids may boost pancreatic IL-1b produc-
In summary, high glucose levels may provide the priming tion in type 2 diabetes.
signal for transcription of IL-1b by activation of TXNIP, which Additional lines of evidence suggest that IAPP may also have
subsequently facilitates enhanced IL-1b expression levels. Addi- extrapancreatic sites of action, since expression of the peptide
tionally, high concentrations of glucose also promote the gener- has been detected in the gastrointestinal tract and sensory
ation of ROS that is sufficient as second signal that promotes in- neurons (Westermark et al., 2011). Moreover, inasmuch as circu-
flammasome activation and subsequently induces release of lating levels of IAPP are elevated upon high-fat diet feeding of
bioactive IL-1b. animals (Westermark et al., 2011), it is plausible to expect that
Uric Acid IAAP-dependent activation of the inflammasome also occurs at
Hyperuricemia often occurs in obese individuals and frequently extrapancreatic sites.
precedes the development of type 2 diabetes. It has been thought Altogether, these data suggest that IAPP functions as a second
that high levels of uric acid are a result of hyperinsulinemia, since signal promoting NLRP3 activation and subsequent caspase-1-
insulin normally reduces the renal secretion of uric acid. Interest- dependent cleavage of IL-1b.
ingly, lowering of circulating levels of uric acid improved insulin
sensitivity in fructose-fed rats (Nakagawa et al., 2006). Therapeutic Interventions
It has been suggested that uric acid drives inflammatory and Given the wealth of data that have implicated activation of the in-
oxidative changes in adipocytes. Uric acid directly affects the flammasome in the pathogenesis of obesity-induced insulin
inflammatory status of the adipose tissue by regulating MCP-1 resistance, inhibition of IL-1b and caspase-1 may represent an
levels in adipocytes (Baldwin et al., 2011), thereby promoting attractive therapeutic target. So far, only a few studies have
infiltration of macrophages (Ito et al., 2008). Interestingly, crys- been conducted aimed at limiting IL-1b action, either by using
tals of monosodium urate, frequently observed in patients diag- receptor antagonists or by neutralizing IL-1b. Blockade of
nosed with gout, have been uncovered as a robust inducer of the IL-1R-mediated signaling by the recombinant IL-1 receptor
NLRP3 inflammasome in macrophages (Martinon et al., antagonist (IL-1Ra) Anakinra was effective in improving glycemic
2006).Can uric acid also drive obesity-induced inflammation by control in patients with type 2 diabetes (Larsen et al., 2007).
activating the inflammasome? However, no improvement in insulin sensitivity levels as deter-
Interestingly, uric acid is indispensable for normal adipogene- mined by euglycemic clamp was observed. In addition, no
sis, as animals that lack xanthine oxidoreductase (XOR), an changes in muscle gene expression or serum adipokine levels
enzyme that generates uric acid from xanthine, exhibit a were seen. Additionally, treatment with Anakinra had no direct
decrease in adipose content and are characterized by a reduc- beneficial effect on insulin sensitivity in nondiabetic insulin-resis-
tion in PPARg activity as compared to wild-type mice. More tant subjects (van Asseldonk et al., 2011). Although these obser-
importantly, uric acid levels within adipose tissue are enhanced vations suggest that IL-1 blockade in human subjects does not
in obese animals as compared to lean mice, and this is accom- reverse insulin resistance, only a limited number of studies
panied by elevated transcription levels of XOR (Cheung et al., have been conducted so far. Additionally, Anakinra has a short
2007). Thus, it may be hypothesized that uric acid can directly half-life and requires daily injections that often cause side effects
promote the inflammasome or modulate IL-1b mRNA transcrip- at the site of the subcutaneous injection, limiting its therapeutic
tion. In theory, adipocyte death, which has been suggested to be potential. Inhibition of IL-1 may be of more benefit in reversing
the driving force of adipose tissue inflammation by attracting hyperglycemia-associated insulin resistance. It should be also
macrophages (Cinti et al., 2005), may also activate the inflamma- underlined that one cannot exclude the possibility that effects
some, since dying cells are known to release uric acid to alert the on hepatic insulin resistance explain the effects observed in
immune system (Shi et al., 2003). the study of Larsen et al. (2007).

Cell Metabolism 15, January 4, 2012 ª2012 Elsevier Inc. 15


Cell Metabolism

Review

More recently, the development of IL-1b-neutralizing anti- REFERENCES


bodies that have successfully ameliorated insulin resistance
(Donath et al.,2008; Sloan-Lancaster et al.,2011; Rissanen Ali, S.R., Timmer, A.M., Bilgrami, S., Park, E.J., Eckmann, L., Nizet, V., and
Karin, M. (2011). Anthrax toxin induces macrophage death by p38 MAPK inhi-
et al.,2011) has been reported. These preparations have a longer bition but leads to inflammasome activation via ATP leakage. Immunity 35,
half-life and require fewer injections and may therefore bear 34–44.
superior therapeutic potential. Nevertheless, it must be empha- Anderson, E.K., Gutierrez, D.A., and Hasty, A.H. (2010). Adipose tissue recruit-
sized that, to date, no clinical studies using anti-IL-1b therapies ment of leukocytes. Curr. Opin. Lipidol. 21, 172–177.
have reported any improvement in insulin sensitivity levels.
Arend, W.P., Palmer, G., and Gabay, C. (2008). IL-1, IL-18, and IL-33 families of
A second potential approach would be direct inhibition of cas- cytokines. Immunol. Rev. 223, 20–38.
pase-1. Specific caspase-1 inhibitors have potent beneficial
Baldwin, W., McRae, S., Marek, G., Wymer, D., Pannu, V., Baylis, C., Johnson,
effects in animal models of insulin resistance and type 2 diabetes R.J., and Sautin, Y.Y. (2011). Hyperuricemia as a mediator of the proinflamma-
(Stienstra et al., 2010), and such pharmacological agents may tory endocrine imbalance in the adipose tissue in a murine model of the meta-
prove useful in humans as well. However, at this moment, no bolic syndrome. Diabetes 60, 1258–1269.

caspase-1 inhibitors are available for human use. Interestingly, Böni-Schnetzler, M., Thorne, J., Parnaud, G., Marselli, L., Ehses, J.A., Kerr-
Glyburide, a frequently used sulfonylurea drug for the treatment Conte, J., Pattou, F., Halban, P.A., Weir, G.C., and Donath, M.Y. (2008).
Increased interleukin (IL)-1beta messenger ribonucleic acid expression in
of type 2 diabetes, has been shown to inhibit the NLRP3 inflam- beta -cells of individuals with type 2 diabetes and regulation of IL-1beta in
masome (Lamkanfi et al., 2009; Masters et al., 2010), and one human islets by glucose and autostimulation. J. Clin. Endocrinol. Metab. 93,
may hypothesize that at least part of its effects may be due to 4065–4074.

its anti-inflammatory action. Browning, L.M. (2003). n-3 Polyunsaturated fatty acids, inflammation and
However, one has to keep in mind that caspase-1 cleaves obesity-related disease. Proc. Nutr. Soc. 62, 447–453.
alternative substrates including caspase-7 (Lamkanfi et al., Cani, P.D., Bibiloni, R., Knauf, C., Waget, A., Neyrinck, A.M., Delzenne, N.M.,
2008), which may mediate its deleterious effects during the and Burcelin, R. (2008). Changes in gut microbiota control metabolic endotox-
development of obesity and insulin resistance. Therefore, thera- emia-induced inflammation in high-fat diet-induced obesity and diabetes in
mice. Diabetes 57, 1470–1481.
pies aimed at specifically blocking caspase-1 may prove to be
more successful in attenuating type 2 diabetes, as compared Chavez, J.A., Knotts, T.A., Wang, L.P., Li, G., Dobrowsky, R.T., Florant, G.L.,
and Summers, S.A. (2003). A role for ceramide, but not diacylglycerol, in the
to strategies aimed solely at blockage of IL-1b. antagonism of insulin signal transduction by saturated fatty acids. J. Biol.
Chem. 278, 10297–10303.

Summary Cheung, K.J., Tzameli, I., Pissios, P., Rovira, I., Gavrilova, O., Ohtsubo, T.,
Chen, Z., Finkel, T., Flier, J.S., and Friedman, J.M. (2007). Xanthine oxidore-
Inflammation is one of the key events that underlie the develop- ductase is a regulator of adipogenesis and PPARgamma activity. Cell Metab.
ment of obesity-induced insulin resistance. Although different 5, 115–128.
roads may lead to its activation, the contribution of IL-1b to
Cinti, S., Mitchell, G., Barbatelli, G., Murano, I., Ceresi, E., Faloia, E., Wang, S.,
the development of insulin resistance at the level of the b cell, Fortier, M., Greenberg, A.S., and Obin, M.S. (2005). Adipocyte death defines
as well as peripherally, in obese individuals is now well estab- macrophage localization and function in adipose tissue of obese mice and hu-
mans. J. Lipid Res. 46, 2347–2355.
lished. Active IL-1b is produced by cleavage of pro-IL-1b by
caspase-1, which is part of the inflammasome protein complex. Csak, T., Ganz, M., Pespisa, J., Kodys, K., Dolganiuc, A., and Szabo, G. (2011).
Although most studies performed to date provide indirect and Fatty acid and endotoxin activate inflammasomes in mouse hepatocytes that
release danger signals to stimulate immune cells. Hepatology 54, 133–144.
associative evidence, growing evidence indicates that the in-
flammasome can be activated by fatty acids, high glucose Dasu, M.R., Devaraj, S., and Jialal, I. (2007). High glucose induces IL-1beta
expression in human monocytes: mechanistic insights. Am. J. Physiol. Endo-
levels, uric acid, and IAPP, linking metabolic danger signals to crinol. Metab. 293, E337–E346.
activation of IL-1b synthesis. The described correlations and
associations, however, do not necessarily prove causative rela- Dinarello, C.A. (2011). Interleukin-1 in the pathogenesis and treatment of
inflammatory diseases. Blood 117, 3720–3732.
tionships.
Inhibition of the IL-1R signaling or IL-1b production by the Donath, M.Y., and Shoelson, S.E. (2011). Type 2 diabetes as an inflammatory
disease. Nat. Rev. Immunol. 11, 98–107.
NLRP3-mediated activation of caspase-1 may ward off loss of
pancreatic b cell function, yet may also prevent the development Donath, M.Y., Størling, J., Maedler, K., and Mandrup-Poulsen, T. (2003).
Inflammatory mediators and islet beta-cell failure: a link between type 1 and
of insulin resistance in liver, muscle, and adipose tissue. type 2 diabetes. J. Mol. Med. (Berl) 81, 455–470.
Although clinical evidence is currently lacking, inhibition of the
IL-1R signaling or IL-1b production by the NLRP3-mediated acti- Donath, M.Y., Weder, C., Whitmore, J., Bauer, R.J., Der, K., Scannon, P.J.,
Dinarello, C.A., and Solinger, A.M. (2008). XOMA 052, an anti-IL-1b antibody,
vation of caspase-1 may avert the development of insulin resis- in a double-blind, placebo-controlled, does escalation study of the safety and
tance and represents an attractive therapeutic target to limit pharmacokinetics in patients with type 2 diabetes mellitus- a new approach to
therapy. Diabetologia 51, s7.
pathological complications associated with obesity, insulin
resistance, and type 2 diabetes. Dunne, A. (2011). Inflammasome activation: from inflammatory disease to
infection. Biochem. Soc. Trans. 39, 669–673.

ACKNOWLEDGMENTS Ehses, J.A., Perren, A., Eppler, E., Ribaux, P., Pospisilik, J.A., Maor-Cahn, R.,
Gueripel, X., Ellingsgaard, H., Schneider, M.K., Biollaz, G., et al. (2007).
Increased number of islet-associated macrophages in type 2 diabetes. Dia-
The authors would like to thank Sander Kersten for his valuable comments. betes 56, 2356–2370.
R.S. was supported by a Ruby Grant of the Dutch Diabetes Research Founda-
tion. M.G.N. was supported by a Vici Grant from the Netherlands Organization Elinav, E., Strowig, T., Kau, A.L., Henao-Mejia, J., Thaiss, C.A., Booth, C.J.,
for Scientific Research (NWO). Peaper, D.R., Bertin, J., Eisenbarth, S.C., Gordon, J.I., and Flavell, R.A.

16 Cell Metabolism 15, January 4, 2012 ª2012 Elsevier Inc.


Cell Metabolism

Review
(2011). NLRP6 inflammasome regulates colonic microbial ecology and risk for Lamkanfi, M., Mueller, J.L., Vitari, A.C., Misaghi, S., Fedorova, A., Deshayes,
colitis. Cell 145, 745–757. K., Lee, W.P., Hoffman, H.M., and Dixit, V.M. (2009). Glyburide inhibits the Cry-
opyrin/Nalp3 inflammasome. J. Cell Biol. 187, 61–70.
Erridge, C., and Samani, N.J. (2009). Saturated fatty acids do not directly stim-
ulate Toll-like receptor signaling. Arterioscler. Thromb. Vasc. Biol. 29, 1944– Larsen, C.M., Faulenbach, M., Vaag, A., Vølund, A., Ehses, J.A., Seifert, B.,
1949. Mandrup-Poulsen, T., and Donath, M.Y. (2007). Interleukin-1-receptor antag-
onist in type 2 diabetes mellitus. N. Engl. J. Med. 356, 1517–1526.
Goldfine, A.B., Silver, R., Aldhahi, W., Cai, D., Tatro, E., Lee, J., and Shoelson,
S.E. (2008). Use of salsalate to target inflammation in the treatment of insulin Lee, J.Y., Sohn, K.H., Rhee, S.H., and Hwang, D. (2001). Saturated fatty acids,
resistance and type 2 diabetes. Clin Transl Sci 1, 36–43. but not unsaturated fatty acids, induce the expression of cyclooxygenase-2
mediated through Toll-like receptor 4. J. Biol. Chem. 276, 16683–16689.
Gong, Y.N., Wang, X., Wang, J., Yang, Z., Li, S., Yang, J., Liu, L., Lei, X., and
Shao, F. (2010). Chemical probing reveals insights into the signaling mecha- Maedler, K., Sergeev, P., Ris, F., Oberholzer, J., Joller-Jemelka, H.I., Spinas,
nism of inflammasome activation. Cell Res. 20, 1289–1305. G.A., Kaiser, N., Halban, P.A., and Donath, M.Y. (2002). Glucose-induced
beta cell production of IL-1beta contributes to glucotoxicity in human pancre-
Gonzalez-Gay, M.A., Gonzalez-Juanatey, C., Vazquez-Rodriguez, T.R., atic islets. J. Clin. Invest. 110, 851–860.
Miranda-Filloy, J.A., and Llorca, J. (2010). Insulin resistance in rheumatoid
Mandrup-Poulsen, T. (2010). IAPP boosts islet macrophage IL-1 in type 2 dia-
arthritis: the impact of the anti-TNF-alpha therapy. Ann. N Y Acad. Sci.
betes. Nat. Immunol. 11, 881–883.
1193, 153–159.
Mandrup-Poulsen, T., Bendtzen, K., Nerup, J., Dinarello, C.A., Svenson, M.,
Gregor, M.F., and Hotamisligil, G.S. (2011). Inflammatory mechanisms in
and Nielsen, J.H. (1986). Affinity-purified human interleukin I is cytotoxic to iso-
obesity. Annu. Rev. Immunol. 29, 415–445.
lated islets of Langerhans. Diabetologia 29, 63–67.
Håversen, L., Danielsson, K.N., Fogelstrand, L., and Wiklund, O. (2009). Induc- Mandrup-Poulsen, T., Pickersgill, L., and Donath, M.Y. (2010). Blockade of
tion of proinflammatory cytokines by long-chain saturated fatty acids in human interleukin 1 in type 1 diabetes mellitus. Nat Rev Endocrinol 6, 158–166.
macrophages. Atherosclerosis 202, 382–393.
Mariathasan, S., Weiss, D.S., Newton, K., McBride, J., O’Rourke, K., Roose-
Hornung, V., Ablasser, A., Charrel-Dennis, M., Bauernfeind, F., Horvath, G., Girma, M., Lee, W.P., Weinrauch, Y., Monack, D.M., and Dixit, V.M. (2006).
Caffrey, D.R., Latz, E., and Fitzgerald, K.A. (2009). AIM2 recognizes cytosolic Cryopyrin activates the inflammasome in response to toxins and ATP. Nature
dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 440, 228–232.
458, 514–518.
Martinon, F., Pétrilli, V., Mayor, A., Tardivel, A., and Tschopp, J. (2006). Gout-
Huvers, F.C., Popa, C., Netea, M.G., van den Hoogen, F.H., and Tack, C.J. associated uric acid crystals activate the NALP3 inflammasome. Nature 440,
(2007). Improved insulin sensitivity by anti-TNFalpha antibody treatment in 237–241.
patients with rheumatic diseases. Ann. Rheum. Dis. 66, 558–559.
Masters, S.L., Dunne, A., Subramanian, S.L., Hull, R.L., Tannahill, G.M., Sharp,
Ito, A., Suganami, T., Yamauchi, A., Degawa-Yamauchi, M., Tanaka, M., F.A., Becker, C., Franchi, L., Yoshihara, E., Chen, Z., et al. (2010). Activation of
Kouyama, R., Kobayashi, Y., Nitta, N., Yasuda, K., Hirata, Y., et al. (2008). the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism
Role of CC chemokine receptor 2 in bone marrow cells in the recruitment of for enhanced IL-1b in type 2 diabetes. Nat. Immunol. 11, 897–904.
macrophages into obese adipose tissue. J. Biol. Chem. 283, 35715–35723.
Masters, S.L., Latz, E., and O’Neill, L.A. (2011). The inflammasome in athero-
Jager, J., Grémeaux, T., Cormont, M., Le Marchand-Brustel, Y., and Tanti, J.F. sclerosis and type 2 diabetes. Sci. Transl. Med. 3, ps17.
(2007). Interleukin-1beta-induced insulin resistance in adipocytes through
down-regulation of insulin receptor substrate-1 expression. Endocrinology McGillicuddy, F.C., Harford, K.A., Reynolds, C.M., Oliver, E., Claessens, M.,
148, 241–251. Mills, K.H., and Roche, H.M. (2011). Lack of interleukin-1 receptor I (IL-1RI)
protects mice from high-fat diet-induced adipose tissue inflammation coinci-
Jin, W., and Patti, M.E. (2009). Genetic determinants and molecular pathways dent with improved glucose homeostasis. Diabetes 60, 1688–1698.
in the pathogenesis of Type 2 diabetes. Clin. Sci. 116, 99–111.
Meijer, K., de Vries, M., Al-Lahham, S., Bruinenberg, M., Weening, D., Dijkstra,
Joosten, L.A., Netea, M.G., Fantuzzi, G., Koenders, M.I., Helsen, M.M., Spar- M., Kloosterhuis, N., van der Leij, R.J., van der Want, H., Kroesen, B.J., et al.
rer, H., Pham, C.T., van der Meer, J.W., Dinarello, C.A., and van den Berg, W.B. (2011). Human primary adipocytes exhibit immune cell function: adipocytes
(2009). Inflammatory arthritis in caspase 1 gene-deficient mice: contribution of prime inflammation independent of macrophages. PLoS ONE 6, e17154.
proteinase 3 to caspase 1-independent production of bioactive interleukin-
Meissner, F., Seger, R.A., Moshous, D., Fischer, A., Reichenbach, J., and Zy-
1beta. Arthritis Rheum. 60, 3651–3662.
chlinsky, A. (2010). Inflammasome activation in NADPH oxidase defective
mononuclear phagocytes from patients with chronic granulomatous disease.
Joosten, L.A., Netea, M.G., Mylona, E., Koenders, M.I., Malireddi, R.K., Oost-
ing, M., Stienstra, R., van de Veerdonk, F.L., Stalenhoef, A.F., Giamarellos- Blood 116, 1570–1573.
Bourboulis, E.J., et al. (2010). Engagement of fatty acids with Toll-like receptor
Minn, A.H., Hafele, C., and Shalev, A. (2005). Thioredoxin-interacting protein is
2 drives interleukin-1b production via the ASC/caspase 1 pathway in monoso-
stimulated by glucose through a carbohydrate response element and induces
dium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum. 62,
beta-cell apoptosis. Endocrinology 146, 2397–2405.
3237–3248.
Muruve, D.A., Pétrilli, V., Zaiss, A.K., White, L.R., Clark, S.A., Ross, P.J., Parks,
Koenen, T.B., Stienstra, R., van Tits, L.J., de Graaf, J., Stalenhoef, A.F., Joos- R.J., and Tschopp, J. (2008). The inflammasome recognizes cytosolic micro-
ten, L.A., Tack, C.J., and Netea, M.G. (2011a). Hyperglycemia activates cas- bial and host DNA and triggers an innate immune response. Nature 452,
pase-1 and TXNIP-mediated IL-1beta transcription in human adipose tissue. 103–107.
Diabetes 60, 517–524.
Nakagawa, T., Hu, H., Zharikov, S., Tuttle, K.R., Short, R.A., Glushakova, O.,
Koenen, T.B., Stienstra, R., van Tits, L.J., Joosten, L.A., van Velzen, J.F., Hij- Ouyang, X., Feig, D.I., Block, E.R., Herrera-Acosta, J., et al. (2006). A causal
mans, A., Pol, J.A., van der Vliet, J.A., Netea, M.G., Tack, C.J., et al. (2011b). role for uric acid in fructose-induced metabolic syndrome. Am. J. Physiol.
The inflammasome and caspase-1 activation: a new mechanism underlying Renal Physiol. 290, F625–F631.
increased inflammatory activity in human visceral adipose tissue. Endocri-
nology 152, 3769–3778. Netea, M.G., Joosten, L.A., Lewis, E., Jensen, D.R., Voshol, P.J., Kullberg,
B.J., Tack, C.J., van Krieken, H., Kim, S.H., Stalenhoef, A.F., et al. (2006). Defi-
Krebs, M., and Roden, M. (2005). Molecular mechanisms of lipid-induced ciency of interleukin-18 in mice leads to hyperphagia, obesity and insulin resis-
insulin resistance in muscle, liver and vasculature. Diabetes Obes. Metab. 7, tance. Nat. Med. 12, 650–656.
621–632.
Netea, M.G., Nold-Petry, C.A., Nold, M.F., Joosten, L.A., Opitz, B., van der
Lamkanfi, M., Kanneganti, T.D., Van Damme, P., Vanden Berghe, T., Vanover- Meer, J.H., van de Veerdonk, F.L., Ferwerda, G., Heinhuis, B., Devesa, I.,
berghe, I., Vandekerckhove, J., Vandenabeele, P., Gevaert, K., and Núñez, G. et al. (2009). Differential requirement for the activation of the inflammasome
(2008). Targeted peptidecentric proteomics reveals caspase-7 as a substrate for processing and release of IL-1beta in monocytes and macrophages. Blood
of the caspase-1 inflammasomes. Mol. Cell. Proteomics 7, 2350–2363. 113, 2324–2335.

Cell Metabolism 15, January 4, 2012 ª2012 Elsevier Inc. 17


Cell Metabolism

Review
Parikh, H., Carlsson, E., Chutkow, W.A., Johansson, L.E., Storgaard, H., Poul- Stienstra, R., van Diepen, J.A., Tack, C.J., Zaki, M.H., van de Veerdonk, F.L.,
sen, P., Saxena, R., Ladd, C., Schulze, P.C., Mazzini, M.J., et al. (2007). TXNIP Perera, D., Neale, G.A., Hooiveld, G.J., Hijmans, A., Vroegrijk, I., et al. (2011).
regulates peripheral glucose metabolism in humans. PLoS Med. 4, e158. Inflammasome is a central player in the induction of obesity and insulin resis-
tance. Proc. Natl. Acad. Sci. USA 108, 15324–15329.
Perrone, L., Devi, T.S., Hosoya, K., Terasaki, T., and Singh, L.P. (2009). Thio-
redoxin interacting protein (TXNIP) induces inflammation through chromatin Stoltzman, C.A., Peterson, C.W., Breen, K.T., Muoio, D.M., Billin, A.N., and
modification in retinal capillary endothelial cells under diabetic conditions. J. Ayer, D.E. (2008). Glucose sensing by MondoA:Mlx complexes: a role for
Cell. Physiol. 221, 262–272. hexokinases and direct regulation of thioredoxin-interacting protein expres-
sion. Proc. Natl. Acad. Sci. USA 105, 6912–6917.
Rissanen, A., Howard, C., Botha, J., and Thuren, T. (2011). IL-1b antibody
(Canakinumab) improves insulin secretion rates in subjects with impaired Tschopp, J. (2011). Mitochondria: Sovereign of inflammation? Eur. J. Immunol.
glucose tolerance (IGT) and type 2 diabetes (T2DM). Diabetes 60 (Suppl 1A ), 41, 1196–1202.
LB11.
van Asseldonk, E.J., Stienstra, R., Koenen, T.B., Joosten, L.A., Netea, M.G.,
Shanmugam, N., Reddy, M.A., Guha, M., and Natarajan, R. (2003). High and Tack, C.J. (2011). Treatment with Anakinra improves disposition index
glucose-induced expression of proinflammatory cytokine and chemokine but not insulin sensitivity in nondiabetic subjects with the metabolic syndrome:
genes in monocytic cells. Diabetes 52, 1256–1264. a randomized, double-blind, placebo-controlled study. J. Clin. Endocrinol.
Metab. 96, 2119–2126.
Shi, Y., Evans, J.E., and Rock, K.L. (2003). Molecular identification of a danger
van de Veerdonk, F.L., Smeekens, S.P., Joosten, L.A., Kullberg, B.J., Dinar-
signal that alerts the immune system to dying cells. Nature 425, 516–521.
ello, C.A., van der Meer, J.W., and Netea, M.G. (2010). Reactive oxygen
species-independent activation of the IL-1beta inflammasome in cells from
Shi, H., Kokoeva, M.V., Inouye, K., Tzameli, I., Yin, H., and Flier, J.S. (2006).
patients with chronic granulomatous disease. Proc. Natl. Acad. Sci. USA
TLR4 links innate immunity and fatty acid-induced insulin resistance. J. Clin.
107, 3030–3033.
Invest. 116, 3015–3025.
Vandanmagsar, B., Youm, Y.H., Ravussin, A., Galgani, J.E., Stadler, K.,
Sloan-Lancaster, J., Abu-Raddad, E., Polzer, J., Miller, J.W., Scherer, J.C., Mynatt, R.L., Ravussin, E., Stephens, J.M., and Dixit, V.D. (2011). The
Berg, J.K., and Landschulz, W.H. (2011). Safety, tolerability and efficacy of NLRP3 inflammasome instigates obesity-induced inflammation and insulin
subcutaneous (SC) LY2189102 (LY), a neutralizing IL-1b antibody, in patients resistance. Nat. Med. 17, 179–188.
(Pts) with type 2 diabetes (T2DM). Diabetes 60 (Suppl 1A ), LB14.
Vincent, J.A., and Mohr, S. (2007). Inhibition of caspase-1/interleukin-1beta
Spranger, J., Kroke, A., Möhlig, M., Hoffmann, K., Bergmann, M.M., Ristow, signaling prevents degeneration of retinal capillaries in diabetes and galacto-
M., Boeing, H., and Pfeiffer, A.F. (2003). Inflammatory cytokines and the risk semia. Diabetes 56, 224–230.
to develop type 2 diabetes: results of the prospective population-based Euro-
pean Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Wen, H., Gris, D., Lei, Y., Jha, S., Zhang, L., Huang, M.T., Brickey, W.J., and
Study. Diabetes 52, 812–817. Ting, J.P. (2011). Fatty acid-induced NLRP3-ASC inflammasome activation
interferes with insulin signaling. Nat. Immunol. 12, 408–415.
Steinberg, G.R., and Kemp, B.E. (2009). AMPK in Health and Disease. Physiol.
Rev. 89, 1025–1078. Westermark, P., Andersson, A., and Westermark, G.T. (2011). Islet amyloid
polypeptide, islet amyloid, and diabetes mellitus. Physiol. Rev. 91, 795–826.
Stienstra, R., Joosten, L.A., Koenen, T., van Tits, B., van Diepen, J.A., van den
Berg, S.A., Rensen, P.C., Voshol, P.J., Fantuzzi, G., Hijmans, A., et al. (2010). Zhou, R., Tardivel, A., Thorens, B., Choi, I., and Tschopp, J. (2010). Thiore-
The inflammasome-mediated caspase-1 activation controls adipocyte differ- doxin-interacting protein links oxidative stress to inflammasome activation.
entiation and insulin sensitivity. Cell Metab. 12, 593–605. Nat. Immunol. 11, 136–140.

18 Cell Metabolism 15, January 4, 2012 ª2012 Elsevier Inc.

You might also like