ArticlePDF AvailableLiterature Review

Emerging insights into molecular mechanisms underlying pyroptosis and functions of inflammasomes in diseases

Authors:

Abstract

Pyroptosis is a form of necrotic and inflammatory programmed cell death, which could be characterized by cell swelling, pore formation on plasma membranes, and release of proinflammatory cytokines (IL‐1β and IL‐18). The process of pyroptosis presents as dual effects: protecting multicellular organisms from microbial infection and endogenous dangers; leading to pathological inflammation if overactivated. Two pathways have been found to trigger pyroptosis: caspase‐1 mediated inflammasome pathway with the involvement of NLRP1‐, NLRP3‐, NLRC4‐, AIM2‐, pyrin‐inflammasome (canonical inflammasome pathway) and caspase‐4/5/11‐mediated inflammasome pathway (noncanonical inflammasome pathway). Gasdermin D (GSDMD) has been proved to be a substrate of inflammatory caspases (caspase‐1/4/5/11), and the cleaved N‐terminal domain of GSDMD oligomerizes to form cytotoxic pores on the plasma membrane. Here, we mainly reviewed the up to date mechanisms of pyroptosis, and began with the inflammasomes as the activator of caspase‐1/caspase‐11, 4, and 5. We further discussed these inflammasomes functions in diseases, including infectious diseases, sepsis, inflammatory autoimmune diseases, and neuroinflammatory diseases. We summarized the updated molecular mechanisms implicated in the regulation of inflammasomes which present as the activators of canonical/noncanonical inflammasome pathways of pyroptosis, and these inflammasomes function in relevant infectious, autoinflammatory, and neuroinflammatory diseases.
J Cell Physiol. 2019;115. wileyonlinelibrary.com/journal/jcp © 2019 Wiley Periodicals, Inc.
|
1
Received: 19 June 2019
|
Accepted: 27 September 2019
DOI: 10.1002/jcp.29268
REVIEW ARTICLE
Emerging insights into molecular mechanisms underlying
pyroptosis and functions of inflammasomes in diseases
Fangfang Lu
1,2
|
Zhixin Lan
2
|
Zhaoqi Xin
2
|
Chunrong He
2
|
Zimeng Guo
2
|
Xiaobo Xia
1
|
Tu Hu
1
1
Department of Ophthalmology, Xiangya
Hospital, Central South University, Changsha,
Hunan Province, China
2
Xiangya School of Medicine, Central South
University, Changsha, Hunan Province, China
Correspondence
Tu Hu, Department of Ophthalmology,
Xiangya Hospital, Central South University,
Changsha, 410013 Hunan Province, China.
Email: hutu1986@csu.edu.cn
Funding information
National Natural Science Foundation of China,
Grant/Award Number: 81900890; College
studentsinnovative project of CSU, Grant/
Award Number: ZY2016698; China
Postdoctoral Science Foundation, Grant/
Award Number: 2018M643004
Abstract
Pyroptosis is a form of necrotic and inflammatory programmed cell death, which
could be characterized by cell swelling, pore formation on plasma membranes, and
release of proinflammatory cytokines (IL1βand IL18). The process of pyroptosis
presents as dual effects: protecting multicellular organisms from microbial infection
and endogenous dangers; leading to pathological inflammation if overactivated. Two
pathways have been found to trigger pyroptosis: caspase1 mediated inflammasome
pathway with the involvement of NLRP1, NLRP3, NLRC4, AIM2, pyrininflamma-
some (canonical inflammasome pathway) and caspase4/5/11mediated inflamma-
some pathway (noncanonical inflammasome pathway). Gasdermin D (GSDMD) has
been proved to be a substrate of inflammatory caspases (caspase1/4/5/11), and the
cleaved Nterminal domain of GSDMD oligomerizes to form cytotoxic pores on the
plasma membrane. Here, we mainly reviewed the up to date mechanisms of
pyroptosis, and began with the inflammasomes as the activator of caspase1/caspase
11, 4, and 5. We further discussed these inflammasomes functions in diseases,
including infectious diseases, sepsis, inflammatory autoimmune diseases, and
neuroinflammatory diseases.
KEYWORDS
caspases, GSDMD, inflammasomes, pyroptosis
1
|
INTRODUCTION
Pyroptosis was first described as lysis occurring following infection of
Shigella Flexner in macrophages in 1992, which was classified as
apoptosis mistakenly since initial recognition of programmed cell
death was limited to apoptosis (Zychlinsky, Prevost, & Sansonetti,
1992). In 2001, pyroptosis was first proposed by Cookson and
Brennan (2001), and was originally defined to describe the
nonapoptotic, caspase1 dependent cell death of Salmonellainfected
macrophages.The subsequent research works identified that
pyroptosis was characterized by pore formation in the plasma
membranes, cell swelling and release of inflammatory intracellular
contents, which was substantially different from apoptosis (Table 1;
Aachoui, Sagulenko, Miao, & Stacey, 2013; Bergsbaken, Fink, &
Cookson, 2009). Pyroptosis is emerging as a form of inflammatory
programmed cell death mediated by inflammatory caspases (Man,
Karki, & Kanneganti, 2017). As the discovery of gasdermin D
(GSDMD), pyroptosis was redefined as gasderminmediated pro-
grammed necrosis (Figure 1; Shi, Gao, & Shao, 2017).
Pyroptosis is substantially classified as innate immune response
which can be triggered by the process of recognizing structures
damageassociated molecular patterns (DAMPs) and conserved patho-
genassociated molecular patterns (PAMPs) by host pattern recognition
receptors (PRRs; Takeuchi & Akira, 2010). Previous studies revealed
that PRRs presented as two types: the transmembrane and the
cytoplasmic. And the latter one facilitated the secretion of proin-
flammatory cytokines and removed the intracellular replicative niche of
the bacteria through mediating a series of signal cascades in cells at the
first line of defense against infection. These were considered as the key
events of pyroptosis (Lamkanfi & Dixit, 2014). In case of sensing
PAMPs/DAMPs, the cytoplasmic PRRs, including nucleotidebinding
domain (NBD)like receptors (NLRs), absent in melanoma 2like
receptors (ALRs), and Pyrin, assembled into highmolecular caspase
activating molecular platforms—“inflammasomes,which served for
recruitment of the inflammatory caspases (Martinon, Burns, & Tschopp,
2002). Canonical inflammasomes are composed of cytoplasmic PRRs,
adapter protein apoptosisassociated specklike protein containing a
CARD (ASC), and procaspase1 (Proell, Gerlic, Mace, Reed, & Riedl,
2013). Several canonical inflammasomes have been identified to
mediate pyroptosis in vivo, denoted by different cytoplasmic PRRs,
including NLRP1 inflammasome (ChavarriaSmith & Vance, 2015),
NLRP3 inflammasome (Agostini et al., 2004), NLRC4 inflammasome
(Poyet et al., 2001), AIM2 inflammasome (Burckstummer et al., 2009),
and Pyrin inflammasome (de Zoete & Flavell, 2014). The activated
canonical inflammasomes could process zymogen procaspase1intothe
active noncovalently linked subunits p10 and p20 (active caspase1),
and the latter could facilitate the maturation of proinflammatory
cytokines (proIL18 and proIL1β) and induce the formation of pore on
plasma membranes (Elliott, Rouge, Wiesmann, & Scheer, 2009).
Previous studies demonstrated that the pores on plasma membranes
permitted ions to permeate and dissipated the cellular ionic gradient,
which led to a multifaceted cascade of events: cell swelling, release of
cytosolic pathogens, and inflammatory cytokines (IL1βand IL18; Miao
et al., 2010). Due to these pores, the immune defense function of
pyroptosis is realized by the released mature inflammatory cytokines
for recruitment of neutrophils and expelled bacteria for neutrophil
mediated killing in situ (Xia, Wang, Zheng, Jiang, & Hu, 2019). Recently,
Feng et al. reported GSDMD as a generic substrate for caspase1and
highlighted the crucial role of gasdermin DN terminus (GSDMDNT)in
the formation of these pores (Ding et al., 2016; Shi et al., 2015). Except
the caspase1 meditated canonical inflammasome pathway of pyropto-
sis, a series of subsequent studies identified murine caspase11 and
human caspase4/5 as cytosolic sensors in response to cytosolic
lipopolysaccharide (LPS) to meditate noncanonical inflammasome
pathway of pyroptosis (Baker et al., 2015; Kayagaki et al., 2011). In
depth studies demonstrated that noncanonical inflammasome could
also cleave GSDMD, triggering caspase1 independent pyroptosis, and
proinflammatory cytokines secretion (Figure 2; Kayagaki et al., 2015;
Yang, Zhao, & Shao, 2015).
In this review, we summarized the updated molecular mechan-
isms implicated in the regulation of inflammasomes which present as
the activators of canonical/noncanonical inflammasome pathways of
pyroptosis, and these inflammasomes function in relevant infectious,
autoinflammatory, and neuroinflammatory diseases.
2
|
NLR FAMILY RELEVENT
INFLAMMASOME
NLRs structurally characterized by a Cterminal LRR domain that
could recognize cytosolic ligands and retain the NLRs in an
TABLE 1 Characteristic comparisons between pyroptosis and apoptosis
Mechanism Morphology Function
Caspases Cellular contents DNA
Plasma
membrane Nucleus
Cyto-
plasm Reaction Result
Pyroptosis Caspase1, 4, 5,
and 11
Released through pores
on plasma membrane
DNA fragmentation Pores formation Intact nucleus Swelling Inflammatory response Expulsion of cytoplasmic
pathogens
Apoptosis Caspase3, 6, 7,
and 8
Packaged in apoptotic
body
Characteristic oligonucleosomal
DNA fragments
Intact plasma
membrane
Damaged
nucleus
Shrinkage Noninflammatory
response
Clearance of damaged
cells
2
|
LU ET AL.
FIGURE 1 Milestones in the history of
pyroptosis exploration
FIGURE 2 Caspase1 mediated canonical inflammasome pathway and caspase4/5/11 mediated noncanonical inflammasome pathway of
pyroptosis. (Domain positions arent drawn to scale). In caspase1 mediated canonical inflammasome pathway of pyroptosis, different PAMPs or
DAMPs activate corresponding cytoplasm inflammasome sensors respectively, including NLRP1, NLRP3, NLRC4, AIM2, and Pyrin, and then
recruit caspase1 with or without adapter protein ASC to form inflammasome. The inflammasome subsequently triggers its dimerization and
autoproteolytic converting of procaspase1 into the active subunits p10 and p20. Activated caspase1 subsequently cleaves its downstream
effectors: the executioner GSDMD and proIL1β/proIL18. In noncanonical inflammasome pathway, cytosolic LPS from Gramnegative bacteria
promotes activation of murine caspase11 or human caspase4/5 directly. The activated caspase11/4/5 also cleaves GSDMD, but it stimulates the
cleavage of proIL1βand proIL18 by activating the Nlrp3Asccaspase1 noncanonical inflammasome. The cleaved GSDMD via canonical or
noncanonical inflammasome pathway release the functional GSDMDNT, which forms pores on the plasma membrane and subsequently induces
pyroptosis and release of mature IL1β/IL18. ASC, apoptosisassociated specklike protein containing a CARD; CARD, caspase activation and
recruitment domain; FIIND, functiontofind domain; GSDMD, gasdermin D; GSDMDNT, gasdermin D Nterminal; GSDMDCT, gasdermin D
Cterminal; LRR, leucinerich repeat; NATCH/ NOD, nucleotidebinding oligomerization domain; PYD, Pyrin domain
LU ET AL.
|
3
nonactivated status, a central highly conserved nucleotidebinding
oligomerization domain (NACHT) that functions in process of
oligomerization, and a variable Nterminal that mediates homotypic
interactions with downstream proteins (Lechtenberg, Mace, & Riedl,
2014). Different Nterminal domains have been used as a structural
sub classification, like NLRP with a PYD (Pyrin domain), NLRB with
BIR domain (baculovirus inhibitory repeat domain), and NLRC with a
CARD (caspase activation and recruitment domain; Sharma &
Kanneganti, 2016). The most wellrecognized inflammasomeforming
members in NLR family include NLRP3, NLRP1, and NLRC4 (Guo,
Callaway, & Ting, 2015). Upon recognizing specific stimuli, the
corresponding NLRs (NLRP3, NLRP1, and NLRC4) nucleates an
adapter protein ASC (composed of PYDCARD), forming an ASC
speck structure around nucleus for the subsequently clustering of
procaspase1 (Lu et al., 2014). Previous studies showed that the PYD
containing NLRP3 exclusively requires ASC for inflammasome
assembly via NLRP3
PYD
ASC
PYD
and ASC
CARD
procaspase1
CARD
interaction, whereas CARDcontaining NLRP1 and NLRC4 could
recruit procaspase1 directly through CARDCARD interaction
(Figure 2; Lechtenberg et al., 2014). Howbeit, some other studies
also claimed that NLRC4/NLRP1 induced procaspase1 activation
and cytokine processing could be significantly enhanced in the
presence of ASC, since its bipartite PYDCARD structure might
stabilize the CARDCARD interaction (Van Opdenbosch et al., 2014).
Thus, there remains a dispute on whether ASC is unnecessary for the
activation of NLRP1/NLRC4 inflammasome.
3
|
NLRP3 INFLAMMASOME
NLRP3 is the most extensively investigated sensor proteins in NLR
family and features a PYD in the Nterminal. To date, a twostep
model has been wellestablished in NLRP3 inflammasome activation:
a priming signal requested to upregulate NLRP3 and proIL1β, and a
second signal required for creating a functional inflammasome (He,
Hara, & Núñez, 2016). The priming signal could be simulated by
pattern recognition receptors, like TLR4, NOD2, TNFR, and IL1R. All
these signaling could cause activation of nuclear factor kappa B (NF
κB), which led to increased synthesis of proIL1βand NLRP3
(Bauernfeind et al., 2009). Juliana et al. (2012) found an additional
rapid priming event: pattern recognition receptor TLR4 mediated
NLRP3 posttranslational deubiquitylation signaling by MyD88. Py,
Kim, VakifahmetogluNorberg, & Yuan (2013) further discovered that
BRCC3 functioned as the deubiquitinase in mouse cells that
specifically stimulated the deubiquitylation of the LRR domain of
NLRP3.Notably, posttranslational modification of protein is not
unique to NLRP3 deubiquitylation, subsequent research works
discovered SYKand JNKdependent ASC phosphorylation and
LUBACmediated ASC ubiquitination to be novel priming processes
(Elliott & Sutterwala, 2015). In general, PRR signaldependent
priming in transcriptional or posttranslational modification level
enables NLRP3 to be responsive to agonists. The second signal of
activation of the NLRP3 inflammasome was triggered by extensive
structurally dissimilar agonists, including environmental crystalline
pollutants like silica, pathogenderived ligands like poreforming
toxins and nucleic acids as well as endogenous danger signals like
serum amyloid A and ATP (Elliott & Sutterwala, 2015).Due to the
diversity of its agonists, it was suggested that these unrelated
upstream activators might trigger cognate cellular downstream
events that directly activate NLRP3. Through further exploration,
researchers found that K
+
efflux, lysosomal disruption, calcium
signaling, mitochondriaderived factors like oxidized mitochondrial
DNA (mt DNA), reactive oxygen species (ROS) and cardiolipin, and
microtubuledriven spatial arrangement of mitochondria were all
direct upstream signals required for NLRP3 inflammasome activation
(Tang et al., 2019; Wang, Yuan, Chen, & Wang, 2019; Yabal, Calleja,
Simpson, & Lawlor, 2019). Despite intensive scrutiny on this topic, a
unified mechanism has not emerged yet. He et al. (2016) proposed
that the activation of the NLRP3 inflammasome is the integration of
above cellular signaling events. However, how these multiple signals
are interacted to activate the NLRP3 inflammasome remains elusive.
The negative regulation of NLRP3 equals the importance of the
priming and activation events described above (Sutterwala, Haasken,
& Cassel, 2014).In the priming level, the myeloidspecific microRNA
223 (miR223) could specifically interact with the 3′‐untranslated
region of NLRP3 mRNA and target it for its degradation, thereby
interfering with the translation of NLRP3. Besides, type I interferons
(IFNs) could also suppress NLRP3 inflammasome priming by inducing
the upregulation of the antiinflammatory cytokine IL10 which
signaled through STAT3 to lessen expression levels of proIL1βand
proIL1α(Guarda et al., 2011). In the activating process, it was
suggested that nitric oxide (NO) inhibited the assembly of NLRP3
inflammasome by stabilizing mitochondria, thereby, obstructing IL1β
maturation and caspase1 activation (Mishra et al., 2013). In addition,
immunityrelated GTPase M (IRGM) was recently found to bind to
the conserved NACHT domain of NLRP3 and the PYD of ASC,
thereby limiting NLRP3/ASC oligomerization. On top of that, IRGM
could also target NLRP3 and ASC for their autophagic degradation
(Mehto et al., 2019; Nabar & Kehrl, 2019). To conclude, the
components both contributing to and negatively modulating the
activation of NLRP3 inflammasome ensure the validity of a perceived
threat with an appropriate response.
The activation of NLRP3 inflammasome exerts protective effects
against infection, including bacterium, virus, and fungi, through
caspase1 mediated pyroptosis (Xia et al., 2019). Nevertheless,
dysregulated NLRP3 inflammasome activation causes chronic in-
flammation, leading to neuroinflammatory diseases, and metabolic
diseases (Guo et al., 2015; Yin et al., 2019). For example, both spinal
cord injury (SCI) and traumatic brain injury (TBI) follow a biphasic
time course: after the initial primary injury resulting in neuronal
death, a complicated secondary phase of central nervous system
(CNS) injury cascade involving neuroinflammation follows, which are
mediated by proinflammatory cytokines. This secondary injury
causes further neurological deterioration and complicates the healing
process (Borgens & LiuSnyder, 2012). Previous studies identified the
expression of NLRP3 in neurons, astrocytes and microglia in rat
4
|
LU ET AL.
model of SCI and TBI (Liu et al., 2013). More recently research works
provided evidence in a SCI or TBI model that during 24 hr72 hr
postinjury, most of NLRP3 inflammasome components including
NLRP3, ASC, and caspase1were increased (Zendedel et al., 2018).
Besides, NLRP3 could be activated by αsynuclein (αsyn) and
amyloidβ(Aβ) via microglial endocytosis and succeeding lysosomal
release of cathepsin B. The activated NLRP3 inflammasome then
processed IL18 and IL1β, and ultimately induce Parkinson disease
(PD) and Alzheimer disease (AD) pathology and tissue damage
respectively (Zhou et al., 2016). Moreover, NLRP3 inflammasome
functioned to promote multiple sclerosis (MS) through IL18 and IL
1β, which induced Th17 and Th1 cell responses and migration (Inoue,
Williams, Gunn, & Shinohara, 2012). These evidence suggested the
crucial role of NLRP3 inflammasome in neuronal disorders. In
addition, it has been wellestablished in the past decade that chronic
inflammation contributes the development of metabolic disorders,
such as atherosclerosis obesity and type 2 diabetes mellitus (T2DM;
Donath & Shoelson, 2011). Recent evidence suggested that choles-
terol crystals could activate the NLRP3 inflammasome through
phagolysosomal damage and generate mature IL1βin both mouse
and human cells (Donath & Shoelson, 2011).Furthermore, elevated
glucose and islet amyloid polypeptide (IAPP) during T2DM could also
trigger the activation of NLRP3 inflammasome and result in
succeeding release of IL1β(Vandanmagsar et al., 2011; Zheng
et al., 2013).Based on these evidence, NLRP3 inflammasome induced
cellular pyroptosis may promote the progress of neurodegenerative
and metabolic diseases. Therefore, further explorations could be
focused on the inventions which compete against the formation of
NLRP3 inflammasomes to improve the treatment for these diseases
4
|
NLRP1 INFLAMMASOME
The structure of NLRP1 proteins shows speciesspecific differences
(Figure 3). It differs from the other NLRs in two respects. First, most
primates including human encode a single NLRP1, which contains
two annotated recruitment domains: A Cterminal CARD and a N
terminal PYD. In other NLRs, the PYD functions to recruit caspase1
via ASC, while in human NLRP1 (hNLRP) it was Cterminal CARD of
NLRP1 rather than PYD that mediated homotypic interactions with
downstream ASC and procaspase1 (Faustin et al., 2007).Notably, N
terminal PYD has recently been shown to be essential in sustaining
NLRP1 in its inactive state based on the observation that destabiliz-
ing the PYD of human NLRP1 increased selfoligomerization (Zhong
et al., 2016). NLRP1 in mice has undergone three paralogs (Nlrp1a, b,
and c) that contain an NR100 (Nterminal domain of rodent Nlrp1
proteins, approximately 100 amino acids) domain preceding the
NACHT domain (Moayeri, Sastalla, & Leppla, 2012). It was discovered
that proteolytic cleavage in the NR100 domain was necessary for
NLRP1B inflammasome formation and caspase activation (Chavarria
Smith & Vance, 2013). Second, between the LRR and the Cterminal
CARD of NLRP1 proteins, there is a unique stretch of 30 kDa domain
called functiontofinddomain (FIIND) (Finger et al., 2012). It was
reported that this domain possessed autoproteolytic activity and the
generated associated Cterminal and Nterminal fragments of NLRP1
was required for activating NLRP1 inflammasome. This autoproteo-
lytic activity is also identified as a maturation event before the
NLRP1 becoming responsive to activators (ChavarríaSmith, Mitchell,
Ho, Daugherty, & Vance, 2016). To date, activators of NLRP1 remain
partially elucidated. Some researchers identified that Nlrp1b in mice
and rats could be activated by reduced intracellular levels of ATP and
Toxoplasma gondii infections (Ewald, ChavarriaSmith, & Boothroyd,
2014; Liao & Mogridge, 2013). However, there are deficient evidence
to convincingly ascertain the role of ATP binding for NLRP1
activation and how T.gondii activates NLRP1 remains to be
discovered. For human NLRP1, there are no defined activators yet.
Although previous study showed that bacterial ligand muramyl
dipeptide (MDP) could induce NLRP1 oligomerization (Faustin et al.,
2007), growing evidence suggested that MDP is actually sensed by
other PRR pathway (Martino et al., 2016). Until now, the only well
FIGURE 3 Domain structure of NLRP1 proteins in human, mouse and rats. (Domain positions arent drawn to scale). All members of NLRP1
proteins contain NATCH, LRR, and CARD domains. Human NLRP1 also consists of a pyrin domain (PYD) in the Nterminal. Compared with
human NLRP1, mouse NLRP1 has undergone three paralogs, termed Nlrp1a, b, and c, that contain an NR100 domain preceding the NACHT
domain instead of a PYD and the linker connecting this two units is shorter. Autolytic cleavage in the NR100 domain and FIIND is indicated
respectively by the split in the structure of corresponding domains. The FIIND and CARD domains are missing in the structure of Nlrp1c, which
is not expected to form a functional inflammasome
LU ET AL.
|
5
identified activator of NLRP1 is anthrax lethal toxin (LT) which is
produced and secreted by Bacillus anthracis. It was show that LT
directly cleaved mouse Nlrp1b and rat Nlrp1at Nterminal and such
cleavage is needed for activation of Nlrp1 inflammasome and
following macrophage pyroptosis (ChavarriaSmith & Vance, 2013).
More recent studies indicate that the activation of human NLRP1 can
also be triggered by proteolysis in a specific Nterminal linker region
between the PYD and NBD domains, which also confirms the auto
inhibitory role of Nterminal PYD (ChavarriaSmith et al., 2016).
Based on these evidence, we could conclude that the prerequisites of
NLRP1 activation are as follows: autoproteolytic cleavage of FIIND,
the sensation of stimulus and the Nterminal proteolytic cleavage.
Anthrax disease is characterized by lung tissue necrosis,
circulatory collapse and pulmonary edema, which is resulted from
infection of B. anthracis whose major virulence determinant is lethal
toxin (LT; Quintiliani & Quintiliani, 2003). Studies indicated LT could
cleave the Nterminus of NLRP1b, and thereby activating the
inflammasome and inducing pyroptosis in macrophage after infection
with B. anthracis in susceptible mouse strains (Nour et al., 2009). LT
induced pyroptosis in murine dendritic cells and macrophages was
originally believed to contribute to pathology of anthrax disease due
to the reduced bacterial clearance by these immunocytes (Hanna,
Acosta, & Collier, 1993). Further exploration illustrated that
activation of NLRP1 could defend against B. anthracis infection by
inducing NLRP1caspase1 mediated pyroptosis, inflammatory signal-
ing, and neutrophil recruitment in situ (Moayeri et al., 2012). In
addition, the previous study indicated that infection with Toxoplasma
triggers an NLRP1 inflammasome mediated inflammatory responses,
limiting parasite load and dissemination, which is protective to the
host (Ewald et al., 2014). These evidence suggest that NLRC4
inflammasome mediated pyroptosis and inflammatory responses
contribute to hostdefense against infection. However, aberrant
inflammatory signal could have deleterious effects. Recent multiple
studies demonstrated that activating mutations of NLRP1 in patients
were associated with hereditary autoinflammatory diseases, like
Crohns disease, autoinflammatory skin disorder, systemic lupus
erythematosus, and rheumatoid arthritis (Cummings et al., 2010;
Grandemange et al., 2017; Kitamura, Sasaki, Abe, Kano, & Yasutomo,
2014; Pontillo et al., 2012). Furthermore, similar to NLRP3, NLRP1 is
also implicated in neuroinflammatory process in neurons (Mortezaee,
Khanlarkhani, Beyer, & Zendedel, 2018). A study using immunoblot
analysis of cerebrospinal fluid (CSF) samples from both adult TBI
patients and nontrauma normal control revealed significantly higher
levels of NLRP1 and caspase1 in TBI patients (Adamczak et al.,
2012). Likewise, coimmunoprecipitations of postSCI lysates in the
SCI rat model showed upregulated association between NLRP1 and
caspase1, suggesting NLRP1 contributing to the pathology of CNS
injury (Lin et al., 2016). Besides, growing evidence suggest NLRP1
also have a pathogenic role in AD (Saresella et al., 2016). Recently, a
study using APPswe/PS1dE9 transgenic mice (transgenic mice
developing chronic deposition of amyloidβ) showed that cerebral
NLRP1 levels were upregulated and when NLRP1 or caspase1 was
knock downed, considerably decreased neuronal pyroptosis and
reversed cognitive impairments emerged. In cultured cortical
neurons, it was found that NLRP1mediated pyroptosis increased in
response to Aβ, which further confirmed the pathogenic role of
NLRP1 inflammasome in AD progression (Guo et al., 2015). Of note,
evidence indicate that composition of neuronal NLRP1 inflamma-
some differs from that described in peripheral macrophages. The
NLRP1 inflammasome in neuron is a preassembled multiprotein
complex, comprising of NLRP1, ASC, caspase1, caspase11, and the
inhibitor of apoptosis protein Xlinked inhibitor of apoptosis protein
(XIAP). The existence of XIAP in this preassembled complex in
neurons is not fully understood, although the cleavage of XIAP might
reduce its capability of inhibiting caspases. And whether caspase11
in the structure of NLRP1 inflammasome in neuron has the same
function of caspase1 to induce pyroptosis requires further con-
firmation as the current reports on caspase11 mediated pyroptosis
are limited in infectious diseases (de Rivero Vaccari, Dietrich, &
Keane, 2014). In contrast to the promotional function of NLRP3
inflammasome in metabolic diseases, NLRP1 has been recently
discovered to counteract metabolic syndrome through inflamma-
some activation and IL18 production (Murphy et al., 2016). Soares
et al. (2018) elucidated that diabetes metabolites, like high glucose
and glycated albumin, could modulate NLRP1 expression and further
suggested that NLRP1 functioned as a homeostatic factor protecting
host against diabetic kidney disease (DKD) in type1 diabetes.
Regardless of welldefined NLRP3 inflammasome on neuroinflamma-
tory diseases and metabolic diseases, it should be a worthy research
path to explore new drug via targeting NLRP1.
5
|
NLRC4 INFLAMMASOME
The NLRC4 inflammasome, insisting of a Nterminal CARD, a central
NACHT and a Cterminal LRR, was initially named ICEprotease
activating factor (IPAF) for its function in activating caspase1 via
direct NLRC4
CARD
caspase1
CARD
interaction (Poyet et al., 2001). In
contrast to the diverse stimuli for the activation of NLRP3, the
NLRC4 has been currently believed to be activated to only two
bacterial components: flagellin and type III secretion systems (T3SS)
apparatus (rod protein and needle protein; Miao et al., 2010). Unlike
other inflammasomes, NLRC4 activation required another protein,
neuronal apoptosis inhibitory proteins (NAIPs), to recognize the
cytosolic presence of these bacterial components (Kofoed & Vance,
2011). In the absence of these bacterial agonists, LRR domain in
NAIP and NLRC4 curves back to occlude the NBD, preventing NBD
mediated oligomerization (Hu et al., 2013). And in the presence of
bacterial ligands, NAIP directly recognizes and binds to specific
ligands with an internal region containing several NBDassociated
αhelical domains and thus breaks its autoinhibitory conformational
states, allowing its NBDmediated cooligomerization with NLRC4
(Tenthorey, Kofoed, Daugherty, Malik, & Vance, 2014). Such co
oligomerization induces formation of a large NAIPNLRC4 inflamma-
some for activating caspase1 and eventually leading to release of
IL1β/IL18 and pyroptosis (Zhao & Shao, 2015). Howbeit how
6
|
LU ET AL.
NLRC4 autoinhibition is relieved to allow oligomerization with NAIP
proteins is elusive. NAIP protein, one of the NLR family member, is
featured by three tandem BIR domains at its Nterminus (Kofoed &
Vance, 2012). Mice have four NAIP paralogues: NAIP5/NAIP6
recognize flagellin; NAIP1 and NAIP2 specifically recognize T3SS
needle protein and rod protein respectively. By contrast, human only
encodes one single NAIP gene (hNAIP) and is able to detect both
bacterial T3SS rod/needle protein and flagellin (Reyes Ruiz et al.,
2017). The discovery of NAIPs in mice and human NAIP that act
functionally upstream of NLRC4 maximizes the capacity of innate
immune detection in coping with bacterial infections. It remains
unknown whether the formation of heterooligomeric structure is a
common feature of all NLRs or just unique to NLRC4. In addition to
the essential role of NAIPs functioning upstream of NLRC4
activation, recent several studies proposed that Ser533 phosphor-
ylation of NLRC4 prepared NLRC4 for subsequent activation by
NAIP detection of ligands (Matusiak et al., 2015). However, there are
also contradictory reports indicating that such phosphorylation event
also occurs in the inactive state of NLRC4 (Hu et al., 2013).
Therefore, it needs further investigation to determine the necessity
of the phosphorylation event.
Flagella and T3SS are molecular transport machineries employed
by bacteria to enhance the ability to reach and invade host cells
(Galán, LaraTejero, Marlovits, & Wagner, 2014). It has been well
established that NAIPNLRC4 inflammasome in host immune cells,
like dendritic cells and macrophages, could detect bacterial flagellin,
T3SS rod/needle protein during infection (Zhao & Shao, 2015). And
the NAIPNLRC4 inflammasome mediated pyroptosis promotes host
innate antibacterial responses by expelling bacteria from their
protected intracellular niche and by released IL1β/IL18 caused
inflammation (Bergsbaken et al., 2009; Zhao & Shao, 2015). However,
in vivo,some pathogenic bacteria develop strategies to escape from
NAIPNLRC4 inflammasome mediated innate immune defense. For
instance, S. typhimurium normally represses flagellin and SPI1 T3SS to
evade the detection by NAIPNLRC4 inflammasome and conversely
activates SPI2 T3SS at the late phase of infection, which lacks
inflammasomestimulating activity, to promote its replication within
macrophages (Miao et al., 2010). To identify the exact pathway of
hostdefense against pathogens, Miao et al. use engineered S.
typhimurium constitutively expressing flagellin to infect mice and
the results showed that NLRC4 inflammasome activation efficiently
cleared the infected S. typhimurium in macrophage. Importantly, this
clearance was mediated by caspase1mediated pyroptosis while IL
1β/IL18 induced inflammatory responses only played a minor role,
based on the evidence that mice devoid of both IL18 and IL1βalso
cleared S. typhimurium, whereas pyroptosis released bacteria from
macrophages and exposed them to be cleared by the neutrophil
(Miao et al., 2010). Except the functions of NAIPNLRC4 inflammma-
some in macrophages and dendritic cells, recent studies reported
protection role of NAIPNLRC4 inflammasome against enteric
pathogens, like Salmonella and Citrobacter rodentium, in intestinal
epithelial cells (Nordlander, Pott, & Maloy, 2014). Still, it required
further study to determine whether this represents a general defense
mechanism protecting the intestinal mucosa. Despite the essential
role of NLRP4 inflammasome in controlling bacterial infections,
hyperactivation of the NLRC4 inflammasome could cause autoin-
flammation in human and mice. A spectrum of studies identified a
gainoffunction mutations in the NBD of NLRC4 to be related with
familial cold autoinflammatory syndrome (FCAS), neonatal onset
multisystem inflammatory disease (NOMID), and autoinflammation
with infantile enterocolitis (AIFEC) (Romberg, Vogel, & Canna, 2017).
It is expected that mutations of NLRP4 may destabilize the
interaction between the NBD and wingedhelix domain (WHD), or
directly affect ADP binding, either of which may be crucial for
maintaining NLRC4 in an autoinhibited state (Hu et al., 2013). Thus,
these evidence underscore the essential roles of the NLRC4
inflammasome in autoinflammatory diseases and suggest novel
targets for therapy.
6
|
AIM2 AND IFI16 INFLAMMASOME
AIM2 and IFI16 belong to the IFNinducible p200protein family,
which is also called AIM2like receptors (ALRs). Eight ALRs (like
p202a, p202b, p204, and Aim2) were found in mouse, while four
ALRs have been previously annotated in humans (IFI16, MNDA, IFIX,
and AIM2; Brunette et al., 2012). It was found that AIM2 could
interact with cytosolic double stranded DNA (dsDNA) and assembles
ASC and procaspase1 to form a functional inflammasome complex
(FernandesAlnemri, Yu, Datta, Wu, & Alnemri, 2009). Recent studies
identified that IFI16 could also specifically recognize ASC and
procaspase1 in response to nuclear and cytosolic dsDNA, forming
a functional inflammasome for activating caspase1 (Kerur et al.,
2011). AIM2 and IFI16 are typical proteins of the ALRs, character-
ized by a PYD in the Nterminus and one or two partially conserved
repeat of 200amino acid residues (HIN200 domain) in the C
terminus. The HIN200 domain contains two adjacent OB domains
(oligonucleotide/oligosaccharidebinding domain), which could po-
tentially combine with dsDNA (FernandesAlnemri et al., 2009). PYD
in AIM2 partly binds its HIN domain by electrostatic attraction so
that AIM2 persistently exists in the autoinhibition state in resting
cells. The dsDNA has higher affinity for OB domain of HIN domain
compared with PYD domain. Therefore, after intracellular dsDNA
appears, PYD will be replaced by dsDNA and thereby the auto
inhibited state of AIM2 is liberated (Jin, Perry, Smith, Jiang, & Xiao,
2013). The PYD therefore is exposed to ASC, recruiting ASC through
homotypic PYDPYD interactions. Subsequently, An functional AIM2
inflammasome is assembled via combination of CARD of ASC and
CARD of procaspase1 which could induce pyroptosis and inflam-
matory responses (Choubey et al., 2010). Notably, both AIM2 and
IFI16 recognize dsDNA from a wide range of sources regardless of
their structure or sequences but the dsDNA strand is required to be
80 basepairs in length for optimum inflammasome activation. This
specific DNA recognition pattern is based on electrostatic charge
neutralization between the dsDNA sugarphosphate backbone and
the positively charged HIN domain residues (Jin et al., 2012).
LU ET AL.
|
7
Moreover, despite there is no oligomerization domain in AIM2 or
IFI16, oligomerization could be accomplished by the multivalent
ligand dsDNA, which offered platform for AIM2 to cluster on (Jin
et al., 2012). Recognition of cytosolic DNA by cells is vital to the
innate immune system to initiate immunological responses and also
an essential contributor to autoimmune diseases when implicating
selfDNA. Except the secretion of type I IFNs such as IFNβ,an
effective immune response to the occurrence of cytosolic DNA is
dependent on proinflammatory cytokines (Medzhitov, 2007). Pre-
vious studies have illustrated that AIM2 and IFI16 could recognize
dsDNA and then form functional inflammasome to trigger pyroptosis
and IL18 and IL1βmediated inflammatory responses (Man, Karki, &
Kanneganti, 2016b). AIM2 detects dsDNA in a DNA sequence
independent manner, allowing its response to various microbial
threats and cellular stress. Indeed, AIM2 was shown to contributed
to provide immunosurveillance to microbial pathogens from bacteria
(like Francisella, Listeria, and Mycobacterium), DNA viruses (like
cytomegalovirus, vaccinia virus, and human papillomaviruses), fungi (like
Aspergillus fumigatus), and protozoa (like Plasmodium berghei; Man,
Karki, & Kanneganti, 2016a). These evidence highlighted the crucial
role of AIM2 inflammasome for hostdefense against cytosolic
pathogenic DNA. However, it occurs under certain circumstances
where host cell fails to distinguish host selfDNA from pathogenic
nucleic acids, resulting in upregulated AIM2dependent release of IL
1βthat contributes to the pathogenesis of inflammatory diseases.
Studies have found abundant cytosolic selfDNA and increased AIM2
expression in patients with chronic and acute skin conditions,
including contact dermatitis, psoriasis, and atopic dermatitis (de
Koning et al., 2012). Except the pivotal function of NLRP1 and NLRP3
inflammasome in TBI, recent studies suggested that AIM2 might
contribute to the pathogenesis of TBI. It was indicated that AIM2 was
associated with the pyroptotic cell death induced caspase1in
response to aberrant dsDNA in cortical neurons. It was also observed
that CSF samples from TBI patients contained considerably higher
levels of nucleic acids than CSF from control patients, and when
neurons were cocultured with CSF from TBI patients, AIM2
inflammasome activation emerged (Adamczak et al., 2014). However,
studies focusing on the function of AIM2 in CNS injury are limited. In
addition, IFI16 inflammasome has been described in HIV1 infected
CD4
+
T cells and DNA virus Kaposi's sarcomaassociated herpesvirus
(KSHV) infected endothelial cells (Monroe et al., 2014). KSHV causes
an asymptomatic latent infection in normal individual, whereas it can
lead to Kaposi's sarcoma in immunocompromised patients, which
features an inflammatory microenvironment including IL1β(Ganem,
2006). Kerur et al. (2011) demonstrated that IFI16 acted as a nuclear
KSHV dsDNA sensor to induce the formation of inflammasome which
then translocates from nucleus to cytosol to process proIL1β.Of
note, studies recently have made a breakthrough on the mechanism
underlying CD4 Tcell death in HIVinfected hosts. Doitsh et al.
(2014) suggested that the progressive depletion of more than 95% of
quiescent lymphoid CD4 Tcells, abortively infected with HIV, die by
pyroptosis instead of apoptosis. Further studies identified that IFI16
was responsible for the detection of HIV1 dsDNA and consequently
resulted in pyroptosis of CD4
+
T cells and secretion of inflammatory
cell signaling molecule IL1β, which recruits more cells to die of
pyroptosis. This creates a vicious pathogenic cycle, contributing to
HIV pathogenesis (Monroe et al., 2014). To sum up, these evidence
indicate a novel and potential therapy for AIDS targeting the
pathway of host CD4
+
T cells pyroptosis rather than the virus.
7
|
PYRIN INFLAMMASOME
Pyrin (Marenostrin, TRIM 20), encoded by the human MEFV gene or
mouse analog Mefv, expresses mainly in neutrophils, monocytes and
dendritic (Seshadri, Duncan, Hart, Gavrilin, & Wewers, 2007). There
are four functional domains in human Pyrin, a Nterminal pyrin
domain (PYD), two central zinc finger domain (bBox), a coiled coil
(CC) domain and a Cterminal B30.2/SPRY domain. And mouse pyrin
lacks a B30.2/SPRY domain (Yu et al., 2006). The Bbox domain
interacts with the PYD to sustain Pyrin in an autoinhibited state and
the CC domain mediates selfoligomerization of Pyrin into a trimer.
Thus, Pyrin exists as an inactive homotrimer in resting cells (Yu et al.,
2007). The PYD in Pyrin, which is also found in other known sensors
like NLRP1, NLRP3, and AIM2, mediates homotypic interaction with
ASC to assemble procaspase1 and form functional inflammasome
(Vajjhala et al., 2014). Despite the wellestablished Pyrin inflamma-
some in vitro, the triggers and function of Pyrin in innate immunity
response remained obscure until a recent study elucidated that Pyrin
mediated inflammasome activation in response to pathogen
mediated modifications of Rho GTPases. And the modifications were
caused by effector proteins or pathogenic toxins, like FICdomain
adenylylation by VopS of Vibrio parahemolyticus, FICdomain adeny-
lylation by IbpA of Histophilus somni, Rhoglycosylation by cytotoxin
TcdB of Clostridium difficile, deamination by Burkholderia cenocepacia,
and ADPribosylation by C. botulinum C3 toxin and.These modifica-
tions all resided in SwitchI region of Rho GTPases, and could lead to
the inactivation of the Rho GTPase. Pyrin functioned to recognize the
inactivation of Rho GTPases and thereby triggered Pyrin inflamma-
some activation (Xu et al., 2014). Interestingly, Pyrin seems not to
bind modified Rho GTPases directly, indicating a potential down-
stream signaling event. On topic of this, Park, Wood, Kastner, and
Chae (2016) recently showed that in the resting state, Rho GTPases
activated the serinethreonine kinases PKN1 and PKN2 that bound
and phosphorylate human Pyrin at S208 and S242. Phosphorylated
Pyrin was combined with1433 proteins, which maintained Pyrin
inflammasome in an inactive state (Park et al., 2016). When Rho
GTPases was modified by bacterial toxins or effector proteins, Pyrin
dephosphorylated and 1433 proteins dissociated, resulting in a
conformational change that allowed Pyrin inflammasome activation.
Yet, it remains obscure how Pyrin is dephosphorylated and how 14
33 proteins are released (Gao, Yang, Liu, Wang, & Shao, 2016).
Familial Mediteranean fever (FMF) is an autosomal recessive
autoinflammatory disease featured by attacks of fever and neutro-
philmediated serosal inflammation, which was caused by mutations
of MEFV gene that encodes Pyrin (Centola, Aksentijevich, & Kastner,
8
|
LU ET AL.
1998). In particular, two thirds of such mutations are located in the
B30.2/SPRY domain (Chae et al., 2011). Such missense mutations
effect in FMF was due to gainoffunction of Pyrin, which caused
increased caspase1 activation and inflammatory responses (Chae
et al., 2011). In addition, A newly identified disease, Pyrinassociated
autoinflammation with neutrophilic dermatosis (PAAND), is also
caused by mutations of MEFV gene, but it is distinct from FMF on
molecular mechanism. The mutation results in serinetoarginine
replacement at position 242 (S242R) in pyrin and thereby eliminates
1433 binding motif (Masters et al., 2016). Further studies
conducted by Moghaddas et al. (2017) reported a novel E224K
MEFV mutation that was located in the +2 position of the 1433
binding motif in pyrin. Consistent with S242R mutation in Pyrin,
E224K in Pyrin could also induce spontaneous inflammasome
formation and occurrence of pyroptosis (Moghaddas et al., 2017).
In addition, a recent study identified that hyperimmunoglobulinemia
D syndrome (HIDS) was dependent on a dysregulation of pyrin
activation, which was caused by mutations of mevalonate kinase
(MVK). Lossoffunction mutations of MVK reduced prenylation of
proteins such as RhoGTPase, causing inactivation of RhoGTPase
and consequent activation of Pyrin inflammasome (Park et al., 2016).
Furthermore, Pyrin protein was also involved in syndrome of
pyogenic arthritis, pyoderma gangrenosum and acne (PAPA), an
inherited autoinflammatory disorder caused by prolineserinethreo-
nine phosphataseinteracting protein1 (PSTPIP1) mutations (Wise
et al., 2002). The PAPAassociated mutation was shown to increase
the binding of PSTPIP1 to Bbox of pyrin.Such binding unmasked
PYD of Pyrin protein, which allowed Pyrin to interact with ASC and
recruit procaspase1 to form Pyrin inflammasome (Yu et al., 2007).
Intriguingly, these evidence indicated a possibility that PSTPIP1
might be a downstream protein of RhoGTPase modification and more
studies are required to focus on this topic.
8
|
NONCANONICAL INFLAMMASOME
The functional significance of caspase11 in the process of pyroptosis
was masked for a long time, until Kayagaki et al. (2011) discovered
that proinflammatory caspase11 could trigger pyroptosis and IL1β
maturation in a Nlrp3Asccaspase1 inflammasome dependent
pathway in LPSprimed mouse macrophages. Shi et al. (2014) found
that human caspase4/5 (homologs to murine caspase11) could also
be activated by direct binding to intracellular LPS in human
monocytes and nonmonocytes, like certain epithelial cells and
keratinocytes, which led to pyroptosis. This caspase4/5/11LPS
complex was classified as the noncanonical inflammasome (Ding &
Shao, 2017).
Mounting studies emphasized the crucial role of Gramnegative
bacterium derived LPS in caspase4/5/11dependent pyroptosis (Yi,
2017). LPS of extracellular Gramnegative bacterium was sensed by
transmembrane Tolllike receptor 4 (TLR4). TLR4 activated MyD88/
TRIF/NFkB pathway and IFNR/JAK/STAT pathway to regulate the
transcription of caspase4/5/11, which was essential for initiating the
noncanonical inflammasome pathway (Yang et al., 2015). Caspase
11/4/5 acts as LPS sensors and only recognizes LPS that has entered
the cytoplasm (Hagar, Powell, Aachoui, Ernst, & Miao, 2013).
Howbeit extracellular Gramnegative bacteria or internalized bacter-
ia within membranebound vacuoles are believed to seldom get
access into cytoplasm, so exposure of bacterial LPS to cytoplasm is a
vital event for pathogen recognition. Vanaja et al. (2016) showed that
Gramnegative bacteria produced outer membrane vesicles (OMVs)
that entered cells by endocytosis and the early endosomes could
release LPS into the cytoplasm. Recently, it was further identified
that hemolysin could combine with OMVs and promoted rupture of
OMVcontaining vacuoles in the cytoplasm, enabling OMVderived
LPS to access the cytoplasm (Chen et al., 2018). And for vacuole
residing Gramnegative bacteria, studies suggested that guanylate
binding proteins (GBP) lysed vacuoles and thus exposed bacteria to
caspase11 in the cytoplasm (Meunier et al., 2014). The CARD in the
caspase4/5/11 could directly interact with the lipid A of cytosolic
LPS, leading to activation of the caspases (Hagar et al., 2013). The
active caspase4/5/11 then induced pyroptosis by cleaving GSDMD
into GSDMDNT (Kayagaki et al., 2015; Shi et al., 2015). Interestingly,
active caspase4/5/11 promotes the maturation of proIL18 or pro
IL1βthrough stimulating the Nlrp3Asccaspase1 noncanonical
inflammasome pathway instead of direct interaction (Baker et al.,
2015). However, the underlying mechanisms by which caspase11
induces Nlrp3Asccaspase1 activation have not been welldefined.
Several studies proposed that potassium outflow probably could
bridge caspase11 and NLRP3 activation based on the discovery that
activated LPScaspase11 could cleave the pannexin1 channel to
induce release of ATP, which activated purinergic receptor P2X7 to
open a larger reversible pore that allowed the efflux of potassium.
And the efflux of potassium was considered as one of the agonists of
NLRP3 inflammasome in the canonical inflammasome pathway (Ruhl
& Broz, 2015). However, the functional crosstalk between non-
canonical and canonical inflammasome signaling cascades still needs
further study. Furthermore, saturated fatty acids and nuclear DAMPs
(nDAMPs) induce lipid peroxidation were reported to trigger
caspase4/5 and caspase11 mediated pyroptosis respectively in
vitro (Chen et al., 2019; Pillon et al., 2016). Whether the
noncanonical inflammasome of cell pyroptosis could be activated
by other molecules except above agonists also remains to be
investigated.
Pyroptosis mediated by caspase11 is believed to contribute to
the clearance of cytosolic bacterial pathogens in vivo, such as
Salmonella and Burkholderia.Salmonella normally resides in the
vacuole. Specific mutant strains of S. typhimurium escaped from the
vacuole and were recognized by caspase11, and caspase11 induced
downstream signaling was shown to enhance clearance of the S.
typhimurium in vivo (Aachoui et al., 2013). Besides, caspase11
/
mice
were susceptible to Burkholderia thailandensis and Burkholderia
pseudomallei, which naturally invade the cytosol, whereas wildtype
mice were resistant to the infection (Jorgensen & Miao, 2015).
Despite that caspase11 mediated pyroptosis provides protection
against pathogen by releasing potentially harmful bacteria from the
LU ET AL.
|
9
nutrientrich host cell cytosol and through secreted inflammatory
cytokine induced inflammatory responses, dysregulated caspase11
mediated pyroptosis during overwhelming infection contributes to
sepsis. Sepsis is defined as lifethreatening organ dysfunction caused
by a dysregulated host inflammatory response to infection (Singer
et al., 2016). LPS has been known as an essential stimulus of sepsis
(Sweet & Hume, 1996). Studies recently proposed caspase4/5/11 as
a sensor of cytoplasmic LPS, suggesting that caspase4/5/11
mediated noncanonical inflammasome pathway of pyroptosis might
play a pivotal role in regulating sepsis (Shi et al., 2014). Further
exploration confirmed this idea with the evidence that wildtype mice
were more vulnerable to LPS than caspase11
/
mice when
challenged with the Gramnegative bacteria (Hagar et al., 2013). In
addition, Kayagaki et al. (2015) reported that Gsdmd
/
mice were
protected from a lethal dose of LPS and highlighted that key role of
GSDMD in LPSinduced septic shock. The resistance of mice lacking
GSDMD or caspase11 to LPSinduced lethality inspires the
researchers to attempt to develop antagonists of human caspase4/
5 or GSDMD for the benefit patients suffering from Gramnegative
bacteriainduced sepsis. Moreover, based on the beneficial and
detrimental effects of caspase4/5/11 mediated pyroptosis, how to
weigh the delicate balance between combating infection and sepsis is
a question to be resolved in the future.
9
|
Gasdermin D (GSDMD)
The underlying molecular mechanism downstream of caspase1/4/5/
11 to mediate pyroptosis had remained undefined, until recently two
separated research groups, Vishva M Dixit and Feng Shao,
independently identified GSDMD (gasdermin D) as an essential
pyroptosis substrate of inflammatory caspases (Kayagaki et al., 2015;
Shi et al., 2015). Both group further generated mouse strains with a
genomic deletion of gasdermin D (Gsdmd
/
) to confirm its crucial
role in pyroptosis and found that Gsdmd
/
iBMDMs did not undergo
pyroptosis or secrete mature inflammatory cytokines (Kayagaki et al.,
2015; Shi et al., 2015). Shi et al. (2015) also found GSDMD deficiency
inhabit IL1βsecretion without influencing its cleavage by caspase1,
suggesting GSDMD functioning downstream of caspase. Moreover, it
was showed that after the assembly of canonical or noncanonical
inflammasome, the activated inflammatory caspases (caspase1/4/5/
11) cleaved the 53kDa GSDMD after Asp276 (mouse)/275 (human),
generating the gasderminC terminus (GSDMDCT,22 kDa) and
gasderminN terminus (GSDMDNT, 31 kDa). Of note, it was
GSDMDNT rather than fulllength GSDMD or GSDMDCT that
bore intrinsic pyroptosisinducing activity, based on the evidence
that when GSDMDCT, GSDMDNT, and fulllength GSDMD were
individually expressed in 293T cells, pyroptosis only occurred in
GSDMDNT group. Likewise, ectopic overexpression of GSDMDNT,
but not GSDMDCT or fulllength GSDMD, rapidly triggered
extensive cell death with pyroptosis morphology. As for the function
of GSDMDCT, it was indicated that GSDMDCT bound to GSDMD
NT in resting cells to maintain GSDMD protein in an autoinhibited
state (Kayagaki et al., 2015; Shi et al., 2015). Recently, Kuang et al.
(2017) reported the crystal structure of the GSDMDCT and
revealed that the first loop on GSDMDCT inserted into the N
terminal domain to help stabilize the fulllength GSDMD conforma-
tion. Such autoinhibited conformation could be broken upon
interdomain cleavage by inflammatory caspases. Once the auto
inhibition state is released, the positive potential surface of GSDMD
NT covered by GSDMDCT is exposed and then forms highorder
oligomers via a chargecharge interaction to trigger the pyroptosis
(Kuang et al., 2017; Shi et al., 2015). Thus, it further confirmed that
GSDMDNT was the key mediator of pyroptosis, which was
characterized by pore formation in morphology. This raised a
prediction that GSDMDNT either possessed poreforming activity
or promoted the formation of this pore (Broz, 2015). Through further
exploration on this topic, studies showed that the GSDMDNT
possessed lipidbinding preferences and could specifically bind to
phosphatidylserine and phosphatidylinositol phosphates, which were
constrained to the inner leaflet of cell membrane, as well as
cardiolipin in the inner and outer leaflets of cell membranes (Ding
et al., 2016; Liu et al., 2016). Indeed, GSDMDNT formed 16mer
pore complex on the plasma membrane, which disrupted cell
membrane integrity. Then, cell pyroptosis and secretion of cytosolic
contents emerged (Ding et al., 2016). In addition, researchers
visualized GSDMDNT oligomers on PScontaining liposomes by
using negative staining electron microscopy and observed that pores
in the plasma membrane were approximately 15 nm inner and 32 nm
outer diameters, which were larger than processed IL1βwith a
diameter of approximately 4.5 nm. These pores precisely explain the
release of IL18 and IL1βduring pyroptosis as these inflammatory
cytokines lack an Nterminal signal peptide for endoplasmic
reticulum to Golgi secretion pathway (Liu et al., 2016). Interestingly,
the results from vitro experiments observed that GSDMDNT could
directly interact with cardiolipin on bacterial membranes and killed
them, suggesting a possible antibacterial role of GSDMDNT.
However, more experiments are required to distinguish such direct
killing of bacterium by GSDMDNT from killing of the GSDMD
induced host pyroptosis, and explain how GSDMDNT protein passes
through the thick cell wall to access inner membrane (Liu et al.,
2016).
GSDMD belongs to a gasdermin family that also includes
GSDMA, GSDMB, GSDMC, GSDME/DFNA5, and DFNB59. All
gasdermins (except for DFNB59) adopt GSDMDlike autoinhibited
twodomain architecture: gasderminN and C domains. Recently,
studies showed gasderminN domains of all gasdermins could induce
mammalian cell pyroptosis and kill bacteria. However, except
GSDMD, other gasdermins are insensitive to inflammatory caspases
(Ding et al., 2016; Shi et al., 2017; Shi et al., 2015). Notably, there are
some recent evidence suggesting a role of GSDME in pyroptosis. It
was showed that GSDME could switch caspase3 dependent
chemotherapy druginduced apoptosis into pyroptosis. The shift
from apoptosis to pyroptosis relied on the expression or expression
level of GSDME. Upon processing chemotherapy drug, GSDME high
expression cells undergo pyroptosis; otherwise, undergo apoptosis
10
|
LU ET AL.
(Wang et al., 2017; Wang et al., 2018). In general, GSDME was
silenced in most cancer cells and expressed in many normal tissues.
Studies revealed that GSDME mediated pyroptosis in normal tissues
contributed to the chemotherapy druginduced toxicity, like tissue
damage and weight loss, while in GSDMEexpressed cancer cells, like
gastric cancer cells, GSDME mediated pyroptosis contributed to the
chemotherapy (Wang et al., 2017, 2018). These results break the
traditional concept that activation of caspase3 inevitably induce
apoptosis. More important, in addition to exploration of inflamma-
some inhibitors for future treatment of cancer (Xu et al., 2019), these
data highlighted an innovative and promising therapeutic approach
against cancer by targeting GSDME.
10
|
CONCLUTION AND PERSPECTIVES
Recent advances have now established the key events during process of
pyroptosis, which is inflammasome activation, inflammatory caspases
maturation, and GSDMD cleavage. A wide range of host and microbial
factors trigger inflammasome activation and induce a series of caspase
dependent signaling events, including proinflammatory cytokine ma-
turation, pyroptosis, and inflammatory responses. This caspase1
mediated canonical pathway of pyroptosis is paramount in hostdefense
against intracellular pathogens. Pyroptosis of infected cells could
remove intracellular replicative niche of the bacteria to extracellular
matrix and the released cytokine could recruit more immune cells to
amplify inflammation in situ. The discovery of LPS triggered caspase11/
4/5 mediated pyroptosis has expanded the understanding of signaling
pathway leading to pyroptosis. This newly discovered noncanonical
pathway is essential in hostdefense against pathogenic infection and is
responsible for the dysregulated inflammatory response, sepsis.
Importantly, GSDMD has been identified as substrate of inflammatory
caspases and poreforming activity of GSDMDN T is responsible for
pyroptosis execution.
Recent studies have clarified a number of mysteries that were
prevalent in the activators of canonical inflammasomes and noncanonical
inflammasome. However, whether these inflammasomes can also be
activated by other PAMPs/DAMPs is still uncertain, and the detailed
mechanisms governing the activation process above are still controver-
sial. Newly uncovered GSDMD opens up new avenues to specifically
suppress pyroptosis in health and disease. However, researchers have not
reached a consensus on the downstream molecular mechanisms involved
in GSDMDmediated cytotoxic pores, and the functions of other GSDMs
in pyroptotic process are still required further investigation. As reviewed
above, pyroptosis appears to be a doubleedged sword. Based on the
pathogenic and protective effects of pyroptosis, further exploration on its
therapeutic inhibition to control the excessive inflammation has to be
balanced against its beneficial contribution to infectious diseases.
ACKNOWLEGMENTS
This study is supported by National Natural Science Foundation of
China (no. 81900890), China Postdoctoral Science Foundation (no.
2018M643004), and the College StudentsInnovative Project of CSU
(no. ZY2016698).
AUTHOR CONTRIBUTIONS
F. L. wrote the article. C. H. and Z. G. edited the article and Z. X. and
Z. L. draw the graphical illustrations. T. H. designed the work,
developed the idea, and supervised the writing process.
ETHICAL STATEMENT
Fangfang Lu, Zhixin Lan, Zhaoqi Xin, Chunrong He, Zimeng Guo, and
Tu Hu declare that they have no conflict of interests. This article does
not contain any studies with human or animal subjects performed by
the any of the author
ORCID
Fangfang Lu http://orcid.org/0000-0002-9214-3046
REFERENCES
Aachoui, Y., Leaf, I. A., Hagar, J. A., Fontana, M. F., Campos, C. G., Zak, D.
E., Miao, E. A. (2013). Caspase11 protects against bacteria that
escape the vacuole. Science,339(6122), 975978. https://doi.org/10.
1126/science.1230751
Aachoui, Y., Sagulenko, V., Miao, E. A., & Stacey, K. J. (2013).
Inflammasomemediated pyroptotic and apoptotic cell death, and
defense against infection. Current Opinion in Microbiology,16(3),
319326. https://doi.org/10.1016/j.mib.2013.04.004
Adamczak, S., Dale, G., de Rivero Vaccari, J. P., Bullock, M. R., Dietrich, W.
D., & Keane, R. W. (2012). Inflammasome proteins in cerebrospinal
fluid of braininjured patients as biomarkers of functional outcome:
Clinical article. Journal of Neurosurgery,117(6), 11191125. https://
doi.org/10.3171/2012.9.JNS12815
Adamczak,S.E.,deRiveroVaccari,J.P.,Dale,G.,Brand,F.J.,3rd,Nonner,D.,
Bullock, M., Keane, R. W. (2014). Pyroptotic neuronal cell death
mediated by the AIM2 inflammasome. Journal of Cerebral Blood Flow and
Metabolism,34(4), 621629. https://doi.org/10.1038/jcbfm.2013.236
Agostini, L., Martinon, F., Burns, K., McDermott, M. F., Hawkins, P. N., &
Tschopp, J. (2004). NALP3 forms an IL1βprocessing inflammasome
with increased activity in mucklewells autoinflammatory disorder.
Immunity,20(3), 319325.
Baker, P. J., Boucher, D., Bierschenk, D., Tebartz, C., Whitney, P. G.,
D'Silva, D. B., Masters, S. L. (2015). NLRP3 inflammasome activation
downstream of cytoplasmic LPS recognition by both caspase4 and
caspase5. European Journal of Immunology,45(10), 29182926.
https://doi.org/10.1002/eji.201545655
Bauernfeind, F. G., Horvath, G., Stutz, A., Alnemri, E. S., MacDonald, K.,
Speert, D., Latz, E. (2009). Cutting edge: NFκB activating pattern
recognition and cytokine receptors license NLRP3 inflammasome
activation by regulating NLRP3 expression. Journal of Immunology,
183(2), 787791. https://doi.org/10.4049/jimmunol.0901363
Bergsbaken, T., Fink, S. L., & Cookson, B. T. (2009). Pyroptosis: Host cell
death and inflammation. Nature Reviews Microbiology,7(2), 99109.
https://doi.org/10.1038/nrmicro2070
Borgens, R. B., & LiuSnyder, P. (2012). Understanding secondary injury.
The Quarterly Review of Biology,87(2), 89127.
LU ET AL.
|
11
Broz, P. (2015). Caspase target drives pyroptosis. Nature,526(7575),
642643. https://doi.org/10.1038/nature15632
Brunette,R.L.,Young,J.M.,Whitley,D.G.,Brodsky,I.E.,Malik,H.S.,&
Stetson, D. B. (2012). Extensive evolutionary and functional diversity
among mammalian AIM2like receptors. Journal of Experimetnal Medicine,
209(11), 19691983. https://doi.org/10.1084/jem.20121960
Bürckstümmer, T., Baumann, C., Blüml, S., Dixit, E., Dürnberger, G., Jahn,
H., SupertiFurga, G. (2009). An orthogonal proteomicgenomic
screen identifies AIM2 as a cytoplasmic DNA sensor for the
inflammasome. Nature Immunology,10(3), 266272. https://doi.org/
10.1038/ni.1702
Centola, M. (1998). The hereditary periodic fever syndromes: Molecular
analysis of a new family of inflammatory diseases. Human Molecular
Genetics,7(10), 15811588.
Chae, J. J., Cho, Y. H., Lee, G. S., Cheng, J., Liu, P. P., Feigenbaum, L.,
Kastner, D. L. (2011). Gainoffunction pyrin mutations induce NLRP3
proteinindependent interleukin1βactivation and severe autoinflam-
mation in mice. Immunity,34(5), 755768. https://doi.org/10.1016/j.
immuni.2011.02.020
ChavarríaSmith, J., Mitchell, P. S., Ho, A. M., Daugherty, M. D., & Vance, R.
E. (2016). Functional and evolutionary analyses identify proteolysis as
a general mechanism for NLRP1 inflammasome activation. PLOS
Pathogens,12(12), e1006052. https://doi.org/10.1371/journal.ppat.
1006052
ChavarríaSmith, J., & Vance, R. E. (2013). Direct proteolytic cleavage of
NLRP1B is necessary and sufficient for inflammasome activation by
anthrax lethal factor. PLOS Pathogens,9(6), e1003452. https://doi.org/
10.1371/journal.ppat.1003452
ChavarríaSmith, J., & Vance, R. E. (2015). The NLRP1 inflammasomes.
Immunological Reviews,265(1), 2234. https://doi.org/10.1111/imr.
12283
Chen, R., Zhu, S., Zeng, L., Wang, Q., Sheng, Y., Zhou, B., Kang, R. (2019).
AGERmediated lipid peroxidation drives caspase11 inflammasome
activation in sepsis. Frontiers in Immunology,10, 1904. https://doi.org/
10.3389/fimmu.2019.01904
Chen, S., Yang, D., Wen, Y., Jiang, Z., Zhang, L., Jiang, J., Liu, Q. (2018).
Dysregulated hemolysin liberates bacterial outer membrane vesicles
for cytosolic lipopolysaccharide sensing. PLOS Pathogens,14(8),
e1007240. https://doi.org/10.1371/journal.ppat.1007240
Choubey, D., Duan, X., Dickerson, E., Ponomareva, L., Panchanathan, R.,
Shen, H., & Srivastava, R. (2010). Interferoninducible p200family
proteins as novel sensors of cytoplasmic DNA: Role in inflammation
and autoimmunity. Journal of Interferon & Cytokine Research,30(6),
371380. https://doi.org/10.1089/jir.2009.0096
Cookson, B. T., & Brennan, M. A. (2001). Proinflammatory programmed
cell death. Trends in Microbiology,9(3), 113114.
Cummings, J. R. F., Cooney, R. M., Clarke, G., Beckly, J., Geremia, A.,
Pathan, S., Jewell, D. P. (2010). The genetics of NODlike receptors
in Crohn's disease. Tissue Antigens,76(1), 4856. https://doi.org/10.
1111/j.13990039.2010.01470.x
Ding, J., & Shao, F. (2017). SnapShot: The noncanonical inflammasome. Cell,
168(3), 544544. https://doi.org/10.1016/j.cell.2017.01.008. e541.
Ding, J., Wang, K., Liu, W., She, Y., Sun, Q., Shi, J., Shao, F. (2016). Pore
forming activity and structural autoinhibition of the gasdermin family.
Nature,535(7610), 111116. https://doi.org/10.1038/nature18590
Doitsh, G., Galloway, N. L. K., Geng, X., Yang, Z., Monroe, K. M., Zepeda, O.,
Greene, W. C. (2014). Cell death by pyroptosis drives CD4 Tcell
depletion in HIV1 infection. Nature,505(7484), 509514. https://doi.
org/10.1038/nature12940
Donath, M. Y., & Shoelson, S. E. (2011). Type 2 diabetes as an
inflammatory disease. Nature Reviews Immunology,11(2), 98107.
https://doi.org/10.1038/nri2925
Elliott, E. I., & Sutterwala, F. S. (2015). Initiation and perpetuation of
NLRP3 inflammasome activation and assembly. Immunological Reviews,
265(1), 3552. https://doi.org/10.1111/imr.12286
Elliott, J. M., Rouge, L., Wiesmann, C., & Scheer, J. M. (2009). Crystal
structure of procaspase1 zymogen domain reveals insight into
inflammatory caspase autoactivation. Journal of Biological Chemistry,
284(10), 65466553. https://doi.org/10.1074/jbc.M806121200
Ewald, S. E., ChavarriaSmith, J., & Boothroyd, J. C. (2014). NLRP1 is an
inflammasome sensor for Toxoplasma gondii.Infection and Immunity,
82(1), 460468. https://doi.org/10.1128/IAI.0117013
Faustin, B., Lartigue, L., Bruey, J. M., Luciano, F., Sergienko, E., Bailly
Maitre, B., Reed, J. C. (2007). Reconstituted NALP1 inflammasome
reveals twostep mechanism of caspase1 activation. Molecular Cell,
25(5), 713724. https://doi.org/10.1016/j.molcel.2007.01.032
FernandesAlnemri, T., Yu, J. W., Datta, P., Wu, J., & Alnemri, E. S. (2009).
AIM2 activates the inflammasome and cell death in response to
cytoplasmic DNA. Nature,458(7237), 509513. https://doi.org/10.
1038/nature07710
Finger, J. N., Lich, J. D., Dare, L. C., Cook, M. N., Brown, K. K., Duraiswami,
C., Gough, P. J. (2012). Autolytic proteolysis within the function to
find domain (FIIND) is required for NLRP1 inflammasome activity.
Journal of Biological Chemistry,287(30), 2503025037. https://doi.org/
10.1074/jbc.M112.378323
Galán, J. E., LaraTejero, M., Marlovits, T. C., & Wagner, S. (2014). Bacterial
type III secretion systems: Specialized nanomachines for protein
delivery into target cells. Annual Review of Microbiology,68, 415438.
https://doi.org/10.1146/annurevmicro092412155725
Ganem, D. (2006). KSHV infection and the pathogenesis of Kaposi's
sarcoma. Annual Review of Pathology: Mechanisms of Disease,1,
273296. https://doi.org/10.1146/annurev.pathol.1.110304.100133
Gao, W., Yang, J., Liu, W., Wang, Y., & Shao, F. (2016). Sitespecific
phosphorylation and microtubule dynamics control Pyrin inflamma-
some activation. Proceedings of the National Academy of Sciences of the
United States of America,113(33), E4857E4866. https://doi.org/10.
1073/pnas.1601700113
Grandemange, S., Sanchez, E., LouisPlence, P., Tran MauThem, F., Bessis,
D., Coubes, C., Geneviève, D. (2017). A new autoinflammatory and
autoimmune syndrome associated with NLRP1 mutations: NAIAD
(NLRP1associated autoinflammation with arthritis and dyskeratosis).
Annals of the Rheumatic Diseases,76(7), 11911198. https://doi.org/
10.1136/annrheumdis2016210021
Guarda, G., Braun, M., Staehli, F., Tardivel, A., Mattmann, C., Förster, I.,
Tschopp, J. (2011). Type I interferon inhibits interleukin1 production
and inflammasome activation. Immunity,34(2), 213223. https://doi.
org/10.1016/j.immuni.2011.02.006
Guo, H., Callaway, J. B., & Ting, J. P. Y. (2015). Inflammasomes: Mechanism
of action, role in disease, and therapeutics. Nature Medicine (New York,
NY, United States),21(7), 677687. https://doi.org/10.1038/nm.3893
Hagar, J. A., Powell, D. A., Aachoui, Y., Ernst, R. K., & Miao, E. A. (2013).
Cytoplasmic LPS activates caspase11: Implications in TLR4indepen-
dent endotoxic shock. Science,341(6151), 12501253. https://doi.org/
10.1126/science.1240988
Hanna, P. C., Acosta, D., & Collier, R. J. (1993). On the role of macrophages
in anthrax. Proceedings of the National Academy of Sciences,90(21),
1019810201.
He, Y., Hara, H., & Núñez, G. (2016). Mechanism and regulation of NLRP3
inflammasome activation. Trends in Biochemical Sciences,41(12),
10121021. https://doi.org/10.1016/j.tibs.2016.09.002
Hu, Z., Yan, C., Liu, P., Huang, Z., Ma, R., Zhang, C., Chai,J.(2013).Crystal
structure of NLRC4 reveals its autoinhibition mechanism. Science,
341(6142), 172175. https://doi.org/10.1126/science.1236381
Inoue, M., Williams, K. L., Gunn, M. D., & Shinohara, M. L. (2012). NLRP3
inflammasome induces chemotactic immune cell migration to the CNS
in experimental autoimmune encephalomyelitis. Proceedings of the
National Academy of Sciences,109(26), 1048010485. https://doi.org/
10.1073/pnas.1201836109
Jin, T., Perry, A., Jiang, J., Smith, P., Curry, J. A., Unterholzner, L., Xiao, T.
S. (2012). Structures of the HIN domain: DNA complexes reveal ligand
12
|
LU ET AL.
binding and activation mechanisms of the AIM2 inflammasome and
IFI16 receptor. Immunity,36(4), 561571. https://doi.org/10.1016/j.
immuni.2012.02.014
Jin, T., Perry, A., Smith, P., Jiang, J., & Xiao, T. S. (2013). Structure of the
absent in melanoma 2 (AIM2) pyrin domain provides insights into the
mechanisms of AIM2 autoinhibition and inflammasome assembly.
Journal of Biological Chemistry,288(19), 1322513235. https://doi.org/
10.1074/jbc.M113.468033
Jorgensen, I., & Miao, E. A. (2015). Pyroptotic cell death defends against
intracellular pathogens. Immunological Reviews,265(1), 130142.
https://doi.org/10.1111/imr.12287
Juliana, C., FernandesAlnemri, T., Kang, S., Farias, A., Qin, F., & Alnemri, E.
S. (2012). Nontranscriptional priming and deubiquitination regulate
NLRP3 inflammasome activation. Journal of Biological Chemistry,
287(43), 3661736622. https://doi.org/10.1074/jbc.M112.407130
Kayagaki, N., Stowe, I. B., Lee, B. L., ORourke, K., Anderson, K., Warming,
S., Dixit, V. M. (2015). Caspase11 cleaves gasdermin D for non
canonical inflammasome signalling. Nature,526(7575), 666671.
https://doi.org/10.1038/nature15541
Kayagaki, N., Warming, S., Lamkanfi, M., Walle, L. V., Louie, S., Dong, J.,
Dixit, V. M. (2011). Noncanonical inflammasome activation targets
caspase11. Nature,479(7371), 117121. https://doi.org/10.1038/
nature10558
Kerur, N., Veettil, M. V., SharmaWalia, N., Bottero, V., Sadagopan, S.,
Otageri, P., & Chandran, B. (2011). IFI16 acts as a nuclear pathogen
sensor to induce the inflammasome in response to Kaposi Sarcoma
associated herpesvirus infection. Cell Host & Microbe,9(5), 363375.
https://doi.org/10.1016/j.chom.2011.04.008
Kitamura, A., Sasaki, Y., Abe, T., Kano, H., & Yasutomo, K. (2014). An
inherited mutation in NLRC4 causes autoinflammation in human and
mice. Journal of Experimetnal Medicine,211(12), 23852396. https://
doi.org/10.1084/jem.20141091
Kofoed, E. M., & Vance, R. E. (2011). Innate immune recognition of
bacterial ligands by NAIPs determines inflammasome specificity.
Nature,477(7366), 592595. https://doi.org/10.1038/nature10394
Kofoed, E. M., & Vance, R. E. (2012). NAIPs: Building an innate immune
barrier against bacterial pathogens: NAIPs function as sensors that
initiate innate immunity by detection of bacterial proteins in the host
cell cytosol. BioEssays,34(7), 589598. https://doi.org/10.1002/bies.
201200013
de Koning, H. D., Bergboer, J. G. M., van den Bogaard, E. H., van Vlijmen
Willems, I. M. J. J., RodijkOlthuis, D., Simon, A., Schalkwijk, J.
(2012). Strong induction of AIM2 expression in human epidermis in
acute and chronic inflammatory skin conditions. Experimental Derma-
tology,21(12), 961964. https://doi.org/10.1111/exd.12037
Kuang, S., Zheng, J., Yang, H., Li, S., Duan, S., Shen, Y., Li, J. (2017).
Structure insight of GSDMD reveals the basis of GSDMD autoinhibi-
tion in cell pyroptosis. Proceedings of the National Academy of Sciences
of the United States of America,114(40), 1064210647. https://doi.org/
10.1073/pnas.1708194114
Lamkanfi, M., & Dixit, V. M. (2014). Mechanisms and functions of
inflammasomes. Cell,157(5), 10131022. https://doi.org/10.1016/j.
cell.2014.04.007
Lechtenberg, B. C., Mace, P. D., & Riedl, S. J. (2014). Structural
mechanisms in NLR inflammasome signaling. Current Opinion in
Structural Biology,29,1725. https://doi.org/10.1016/j.sbi.2014.08.
011
Liao, K. C., & Mogridge, J. (2013). Activation of the Nlrp1b inflammasome
by reduction of cytosolic ATP. Infection and Immunity,81(2), 570579.
https://doi.org/10.1128/IAI.0100312
Lin, W. P., Xiong, G. P., Lin, Q., Chen, X. W., Zhang, L. Q., Shi, J. X., Lin, J.
H. (2016). Heme oxygenase1 promotes neuron survival through
downregulation of neuronal NLRP1 expression after spinal cord
injury. Journal of Neuroinflammation,13(1), 52. https://doi.org/10.
1186/s129740160521y
Liu, H. D., Li, W., Chen, Z. R., Hu, Y. C., Zhang, D. D., Shen, W., Hang, C.
H. (2013). Expression of the NLRP3 inflammasome in cerebral cortex
after traumatic brain injury in a rat model. Neurochemical Research,
38(10), 20722083. https://doi.org/10.1007/s110640131115z
Liu, X., Zhang, Z., Ruan, J., Pan, Y., Magupalli, V. G., Wu, H., & Lieberman, J.
(2016). Inflammasomeactivated gasdermin D causes pyroptosis by
forming membrane pores. Nature,535(7610), 153158. https://doi.
org/10.1038/nature18629
Lu, A., Magupalli, V. G., Ruan, J., Yin, Q., Atianand, M. K., Vos, M. R.,
Egelman, E. H. (2014). Unified polymerization mechanism for the
assembly of ASCdependent inflammasomes. Cell,156(6), 11931206.
https://doi.org/10.1016/j.cell.2014.02.008
Man, S. M., Karki, R., & Kanneganti, T. D. (2016a). AIM2 inflammasome in
infection, cancer, and autoimmunity: Role in DNA sensing, inflamma-
tion, and innate immunity. European Journal of Immunology,46(2),
269280. https://doi.org/10.1002/eji.201545839
Man, S. M., Karki, R., & Kanneganti, T. D. (2016b). DNAsensing
inflammasomes: Regulation of bacterial host defense and the gut
microbiota. Pathogens and Disease,74(4), ftw028. https://doi.org/10.
1093/femspd/ftw028
Man, S. M., Karki, R., & Kanneganti, T. D. (2017). Molecular mechanisms
and functions of pyroptosis, inflammatory caspases and inflamma-
somes in infectious diseases. Immunological Reviews,277(1), 6175.
https://doi.org/10.1111/imr.12534
Martino, L., Holland, L., Christodoulou, E., Kunzelmann, S., Esposito, D., &
Rittinger, K. (2016). The biophysical characterisation and SAXS
analysis of human NLRP1 uncover a new level of complexity of NLR
proteins. PLOS One,11(10), e0164662. https://doi.org/10.1371/
journal.pone.0164662
Martinon, F., Burns, K., & Tschopp, J. (2002). The Inflammasome. Molecular
Cell,10(2), 417426.
Masters, S. L., Lagou, V., Jéru, I., Baker, P. J., Van Eyck, L., Parry, D. A.,
Liston, A. (2016). Familial autoinflammation with neutrophilic derma-
tosis reveals a regulatory mechanism of pyrin activation. Science
Translational Medicine,8(332), 332ra45332ra45. https://doi.org/10.
1126/scitranslmed.aaf1471
Matusiak, M., Van Opdenbosch, N., Vande Walle, L., Sirard, J. C.,
Kanneganti, T. D., & Lamkanfi, M. (2015). Flagellininduced NLRC4
phosphorylation primes the inflammasome for activation by NAIP5.
Proceedings of the National Academy of Sciences of the United States of
America,112(5), 15411546. https://doi.org/10.1073/pnas.
1417945112
Medzhitov, R. (2007). Recognition of microorganisms and activation of the
immune response. Nature,449(7164), 819826. https://doi.org/10.
1038/nature06246
Mehto, S., Jena, K. K., Nath, P., Chauhan, S., Kolapalli, S. P., Das, S. K.,
Chauhan, S. (2019). The Crohns disease risk factor IRGM limits
NLRP3 inflammasome activation by impeding its assembly and by
mediating its selective autophagy. Molecular Cell,73(3), 429445.
https://doi.org/10.1016/j.molcel.2018.11.018. e427.
Meunier, E., Dick, M. S., Dreier, R. F., Schürmann, N., Broz, D. K., Warming,
S., Broz, P. (2014). Caspase11 activation requires lysis of pathogen
containing vacuoles by IFNinduced GTPases. Nature,509(7500),
366370. https://doi.org/10.1038/nature13157
Miao, E. A., Leaf, I. A., Treuting, P. M., Mao, D. P., Dors, M., Sarkar, A.,
Aderem, A. (2010). Caspase1induced pyroptosis is an innate immune
effector mechanism against intracellular bacteria. Nature Immunology,
11(12), 11361142. https://doi.org/10.1038/ni.1960
Miao, E. A., Mao, D. P., Yudkovsky, N., Bonneau, R., Lorang, C. G., Warren,
S. E., Aderem, A. (2010). Innate immune detection of the type III
secretion apparatus through the NLRC4 inflammasome. Proceedings of
the National Academy of Sciences,107(7), 30763080. https://doi.org/
10.1073/pnas.0913087107
Mishra, B. B., Rathinam, V. A. K., Martens, G. W., Martinot, A. J., Kornfeld,
H., Fitzgerald, K. A., & Sassetti, C. M. (2013). Nitric oxide controls the
LU ET AL.
|
13
immunopathology of tuberculosis by inhibiting NLRP3 inflammaso-
medependent processing of IL1β.Nature Immunology,14(1), 5260.
https://doi.org/10.1038/ni.2474
Moayeri, M., Sastalla, I., & Leppla, S. H. (2012). Anthrax and the
inflammasome. Microbes and Infection,14(5), 392400. https://doi.
org/10.1016/j.micinf.2011.12.005
Moghaddas, F., Llamas, R., De Nardo, D., MartinezBanaclocha, H.,
MartinezGarcia, J. J., MesaDelCastillo, P., Masters, S. L. (2017).
A novel pyrinassociated autoinflammation with neutrophilic derma-
tosis mutation further defines 1433 binding of pyrin and distinction
to Familial Mediterranean Fever. Annals of the Rheumatic Diseases,
76(12), 20852094. https://doi.org/10.1136/annrheumdis2017
211473
Monroe, K. M., Yang, Z., Johnson, J. R., Geng, X., Doitsh, G., Krogan, N. J., &
Greene, W. C. (2014). IFI16 DNA sensor is required for death of
lymphoid CD4 T cells abortively infected with HIV. Science,
343(6169), 428432. https://doi.org/10.1126/science.1243640
Mortezaee, K., Khanlarkhani, N., Beyer, C., & Zendedel, A. (2018).
Inflammasome: Its role in traumatic brain and spinal cord injury.
Journal of Cellular Physiology,233(7), 51605169. https://doi.org/10.
1002/jcp.26287
Murphy, A. J., Kraakman, M. J., Kammoun, H. L., Dragoljevic, D., Lee, M. K.
S., Lawlor, K. E., Masters, S. L. (2016). IL18 production from the
NLRP1 inflammasome prevents obesity and metabolic syndrome. Cell
Metabolism,23(1), 155164. https://doi.org/10.1016/j.cmet.2015.09.
024
Nabar, N. R., & Kehrl, J. H. (2019). Inflammasome Inhibition Links IRGM to
Innate Immunity. Molecular Cell,73(3), 391392. https://doi.org/10.
1016/j.molcel.2019.01.029
Nordlander, S., Pott, J., & Maloy, K. J. (2014). NLRC4 expression in
intestinal epithelial cells mediates protection against an enteric
pathogen. Mucosal Immunology,7(4), 775785. https://doi.org/10.
1038/mi.2013.95
Nour, A. M., Yeung, Y. G., Santambrogio, L., Boyden, E. D., Stanley, E. R., &
Brojatsch, J. (2009). Anthrax lethal toxin triggers the formation of a
membraneassociated inflammasome complex in murine macro-
phages. Infection and Immunity,77(3), 12621271. https://doi.org/10.
1128/IAI.0103208
Van Opdenbosch, N., Gurung, P., Vande Walle, L., Fossoul, A., Kanneganti,
T. D., & Lamkanfi, M. (2014). Activation of the NLRP1b inflammasome
independently of ASCmediated caspase1 autoproteolysis and speck
formation. Nature Communications,5, 3209. https://doi.org/10.1038/
ncomms4209
Park, Y. H., Wood, G., Kastner, D. L., & Chae, J. J. (2016). Pyrin
inflammasome activation and RhoA signaling in the autoinflammatory
diseases FMF and HIDS. Nature Immunology,17(8), 914921. https://
doi.org/10.1038/ni.3457
Pillon, N. J., Chan, K. L., Zhang, S., Mejdani, M., Jacobson, M. R., Ducos, A.,
Klip, A. (2016). Saturated fatty acids activate caspase4/5 in human
monocytes, triggering IL1βand IL18 release. American Journal of
PhysiologyEndocrinology and Metabolism,311(5), E825E835. https://
doi.org/10.1152/ajpendo.00296.2016
Pontillo, A., Girardelli, M., Kamada, A. J., Pancotto, J. A., Donadi, E. A.,
Crovella, S., & SandrinGarcia, P. (2012). Polimorphisms in inflamma-
some genes are involved in the predisposition to systemic lupus
erythematosus. Autoimmunity,45(4), 271278. https://doi.org/10.
3109/08916934.2011.637532
Poyet, J. L., Srinivasula, S. M., Tnani, M., Razmara, M., FernandesAlnemri,
T., & Alnemri, E. S. (2001). Identification of Ipaf, a human caspase1
activating protein related to Apaf1. Journal of Biological Chemistry,
276(30), 2830928313. https://doi.org/10.1074/jbc.C100250200
Proell, M., Gerlic, M., Mace, P. D., Reed, J. C., & Riedl, S. J. (2013). The
CARD plays a critical role in ASC foci formation and inflammasome
signalling. Biochemical Journal,449(3), 613621. https://doi.org/10.
1042/BJ20121198
Py, B. F., Kim, M. S., VakifahmetogluNorberg, H., & Yuan, J. (2013).
Deubiquitination of NLRP3 by BRCC3 critically regulates inflamma-
some activity. Molecular Cell,49(2), 331338. https://doi.org/10.1016/
j.molcel.2012.11.009
Quintiliani, R., Jr., & Quintiliani, R. (2003). Inhalational anthrax and
bioterrorism. Current Opinion in Pulmonary Medicine,9(3), 221226.
Reyes Ruiz, V. M., Ramirez, J., Naseer, N., Palacio, N. M., Siddarthan, I. J.,
Yan, B. M., Shin, S. (2017). Broad detection of bacterial type III
secretion system and flagellin proteins by the human NAIP/NLRC4
inflammasome. Proceedings of the National Academy of Sciences of the
United States of America,114(50), 1324213247. https://doi.org/10.
1073/pnas.1710433114
de Rivero Vaccari, J. P., Dietrich, W. D., & Keane, R. W. (2014). Activation
and regulation of cellular inflammasomes: Gaps in our knowledge for
central nervous system injury. Journal of Cerebral Blood Flow and
Metabolism,34(3), 369375. https://doi.org/10.1038/jcbfm.2013.227
Romberg, N., Vogel, T. P., & Canna, S. W. (2017). NLRC4 inflammasomo-
pathies. Current Opinion in Allergy and Clinical Immunology,17(6),
398404. https://doi.org/10.1097/ACI.0000000000000396
Ruhl, S., & Broz, P. (2015). Caspase11 activates a canonical NLRP3
inflammasome by promoting K(+) efflux. European Journal of Immunol-
ogy,45(10), 29272936. https://doi.org/10.1002/eji.201545772
Saresella, M., La Rosa, F., Piancone, F., Zoppis, M., Marventano, I.,
Calabrese, E., Clerici, M. (2016). The NLRP3 and NLRP1 inflamma-
somes are activated in Alzheimer's disease. Molecular Neurodegenera-
tion,11, 23. https://doi.org/10.1186/s1302401600881
Seshadri, S., Duncan, M. D., Hart, J. M., Gavrilin, M. A., & Wewers, M. D.
(2007). Pyrin levels in human monocytes and monocytederived
macrophages regulate IL1beta processing and release. Journal of
Immunology,179(2), 12741281.
Sharma, D., & Kanneganti, T. D. (2016). The cell biology of inflammasomes:
Mechanisms of inflammasome activation and regulation. Journal of Cell
Biology,213(6), 617629. https://doi.org/10.1083/jcb.201602089
Shi, J., Gao, W., & Shao, F. (2017). Pyroptosis: Gasderminmediated
programmed necrotic cell death. Trends in Biochemical Sciences,42(4),
245254. https://doi.org/10.1016/j.tibs.2016.10.004
Shi, J., Zhao, Y., Wang, K., Shi, X., Wang, Y., Huang, H., Shao, F. (2015).
Cleavage of GSDMD by inflammatory caspases determines pyroptotic
cell death. Nature,526(7575), 660665. https://doi.org/10.1038/
nature15514
Shi, J., Zhao, Y., Wang, Y., Gao, W., Ding, J., Li, P., Shao, F. (2014).
Inflammatory caspases are innate immune receptors for intracellular
LPS. Nature,514(7521), 187192. https://doi.org/10.1038/
nature13683
Singer, M., Deutschman, C. S., Seymour, C. W., ShankarHari, M., Annane,
D., Bauer, M., Angus, D. C. (2016). The Third International
Consensus Definitions for Sepsis and Septic Shock (Sepsis3). Journal
of the American Medical Association,315(8), 801810. https://doi.org/
10.1001/jama.2016.0287
Soares, J. L. S., Fernandes, F. P., Patente, T. A., Monteiro, M. B., Parisi, M.
C., GiannellaNeto, D., Pontillo, A. (2018). Gainoffunction variants
in NLRP1 protect against the development of diabetic kidney disease:
NLRP1 inflammasome role in metabolic stress sensing? Clinical
Immunology,187,4649. https://doi.org/10.1016/j.clim.2017.10.003
Sutterwala, F. S., Haasken, S., & Cassel, S. L. (2014). Mechanism of NLRP3
inflammasome activation. Annals of the New York Academy of Sciences,
1319,8295. https://doi.org/10.1111/nyas.12458
Sweet, M. J., & Hume, D. A. (1996). Endotoxin signal transduction in
macrophages. Journal of Leukocyte Biology,60(1), 826.
Takeuchi, O., & Akira, S. (2010). Pattern recognition receptors and
inflammation. Cell,140(6), 805820. https://doi.org/10.1016/j.cell.
2010.01.022
Tang, Y. S., Zhao, Y. H., Zhong, Y., Li, X. Z., Pu, J. X., Luo, Y. C., & Zhou, Q. L.
(2019). Neferine inhibits LPSATPinduced endothelial cell pyroptosis
via regulation of ROS/NLRP3/Caspase1 signaling pathway.
14
|
LU ET AL.
Inflammation Research,68(9), 727738. https://doi.org/10.1007/
s00011019012566
Tenthorey, J. L., Kofoed, E. M., Daugherty, M. D., Malik, H. S., & Vance, R.
E. (2014). Molecular basis for specific recognition of bacterial ligands
by NAIP/NLRC4 inflammasomes. Molecular Cell,54(1), 1729. https://
doi.org/10.1016/j.molcel.2014.02.018
Vajjhala, P. R., Kaiser, S., Smith, S. J., Ong, Q. R., Soh, S. L., Stacey, K. J., &
Hill, J. M. (2014). Identification of multifaceted binding modes for
pyrin and ASC pyrin domains gives insights into pyrin inflammasome
assembly. Journal of Biological Chemistry,289(34), 2350423519.
https://doi.org/10.1074/jbc.M114.553305
Vanaja, S. K., Russo, A. J., Behl, B., Banerjee, I., Yankova, M., Deshmukh, S.
D., & Rathinam, V. A. K. (2016). Bacterial outer membrane vesicles
mediate cytosolic localization of LPS and caspase11 activation. Cell,
165(5), 11061119. https://doi.org/10.1016/j.cell.2016.04.015
Vandanmagsar, B., Youm, Y. H., Ravussin, A., Galgani, J. E., Stadler, K.,
Mynatt, R. L., Dixit, V. D. (2011). The NLRP3 inflammasome
instigates obesityinduced inflammation and insulin resistance. Nature
Medicine (New York, NY, United States),17(2), 179188. https://doi.org/
10.1038/nm.2279
Wang, S., Yuan, Y. H., Chen, N. H., & Wang, H. B. (2019). The mechanisms
of NLRP3 inflammasome/pyroptosis activation and their role in
Parkinson's disease. International Immunopharmacology,67, 458464.
https://doi.org/10.1016/j.intimp.2018.12.019
Wang, Y., Gao, W., Shi, X., Ding, J., Liu, W., He, H., Shao, F. (2017).
Chemotherapy drugs induce pyroptosis through caspase3 cleavage
of a gasdermin. Nature,547(7661), 99103. https://doi.org/10.1038/
nature22393
Wang, Y., Yin, B., Li, D., Wang, G., Han, X., & Sun, X. (2018). GSDME
mediates caspase3dependent pyroptosis in gastric cancer. Biochem-
ical and Biophysical Research Communications,495(1), 14181425.
https://doi.org/10.1016/j.bbrc.2017.11.156
Wise, C. A., Gillum, J. D., Seidman, C. E., Lindor, N. M., Veile, R., Bashiardes,
S., & Lovett, M. (2002). Mutations in CD2BP1 disrupt binding to PTP
PEST and are responsible for PAPA syndrome, an autoinflammatory
disorder. Human Molecular Genetics,11(8), 961969.
Xia, X., Wang, X., Zheng, Y., Jiang, J., & Hu, J. (2019). What role does
pyroptosis play in microbial infection? Journal of Cellular Physiology,
234(6), 78857892. https://doi.org/10.1002/jcp.27909
Xu, H., Yang, J., Gao, W., Li, L., Li, P., Zhang, L., Shao, F. (2014). Innate
immune sensing of bacterial modifications of Rho GTPases by the
Pyrin inflammasome. Nature,513(7517), 237241. https://doi.org/10.
1038/nature13449
Xu, S., Li, X., Liu, Y., Xia, Y., Chang, R., & Zhang, C. (2019). Inflammasome
inhibitors: Promising therapeutic approaches against cancer. Journal
of Hematology & Oncology,12(1), 64. https://doi.org/10.1186/s13045
01907550
Yabal, M., Calleja, D. J., Simpson, D. S., & Lawlor, K. E. (2019). Stressing out
the mitochondria: Mechanistic insights into NLRP3 inflammasome
activation. Journal of Leukocyte Biology,105(2), 377399. https://doi.
org/10.1002/JLB.MR0318124R
Yang, J., Zhao, Y., & Shao, F. (2015). Noncanonical activation of
inflammatory caspases by cytosolic LPS in innate immunity. Current
Opinion in Immunology,32,7883. https://doi.org/10.1016/j.coi.2015.
01.007
Yi, Y. S. (2017). Caspase11 noncanonical inflammasome: A critical sensor
of intracellular lipopolysaccharide in macrophagemediated inflam-
matory responses. Immunology,152(2), 207217. https://doi.org/10.
1111/imm.12787
Yin, W., Zhou, Q. L., OuYang, S. X., Chen, Y., Gong, Y. T., & Liang, Y. M.
(2019). Uric acid regulates NLRP3/IL1beta signaling pathway and
further induces vascular endothelial cells injury in early CKD through
ROS activation and K(+) efflux. BMC Nephrology,20(1), 319. https://
doi.org/10.1186/s1288201915068
Yu, J. W., FernandesAlnemri, T., Datta, P., Wu, J., Juliana, C., Solorzano, L.,
Alnemri, E. S. (2007). Pyrin activates the ASC pyroptosome in
response to engagement by autoinflammatory PSTPIP1 mutants.
Molecular Cell,28(2), 214227. https://doi.org/10.1016/j.molcel.2007.
08.029
Yu, J. W., Wu, J., Zhang, Z., Datta, P., Ibrahimi, I., Taniguchi, S., Alnemri,
E. S. (2006). Cryopyrin and pyrin activate caspase1, but not NF
kappaB, via ASC oligomerization. Cell Death and Differentiation,13(2),
236249. https://doi.org/10.1038/sj.cdd.4401734
Zendedel, A., Monnink, F., Hassanzadeh, G., Zaminy, A., Ansar, M. M., Habib,
P., Beyer, C. (2018). Estrogen attenuates local inflammasome
expression and activation after spinal cord injury. Molecular Neurobiol-
ogy,55(2), 13641375. https://doi.org/10.1007/s1203501704002
Zhao, Y., & Shao, F. (2015). The NAIPNLRC4 inflammasome in innate
immune detection of bacterial flagellin and type III secretion
apparatus. Immunological Reviews,265(1), 85102. https://doi.org/10.
1111/imr.12293
Zheng, Y., Zhang, D., Zhang, L., Fu, M., Zeng, Y., & Russell, R. (2013).
Variants of NLRP3 gene are associated with insulin resistance in
Chinese Han population with type2 diabetes. Gene,530(1), 151154.
https://doi.org/10.1016/j.gene.2013.07.082
Zhong, F. L., Mamai, O., Sborgi, L., Boussofara, L., Hopkins, R., Robinson, K.,
Reversade, B. (2016). Germline NLRP1 mutations cause skin
inflammatory and cancer susceptibility syndromes via inflammasome
activation. Cell,167(1), 187202. https://doi.org/10.1016/j.cell.2016.
09.001. e117.
Zhou, Y., Lu, M., Du, R. H., Qiao, C., Jiang, C. Y., Zhang, K. Z., Hu, G.
(2016). MicroRNA7 targets Nodlike receptor protein 3 inflamma-
some to modulate neuroinflammation in the pathogenesis of
Parkinson's disease. Molecular Neurodegeneration,11, 28. https://doi.
org/10.1186/s1302401600943
de Zoete, M. R., & Flavell, R. A. (2014). Detecting different: Pyrin senses
modified GTPases. Cell Research,24(11), 12861287. https://doi.org/
10.1038/cr.2014.101.
Zychlinsky, A., Prevost, M. C., & Sansonetti, P. J. (1992). Shigella flexneri
induces apoptosis in infected macrophages. Nature,358(6382),
167169. https://doi.org/10.1038/358167a0
How to cite this article: Lu F, Lan Z, Xin Z, et al. Emerging
insights into molecular mechanisms underlying pyroptosis and
functions of inflammasomes in diseases. J Cell Physiol.
2019;115. https://doi.org/10.1002/jcp.29268
LU ET AL.
|
15
... Cru-cially, several anticancer agents can prompt pyroptosis in tumour cells by stimulating the NLRP3 inflammasome and the subsequent GSDMD pathway. 15 Pyroptotic cell demise not only directly eliminates cancer cells but also prompts the liberation of inflammatory mediators, which can enhance antitumour immunity and surmount treatment resistance. 16 A variety of tumours show dysregulated expression of the NLRP3 inflammasome, 17,18 its expression and regulatory mechanism in CRC remain unclear. ...
Article
Full-text available
Background Although numerous studies have indicated that activated pyroptosis can enhance the efficacy of antitumour therapy in several tumours, the precise mechanism of pyroptosis in colorectal cancer (CRC) remains unclear. Methods Pyroptosis in CRC cells treated with antitumour agents was assessed using various techniques, including Western blotting, lactate dehydrogenase release assay and microscopy analysis. To uncover the epigenetic mechanisms that regulate NLRP3, chromatin changes and NLRP3 promoter histone modifications were assessed using Assay for Transposase‐Accessible Chromatin using sequencing and RNA sequencing. Chromatin immunoprecipitation‒quantitative polymerase chain reaction was used to investigate the NLRP3 transcriptional regulatory mechanism. Additionally, xenograft and patient‐derived xenograft models were constructed to validate the effects of the drug combinations. Results As the core molecule of the inflammasome, NLRP3 expression was silenced in CRC, thereby limiting gasdermin D (GSDMD)‐mediated pyroptosis. Supplementation with NLRP3 can rescue pyroptosis induced by antitumour therapy. Overexpression of HDAC2 in CRC silences NLRP3 via epigenetic regulation. Mechanistically, HDAC2 suppressed chromatin accessibility by eliminating H3K27 acetylation. HDAC2 knockout promotes H3K27ac‐mediated recruitment of the BRD4‐p‐P65 complex to enhance NLRP3 transcription. Inhibiting HDAC2 by Santacruzamate A in combination with classic antitumour agents (5‐fluorouracil or regorafenib) in CRC xenograft‐bearing animals markedly activated pyroptosis and achieved a significant therapeutic effect. Clinically, HDAC2 is inversely correlated with H3K27ac/p‐P65/NLRP3 and is a prognostic factor for CRC patients. Conclusion Collectively, our data revealed a crucial role for HDAC2 in inhibiting NLRP3/GSDMD‐mediated pyroptosis in CRC cells and highlighted HDAC2 as a potential therapeutic target for antitumour therapy. Highlights Silencing of NLRP3 limits the GSDMD‐dependent pyroptosis in colorectal cancer. HDAC2‐mediated histone deacetylation leads to epigenetic silencing of NLRP3. HDAC2 suppresses the NLRP3 transcription by inhibiting the formation of H3K27ac/BRD4/p‐P65 complex. Targeting HDAC2 activates pyroptosis and enhances therapeutic effect.
... Then the cell suffered from swelling, osmotic dissolution, rupture and death, which indicated the induction of pyroptosis [16]. Once pyroptosis is activated, intracellular contents is released and a series of subsequent physiological processes can be induced [17,18]. Previous studies have shown that pyroptosis can activate TGF-β/Smad pathway in liver, kidney, lung and many other tissues, and then lead to tissue fibrosis [19][20][21]. ...
Article
Full-text available
Joint contracture is one of the common diseases clinically, and joint capsule fibrosis is considered to be one of the most important pathological changes of joint contracture. However, the underlying mechanism of joint capsule fibrosis is still controversial. The present study aims to establish an animal model of knee extending joint contracture in rats, and to investigate the role of hypoxia-mediated pyroptosis in the progression of joint contracture using this animal model. 36 male SD rats were selected, 6 of which were not immobilized and were used as control group, while 30 rats were divided into I-1 group (immobilized for 1 week following 7 weeks of free movement), I-2 group (immobilized for 2 weeks following 6 weeks of free movement), I-4 group (immobilized for 4 weeks following 4 weeks of free movement), I-6 group (immobilized for 6 weeks following 2 weeks of free movement) and I-8 group (immobilized for 8 weeks) according to different immobilizing time. The progression of joint contracture was assessed by the measurement of knee joint range of motion, collagen deposition in joint capsule was examined with Masson staining, protein expression levels of HIF-1α, NLRP3, Caspase-1, GSDMD-N, TGF-β1, α-SMA and p-Smad3 in joint capsule were assessed using western blotting, and the morphological changes of fibroblasts were observed by transmission electron microscopy. The degree of total and arthrogenic contracture progressed from the first week and lasted until the first eight weeks after immobilization. The degree of total and arthrogenic contracture progressed rapidly in the first four weeks after immobilization and then progressed slowly. Masson staining indicated that collagen deposition in joint capsule gradually increased in the first 8 weeks following immobilization. Western blotting analysis showed that the protein levels of HIF-1α continued to increase during the first 8 weeks of immobilization, and the protein levels of pyroptosis-related proteins NLRP3, Caspase-1, GSDMD-N continued to increase in the first 4 weeks after immobilization and then decreased. The protein levels of fibrosis-related proteins TGF-β1, p-Smad3 and α-SMA continued to increase in the first 8 weeks after immobilization. Transmission electron microscopy showed that 4 weeks of immobilization induced cell membrane rupture and cell contents overflow, which further indicated the activation of pyroptosis. Knee extending joint contracture animal model can be established by external immobilization orthosis in rats, and the activation of hypoxia-mediated pyroptosis may play a stimulating role in the process of joint capsule fibrosis and joint contracture.
Article
Full-text available
Pseudomonas aeruginosa is a highly adaptable opportunistic pathogen capable of exploiting barriers and immune defects to cause chronic lung infections in conditions such as cystic fibrosis. In these contexts, host immune responses are ineffective at clearing persistent bacterial infection, instead driving a cycle of inflammatory lung damage. This review outlines key components of the host immune response to chronic P. aeruginosa infection within the lung, beginning with initial pathogen recognition, followed by a robust yet maladaptive innate immune response, and an ineffective adaptive immune response that propagates lung damage while permitting bacterial persistence. Untangling the interplay between host immunity and chronic P. aeruginosa infection will allow for the development and refinement of strategies to modulate immune-associated lung damage and potentiate the immune system to combat chronic infection more effectively.
Article
Full-text available
Noninflammatory apoptosis is transformed into inflammatory pyroptosis by activating caspase‐3 to lyse gasdermin E (GSDME), and this process can be used as an effective therapeutic strategy. Thus, a selective and powerful inducer of activated caspase‐3 plays a vital role in pyroptosis‐based cancer therapy. Herein, a human cell membrane vesicle‐based nanoplatform (HCNP) is designed for photodynamic therapy (PDT). HCNP is modified with vesicular stomatitis virus G‐protein (VSVG) to anchor nano‐photosensitizers on the tumor cell membrane. Photosensitizers are bonded to HCNP by clicking chemical reaction as pyroptosis inducers. The results show that HCNP effectively disrupts the mitochondrial function of cells by generating reactive oxygen species (ROS) upon laser irradiation; concomitantly, GSDME is cleaved by activated caspase‐3 and promotes pyroptosis of lung cancer cells. Here an effective intervention strategy is proposed to induce pyroptosis based on light‐activated PDT.
Article
Intestinal inflammatory disease (IBD) is a chronic intestinal disorder that includes Crohn’s disease and ulcerative colitis. In recent years, its incidence has been increasing, but there is still no breakthrough in its treatment. Cell death was divided into programmed and nonprogrammed cell death (PCD). PCD mainly includes ferroptosis, apoptosis, necroptosis, pyroptosis, and autophagy, while non-PCD refers to necrosis. Increasing evidence shows that PCD, such as ferroptosis and autophagy, plays an important regulatory role in intestinal diseases, which may be involved in the occurrence and progression of IBD and can be used as a therapeutic target for the disease. This article reviews recent studies on ferroptosis, apoptosis, necroptosis, pyroptosis, and autophagy associated with IBD, combs the regulatory mechanisms of PCD in IBD, and summarizes the corresponding therapeutic drugs and new materials, aiming to provide new directions and treatments for IBD research.
Chapter
Inflammation triggers specific metabolic pathways and if not resolved, translates into several painful diseases such as rheumatoid arthritis, lupus, Alzheimer's disease, cardiovascular disorders and psoriasis. Various processes have been explored to understand the factors behind inflammation and consequently, many mechanisms have been examined to suppress it. The nucleotide-binding domain like receptor 3 (NLRP3) inflammasome is an example of such factors which is responsible for triggering sterile and microbe induced inflammation. Studies of genetic variants of the related gene have revealed insights into the mRNA expression pathways that may help researchers to identify crucial disease mechanisms. This book is a review of the scientific findings of distinguished scholars who have studied NLRP3 inflammasome activation and its contribution in worsening the outcomes of inflammatory disorders. This collection of chapters covers many aspects of the multifaceted role of NLRP3 inflammasome. Beginning with airway inflammation and fibrosis, it progresses to explore its involvement in pulmonary hypertension, heart diseases, tuberculosis, cardiovascular complications, and childhood asthma. Additionally, it examines the inflammasome's impact on protozoan parasitic infections and neuropathic pain. The chapters not only elucidate the intricate mechanisms of NLRP3 activation but also discuss potential inhibitors and therapeutic targets. Readers will gain a comprehensive understanding of the NLRP3 inflammasome's diverse implications across different physiological contexts. The book includes references making this book a valuable treatise of insights for researchers, clinicians, and healthcare professionals.
Article
Full-text available
Background: Chronic kidney disease (CKD) has been considered as a major health problem in the world. Increasing uric acid (UA) could induce vascular endothelial injury, which is closely related to microinflammation, oxidative stress, and disorders of lipids metabolism. However, the specific mechanism that UA induces vascular endothelial cells injury in early CKD remains unknown. Methods: Human umbilical vein endothelial cells (HUVECs) were cultured and subjected to different concentrations of UA for different periods. Early CKD rat model with elevated serum UA was established. Western blotting and quantitative real-time PCR (qPCR) were applied for measuring protein and mRNA expression of different cytokines. The animals were sacrificed and blood samples were collected for measurement of creatinine, UA, IL-1β, TNF-α, and ICAM-1. Renal tissues were pathologically examined by periodic acid-Schiff (PAS) or hematoxylin-eosin (HE) staining. Results: The expression of IL-1β, ICAM-1, NLRP3 complexes, and activation of NLRP3 inflammasome could be induced by UA, but the changes induced by UA were partially reversed by siRNA NLRP3 or caspase 1 inhibitor. Furthermore, we identified that UA regulated the activation of NLRP3 inflammasome by activating ROS and K+ efflux. In vivo results showed that UA caused the vascular endothelial injury by activating NLRP3/IL-1β pathway. While allopurinol could reduce UA level and may have protective effects on cardiovascular system. Conclusions: UA could regulate NLRP3/IL-1β signaling pathway through ROS activation and K+ efflux and further induce vascular endothelial cells injury in early stages of CKD.
Article
Full-text available
Inflammasome activation can trigger an inflammatory and innate immune response through the release of cytokines and induction of pyroptosis. A dysfunctional inflammasome has been implicated in the development of human pathologies, including sepsis and septic shock. Here, we show that advanced glycosylation end-product specific receptor (AGER/RAGE) is required for caspase-11 inflammasome activation in macrophages. A nuclear damage-associated molecular pattern (nDAMP) complex, including high-mobility group box 1, histone, and DNA, can promote caspase-11-mediated gasdermin D cleavage, interleukin 1β proteolytic maturation, and lactate dehydrogenase release. The inhibition of AGER-mediated lipid peroxidation via arachidonate 5-lipoxygenase (ALOX5) limits caspase-11 inflammasome activation and pyroptosis in macrophages in response to nDAMPs or cytosolic lipopolysaccharide. Importantly, the pharmacologic inhibition of the AGER-ALOX5 pathway or global depletion (Ager−/−) or conditional depletion of AGER in myeloid cells (AgerMye−/−) protects against lipopolysaccharide-induced septic death in poly(I:C)-primed mice. These data identify a molecular basis for caspase-11 inflammasome activation and provide a potential strategy to treat sepsis.
Article
Full-text available
Inflammation has long been accepted as a key component of carcinogenesis. During inflammation, inflammasomes are potent contributors to the activation of inflammatory cytokines that lead to an inflammatory cascade. Considering the contributing role of inflammasomes in cancer progression, inflammasome inhibitors seem to have a promising future in cancer treatment and prevention. Here, we summarize the structures and signaling pathways of inflammasomes and detail some inflammasome inhibitors used to treat various forms of cancer, which we expect to be used in novel anticancer approaches. However, the practical application of inflammasome inhibitors is limited in regard to specific types of cancer, and the associated clinical trials have not yet been completed. Therefore, additional studies are required to explore more innovative and effective medicines for future clinical treatment of cancer.
Article
Full-text available
Background Oxidative stress-induced endothelial dysfunction and pyroptosis play an important role during chronic kidney disease (CKD) progression. Neferine, which is an alkaloid ingredient from the lotus seed embryo, has many biological actions such as anti-inflammatory, anticancer and antioxidant. However, the role of neferine in endothelial cell pyroptosis and the involved mechanism remain obscure. The aim is to probe the protective effects of neferine on cell pyroptosis and the involved underlying mechanism. Methods After the HUVECs were primed with neferine treatment for 2 h prior to LPS and ATP exposure for 24 h, the cell proliferation was determined by BrdU; the cell LDH release was detected by LDH kits; the levels of intracellular ROS, MDA and SOD were tested by detection kits; Caspase-1 activity kit was used to determine caspase-1 activity; the contents of NLRP3, ASC, caspase-1, IL-1β, IL-18 and GSDMD were tested by RT-PCR and western blot. Results We found that neferine could inhibit LPS-ATP-induced oxidative stress and the activation of NLRP3 inflammasome signaling, and increased the endothelial cell viability and SOD production. siRNA which mediated the knockdown of NLRP3 promoted the neferine-induced inhibition effects of cell pyroptosis. Furthermore, these neferine-induced effects were reversed by the over-expression of NLRP3. Conclusions Our findings indicated neferine may reduce ROS by anti-oxidation and inhibit LPS-ATP-induced endothelial cell pyroptosis via blocking ROS/NLRP3/Caspase-1 signaling pathway, which provides the evidence for therapeutic effect in CKD.
Article
Full-text available
Several large-scale genome-wide association studies genetically linked IRGM to Crohn’s disease and other inflammatory disorders in which the IRGM appears to have a protective function. However, the mechanism by which IRGM accomplishes this anti-inflammatory role remains unclear. Here, we reveal that IRGM/Irgm1 is a negative regulator of the NLRP3 inflammasome activation. We show that IRGM expression, which is increased by PAMPs, DAMPs, and microbes, can suppress the pro-inflammatory responses provoked by the same stimuli. IRGM/Irgm1 negatively regulates IL-1β maturation by suppressing the activation of the NLRP3 inflammasome. Mechanistically, we show that IRGM interacts with NLRP3 and ASC and hinders inflammasome assembly by blocking their oligomerization. Further, IRGM mediates selective autophagic degradation of NLRP3 and ASC. By suppressing inflammasome activation, IRGM/Irgm1 protects from pyroptosis and gut inflammation in a Crohn’s disease experimental mouse model. This study for the first time identifies the mechanism by which IRGM is protective against inflammatory disorders.
Article
Full-text available
Pyroptosis, a type of programmed cell death mediated by gasdermin, is characterized by the swelling and rupture of cells, release of cellular contents and a strong inflammatory response, which is critical for controlling microbial infection. Pattern recognition receptors recognize the intracellular and extracellular pathogenic microbial components and stimulate the organism's inflammatory response by activating the pyroptosis signaling pathway and releasing interleukin‐1β (IL‐1β), IL‐18, and other inflammatory factors to promote pathogen clearance and prevent infection. In the process of continuous evolution, pathogens have developed multiple strategies to modulate the occurrence of pyroptosis and thus enhance their ability to induce disease; that is, the competition between host cells and pathogens controls the occurrence of pyroptosis. Competition can directly affect tissue inflammation outbreaks and even alter cell survival. Studies have shown that various bacterial infections, including Shigella flexneri, Salmonella, Listeria monocytogenes, and Legionella pneumophila, can lead to pyroptosis. Pyroptosis is associated with the occurrence and development of various diseases caused by microbial infection, and the identification of molecules related to the pyroptosis signaling pathway may provide new drug targets for the treatment of related diseases. This study reviews the molecular mechanisms of pyroptosis and the role of pyroptosis in microbial infection‐related diseases.
Article
Full-text available
Inflammatory caspase-11/4/5 recognize cytosolic LPS from invading Gram-negative bacteria and induce pyroptosis and cytokine release, forming rapid innate antibacterial defenses. Since extracellular or vacuole-constrained bacteria are thought to rarely access the cytoplasm, how their LPS are exposed to the cytosolic sensors is a critical event for pathogen recognition. Hemolysin is a pore-forming bacterial toxin, which was generally accepted to rupture cell membrane, leading to cell lysis. Whether and how hemolysin participates in non-canonical inflammasome signaling remains undiscovered. Here, we show that hemolysin-overexpressed enterobacteria triggered significantly increased caspase-4 activation in human intestinal epithelial cell lines. Hemolysin promoted LPS cytosolic delivery from extracellular bacteria through dynamin-dependent endocytosis. Further, we revealed that hemolysin was largely associated with bacterial outer membrane vesicles (OMVs) and induced rupture of OMV-containing vacuoles, subsequently increasing LPS exposure to the cytosolic sensor. Accordingly, overexpression of hemolysin promoted caspase-11 dependent IL-18 secretion and gut inflammation in mice, which was associated with restricting bacterial colonization in vivo. Together, our work reveals a concept that hemolysin promotes noncanonical inflammasome activation via liberating OMVs for cytosolic LPS sensing, which offers insights into innate immune surveillance of dysregulated hemolysin via caspase-11/4 in intestinal antibacterial defenses.
Article
IRGM is a risk factor for several inflammatory diseases, yet no direct link to immune regulation had been shown. In this issue of Molecular Cell, Mehto et al. (2019) report that IRGM limits NLRP3 inflammasome activation—by both direct inhibition of NLRP3/ASC oligomerization and selective autophagic destruction of NLRP3/ASC.
Article
Parkinson's disease (PD) is a typical neurodegenerative disease and the pathological feature of which is the death of dopamine neurons in the substantia nigra region. At present, neuronal death caused by inflammatory cytokine-mediated neuroinflammation is being extensively studied. The nucleotide-binding oligomerization domain-, leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome is an inflammatory complex existing in microglia. Its activation promotes the secretion of the inflammatory cytokine interleukin-1β/18 (IL-1β/18) and induces pyroptosis, a type of cell death that possesses the potential for inflammation, to rupture microglia to further release IL-1β. In this review we focus on the mechanisms of activation of the NLRP3 inflammasome and pyroptosis and their inflammatory effects on the development of PD. In addition, we focus on some inhibitors of NLRP3 inflammatory pathways to alleviate the progression of PD by inhibiting central inflammation and provide new therapeutic strategies for the treatment of PD.
Article
Inflammasomes are multimeric protein complexes that induce the cleavage and release of bioactive IL-1β and cause a lytic form of cell death, termed pyroptosis. Due to its diverse triggers, ranging from infectious pathogens and host danger molecules to environmental irritants, the NOD-like receptor protein 3 (NLRP3) inflammasome remains the most widely studied inflammasome to date. Despite intense scrutiny, a universal mechanism for its activation remains elusive, although, recent research has focused on mitochondrial dysfunction or potassium (K+) efflux as key events. In this review, we give a general overview of NLRP3 inflammasome activation and explore the recently emerging noncanonical and alternative pathways to NLRP3 activation. We highlight the role of the NLRP3 inflammasome in the pathogenesis of metabolic disease that is associated with mitochondrial and oxidative stress. Finally, we interrogate the mechanisms proposed to trigger NLRP3 inflammasome assembly and activation. A greater understanding of how NLRP3 inflammasome activation is triggered may reveal new therapeutic targets for the treatment of inflammatory disease. Review of the ambiguities surrounding mitochondrial damage-mediated activation of NLRP3, including new models for how cell death may activate NLRP3 to influence inflammation.