ArticlePDF Available

Virulent Mycobacterium bovis Beijing Strain Activates the NLRP7 Inflammasome in THP-1 Macrophages

PLOS
PLOS ONE
Authors:
  • Case Western Reserve University Ohio USA

Abstract and Figures

Mycobacterium bovis is the causative agent of tuberculosis in a wide range of mammals, including humans. Macrophages are the first line of host defense. They secrete proinflammatory cytokines, such as interleukin-1 beta (IL-1β), in response to mycobacterial infection, but the underlying mechanisms by which human macrophages are activated and release IL-1β following M. bovis infection are poorly understood. Here we show that the ‘nucleotide binding and oligomerization of domain-like receptor (NLR) family pyrin domain containing 7 protein’ (NLRP7) inflammasome is involved in IL-1β secretion and caspase-1 activation induced by M. bovis infection in THP-1 macrophages. NLRP7 inflammasome activation promotes the induction of pyroptosis as well as the expression of tumor necrosis factor alpha (TNF-α), Chemokine (C-C motif) ligand 3 (CCL3) and IL-1β mRNAs. Thus, the NLRP7 inflammasome contributes to IL-1β secretion and induction of pyroptosis in response to M. bovis infection in THP-1 macrophages.
Content may be subject to copyright.
RESEARCH ARTICLE
Virulent Mycobacterium bovis Beijing Strain
Activates the NLRP7 Inflammasome in THP-1
Macrophages
Yang Zhou
1
, Syed Zahid Ali Shah
1
, Lifeng Yang
1
, Zhongqiu Zhang
2
, Xiangmei Zhou
1
*,
Deming Zhao
1
*
1National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal
Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key
Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China, 2Veterinary Bureau,
Ministry of Agriculture of the Peoples Republic of China, Beijing 100125, China
*zhouxm@cau.edu.cn (XZ); zhaodm@cau.edu.cn (DZ)
Abstract
Mycobacterium bovis is the causative agent of tuberculosis in a wide range of mammals,
including humans. Macrophages are the first line of host defense. They secrete proinflam-
matory cytokines, such as interleukin-1 beta (IL-1β), in response to mycobacterial infection,
but the underlying mechanisms by which human macrophages are activated and release
IL-1βfollowing M.bovis infection are poorly understood. Here we show that the nucleotide
binding and oligomerization of domain-like receptor (NLR) family pyrin domain containing 7
protein(NLRP7) inflammasome is involved in IL-1βsecretion and caspase-1 activation
induced by M.bovis infection in THP-1 macrophages. NLRP7 inflammasome activation pro-
motes the induction of pyroptosis as well as the expression of tumor necrosis factor alpha
(TNF-α), Chemokine (C-C motif) ligand 3 (CCL3) and IL-1βmRNAs. Thus, the NLRP7
inflammasome contributes to IL-1βsecretion and induction of pyroptosis in response to M.
bovis infection in THP-1 macrophages.
Introduction
Mycobacterium bovis, a member of the M.tuberculosis complex, is the etiological agent of
bovine tuberculosis which is estimated to infect more than 50 million cattle per annum with
concomitant economic losses of approximately $3 billion worldwide [1]. M.bovis is also
responsible for a proportion of human tuberculosis (TB) cases, and can be transmitted from
human to human. About 2.8% of all human TB cases in Africa are caused by M.bovis, and
human M.bovis infection accounts for 7.6% of cases in Mexico which also contributes to the
disease incidence in the United States, although overall incidence in the Americas is low [2].
This huge zoonotic risk imposes limitations on the potential for control [3,4].
Macrophages are considered the first line of host defense against invasive microbes. Upon
infection, they initiate inflammatory responses by releasing cytokines and chemokines, such as
IL-1β, IL-18, TNF-α, and CCL3. Among these, IL-1βis a potent mediator of antimicrobial
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 1/13
a11111
OPEN ACCESS
Citation: Zhou Y, Shah SZA, Yang L, Zhang Z, Zhou
X, Zhao D (2016) Virulent Mycobacterium bovis
Beijing Strain Activates the NLRP7 Inflammasome in
THP-1 Macrophages. PLoS ONE 11(4): e0152853.
doi:10.1371/journal.pone.0152853
Editor: Volker Briken, University of Maryland,
UNITED STATES
Received: December 31, 2015
Accepted: March 21, 2016
Published: April 4, 2016
Copyright: © 2016 Zhou et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper.
Funding: This work was funded by MoSTRCUK
international cooperation project (Project No.
2013DFG32500) http://www.istcp.org.cn, National
Natural Science Foundation of China (Project No.
31572487) https://isisn.nsfc.gov.cn, Funding of State
Key Lab of Agrobiotechnology (Project No.
2012SKLAB06-14) http://cbs.cau.edu.cn, 2015 CAU
Foreign Experts Major Projects (Project No:
2012z018) http://cau.edu.cn, and High-end Foreign
Experts Recruitment Program (Project No:
GDW20151100036) http://cepms.safea.gov.cn.
responses. It contributes to the maturation of mycobacterial phagosomes into phagolysosomes,
which enhances mycobacterial elimination by macrophages. Inhibition of IL-1βactivity by
neutralizing antibody or siRNA increases intracellular mycobacterial survival [5]. Conversely,
adding exogenous IL-1βmarkedly inhibits their survival [6]. IL-1βactivity is tightly controlled
at the levels of expression, maturation, and secretion [7]. It is initially synthesized as a precur-
sor molecule, proIL-1β, in the cytosol in response to pathogen-associated molecular patterns
(PAMPs), which are sensed by evolutionarily-conserved toll-like receptors. Its maturation and
secretion requires caspase-1 activation by multiprotein complexes known as inflammasomes
[8]. The inflammasomes consist of a receptor protein, the adaptor apoptosis-associated speck-
like protein containing a caspase-activation recruitment domain (ASC), and caspase-1. Recep-
tor proteins include NLR family pyrin domain containing 3 protein (NLRP3) [9], absent in
melanoma 2 (AIM2) [10], NLR family caspase-activation recruitment domain (CARD)-con-
taining protein 4 (NLRC4) [11], and NLRP7, which is uniquely stimulated by microbial acety-
lated lipopeptides [12]. The majority of research on NLRP7 has been associated with
hydatidiform mole, an abnormal human pregnancy with hyperproliferative vesicular tropho-
blast and no fetal development [13]. A recent study showed that some live and heat killed
microbes, including Mycoplasma spp., Staphylococcus aureus and Listeria monocytogenes, acti-
vate the NLRP7 inflammasome. NLRP7 senses lipopeptides through its leucine-rich repeat
(LRR) domain [12], and results in self-oligomerization to form an inflammasome scaffold
through its nucleotide-binding and oligomerization (NACHT) domain. It interacts with ASC
via homotypic pyrin domain interactions, recruiting procaspase-1 via the CARD domain of
ASC. Procaspase-1 clustering leads to caspase-1 auto-activation and generation of active cas-
pase-1, which cleaves inactive proinflammatory cytokines into their active forms.
Little is known about the protective role of the NLRP7 inflammasome against either M.
bovis or M.tuberculosis in macrophages. M.bovis is an intracellular pathogen expressing and
secreting lipoproteins [1416] and we demonstrate here that M.bovis infection triggers NLRP7
inflammasome activation and induction of pyroptosis in human THP-1 macrophages.
Materials and Methods
Reagents
The following antibodies and reagents were purchased from the indicated suppliers: the mouse
monoclonal antibody against NLRP7 used for immunofluorescence assay and rabbit polyclonal
anti-AIM2 antibody, Santa Cruz Biotechnology; rabbit polyclonal anti-NLRP7 antibody used
for western blotting, Pierce/Thermo Fisher Scientific; rabbit polyclonal anti-ASC antibody and
rabbit polyclonal anti-NLRP3 antibody, Sangon Biotech, Shanghai, China; goat polyclonal
anti-IL-1βantibody, R&D Systems; rabbit polyclonal anti-β-actin antibody, Proteintech,
Wuchan, China; rabbit polyclonal anti-caspase-1 antibody, ProSci Incorporated; phorbol
12-myristate-13-acetate (PMA), Sigma-Aldrich; glycine, Beijing Solarbio Science & Technol-
ogy Co., Ltd.; cytochalasin D, Cayman Chemical; and Z-YVAD-FMK, BioVision Incorporated.
THP-1 cell culture and differentiation
THP-1 cells were obtained from American Type Culture Collection (Manassas, VA, USA) and
maintained in RPMI 1640 medium (Gibco, Grand Island, NY, USA) containing 10% fetal
bovine serum (FBS, Gibco). THP-1 cells were stimulated with PMA (5 ng/mL) to differentiate
into macrophages for 2 days, after which the cells were washed three times with warm phos-
phate-buffered saline (PBS). Cells were then incubated in PMA-free culture medium and rested
for a further 2 days.
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 2/13
Competing Interests: The authors declare that no
competing interests exist.
Bacterial culture and infection
Virulent M.bovis Beijing strain was obtained from the China Institute of Veterinary Drug Con-
trol, Beijing and grown in 7H9 Middlebrook media (BD Biosciences) supplemented with albu-
min-dextrose-catalase (ADC) enrichment solution and 0.05% Tween-80 (Difco) at 37°C. THP-
1 macrophages were infected with M.bovis at a multiplicity of infection (MOI) indicated for 2
h and then washed three times with warm PBS to remove extracellular bacteria. The samples
were harvested at the indicated time.
Small interference RNA (siRNA) transfection
THP-1 macrophages were transfected with gene-specific siRNA pools to knock down NLRP7
or ASC. Human NLRP7-targeting siRNA oligonucleotides, ASC-targeting siRNA oligonucleo-
tides and non-targeting control siRNA oligonucleotides were obtained from Shanghai Gene-
Pharma Co., Ltd (Table 1). THP-1 cells were differentiated and then incubated overnight in a
24-well plate. Prior to transfection, all medium was removed and 400 μL of fresh medium was
added. Lipofectamine 3000 transfection reagent and siRNA were added into 100 μL of serum-
free culture medium, and incubated for 10 min at room temperature. The resulting mixture
was added drop-wise onto the cells and culture medium was replaced after 24 h.
Quantitative real-time PCR
Total RNA extraction was performed using RN28-EASYspin Plus Tissue/Cell RNA Kit (Aidlab
Biotech, Beijing) and reverse transcription was performed using RevertAid First Strand cDNA
Synthesis Kit (Thermo Fisher). Quantitative PCR was carried out in a Roche LightCycler480 II
using TransStart Green qPCR SuperMix UDG (Beijing TransGen Biotech). Primers for NLRP7
were 5´-TAAGGAATGCGACTGTGAACATC-3´ forward and 5´-TGCTAACTCCGAGTCTTC
TTCT-3´ reverse. Primers for NLRP3, AIM2, and GAPDH were the same as those used in our
previous study [17].
Table 1. Sequences of ASC-targeting siRNA and non-targeting control siRNA.
Name Sequence (sense, antisense)
NLRP7-targeting siRNA:
Target Sequence 1: GACGUCACUCUGAGAAACCAATT
UUGGUUUCUCAGAGUGACGUCTT
Target Sequence 2: GUCAGAGGGUCACAUGUUATT
UAACAUGUGACCCUCUGACTT [12]
Target Sequence 3: GUGUUCCUGGAGAAUUACATT
UGUAAUUCUCCAGGAACACTT [12]
ASC-targeting siRNA:
Target Sequence 1: UCGCGAGGGUCACAAACGUTT
ACGUUUGUGACCCUCGCGATT
Target Sequence 2: UGCUGUCCAUGGACGCCUUTT
AAGGCGUCCAUGGACAGCATT
Target Sequence 3: GCAAGAUGCGGAAGCUCUUTT
AAGAGCUUCCGCAUCUUGCTT
Non-targeting siRNA: UUCUCCGAACGUGUCACGUTT
ACGUGACACGUUCGGAGAATT
doi:10.1371/journal.pone.0152853.t001
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 3/13
Western blotting
Cells were washed in PBS, and lysed in cold lysis buffer (Beyotime Institute of Biotechnology,
China) for 20 min. Samples were centrifuged at 12,000 ×gfor 20min and the supernatant was
boiled for 10 min after addition of loading buffer (250 mM Tris-HCl pH 6.8, 10% SDS, 0.5%
BPB, 50% glycerol, 0.5 M DTT). For detection of IL-1βand caspase-1 released into the culture
medium, proteins were precipitated as described previously [18]. Aliquots were separated via
SDS-PAGE and the proteins were transferred to PVDF membranes (Immobilon-PSQ,
ISEQ00010, 0.2 μm). Blots were blocked by 5% non-fat milk in TBST (25 mMTris base, 137
mM sodium chloride, 2.7 mM potassium chloride and 0.05% Tween-20, pH7.4) for 1 h at
room temperature, incubated with the indicated primary antibody overnight at 4°C and the
corresponding HRP-labeled secondary antibody for 50 min at 37°C, and the signal detected
using an enhanced chemiluminescence (ECL) detection kit (Bio-Rad, USA).
Immunofluorescence
THP-1 cells were fixed with 4% paraformaldehyde for 10 min at room temperature. Following
permeabilization with 0.1% Triton X-100 for 10 min, the cells were blocked with 1% BSA for 1
h, incubated with primary antibodies overnight at 4°C and secondary antibodies at 37°C for 1
h. Nuclei were stained with DAPI for 1 min. Finally, coverslips were mounted on slides, and
the cells were imaged using confocal microscopy. Colocalization was quantified using ImageJ
software.
Lactate dehydrogenase (LDH) release assay
LDH release was measured using LDH Cytotoxicity Assay Kit (Cayman Incorporated) accord-
ing to the manufacturers instructions.
Statistical analysis
All assays were performed in three independent experiments and data were analyzed using
GraphPad Prism 5.0 software and Students t test; p <0.05 values were considered statistically
significant.
Results
M.bovis infection induces caspase-1 activation and IL-1βsecretion in
THP-1 macrophages
We first examined whether M.bovis could induce caspase-1 activation and IL-1βsecretion in
THP-1 monocyte-derived macrophages at various MOIs (Fig 1A). Infection of THP-1 macro-
phages with M.bovis led to release of IL-1βinto the supernatant in a dose-dependent fashion
at MOIs ranging from 0.1 to 10, but IL-1βsecretion was not enhanced further at an MOI of
100. Meanwhile, bacterial challenge resulted in increased production of proIL-1β, a precursor
of IL-1β.M.bovis also induced caspase-1 maturation, as evidenced by increased levels of the
cleaved p20 subunit in the supernatant, which directly correlated with MOI. To investigate
whether capase-1 activation is required for IL-1βsecretion induced by M.bovis, THP-1 macro-
phages were pretreated with Z-YVAD-FMK, a cell permeable inhibitor of caspase-1; this con-
siderably reduced IL-1βsecretion (but not proIL-1βproduction) upon M.bovis infection (Fig
1B). As M.bovis can be internalized by macrophages, the contribution of intracellular bacteria
to IL-1βsecretion and caspase-1 activation was examined. THP-1 macrophages were treated
with cytochalasin D, a drug that inhibits actin polymerization and thus blocks phagocytosis of
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 4/13
M.bovis. Substantial inhibition of caspase-1 activation and IL-1βrelease were observed follow-
ing M.bovis infection (Fig 1C). Taken together, these data show that M.bovis induces IL-1β
secretion in a caspase-1-dependent manner in THP-1 macrophages.
M.bovis infection upregulates the expression of NLRP7 mRNA
The NLRP7 inflammasome is activated by a variety of microorganisms through the recogni-
tion of microbial acetylated lipopeptides. Since M.bovis expresses secreted and membrane-
associated lipoproteins, such as MPB70/80, MPB83 [16], and P27 [15]. To explore whether the
NLRP7 inflammasome is activated by M.bovis, we initially examined the time course of the
mRNA expression of NLRP7 in THP-1 macrophages. Treatment with M.bovis significantly
upregulated the expression of NLRP7 mRNA at 14 h post-infection (hpi). The levels increased
to approximately 3-fold relative to negative control, and then decreased to 1.3-fold at 50 hpi
(Fig 2A). Stimulation with M.bovis at different MOIs ranging from 0.1 to 100 revealed that
NLRP7 was upregulated in a dose-dependent manner (Fig 2B). Although there was an increase
in transcriptional level, infection with M.bovis at MOIs of 0.1 and 1 failed to produce a signifi-
cant change. In spite of upregulation of NLRP7 at the mRNA level, there is no change at the
protein level even at an MOI of 10 at 50 hpi (Fig 2C), or at an MOI of 100 at 14 hpi (Fig 2D).
The AIM2 inflammasome is activated through the recognition of DNA during M.bovis infec-
tion [19,20], and the NLRP3 inflammasome is thought to be activated after exposure to the
secreted protein, ESAT-6 [21,22]. To clarify the role of the AIM2 and NLRP3 inflammasomes
in M.bovis-infected THP-1 macrophages, we quantified their expression following infection.
There was a rapid induction of NLRP3 mRNA within 2 hpi, which then fell gradually in a
time-dependent manner, reaching control levels at 26 hpi (Fig 2E). AIM2 mRNA increased as
early as 26 hpi, and continued to increase at 50 hpi (Fig 2F). M.bovis infection failed to induce
any changes in protein levels of NLRP3 and AIM2 (Fig 2C). Infections at various MOIs
showed that M.bovis-induced upregulation of both NLRP3 and AIM2 was dose-dependent
Fig 1. M.bovis triggers caspase-1 activation and IL-1βsecretion in THP-1 macrophages. A. Cells were infected at the indicated MOIs and samples
were harvested at 14 hpi. Culture supernatant was analyzed for IL-1βand caspase-1, and cell lysates were analyzed for proIL-1β, procaspase-1, and β-actin
by immunoblotting. B. Cells were pretreated with 50 μM Z-YVAD-FMK for 1 h, and then infected with M.bovis at an MOI of 10 in the presence or absence of
Z-YVAD-FMK. Culture supernatant and lysates were analyzed by immunoblotting. C. Cells were treated with 1 μg/mL cytochalasin D to block phagocytosis
for 1 h, and then infected with M.bovis in the presence or absence of cytochalasin D. Culture supernatant and lysates were analyzed by immunoblotting.
Abbreviations: Sup, culture supernatant; Lys, cell lysates; Z-YVAD, Z-YVAD-FMK; cyto D, cytochalasin D. Data from one representative experiment of three
are presented.
doi:10.1371/journal.pone.0152853.g001
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 5/13
(Fig 2G and 2H). Thus, M.bovis induces upregulation of the mRNA level of NLRP7 besides
NLRP3 and AIM2.
The NLRP7 inflammasome contributes to caspase-1 activation and IL-
1βsecretion during M.bovis infection
To further investigate whether the NLRP7 inflammasome plays a role in M.bovis-induced IL-
1β, we utilized a pool of siRNAs to knock down NLRP7 in THP-1 macrophages. Compared to
non-targeting control, siRNA-mediated knockdown significantly reduced the protein levels of
NLRP7, and also attenuated caspase-1 activation and IL-1βsecretion following stimulation
with M.bovis (Fig 3A). NLRP7 promotes IL-1βsecretion via activation of the inflammasome,
Fig 2. M.bovis infection leads to upregulation of NLRP7 mRNA expression. A, E, F. THP-1 macrophages were infected with M.bovis for the indicated
times. Cell lysates were subjected to quantitative real-time PCR analysis. *0.01 <P<0.05, **P<0.01. B, G, H. Cells were infected with M.bovis at the
indicated MOI. Lysates were harvested at 14 hpi, and subjected to quantitative real-time PCR analysis. C. Cells were infected with M.bovis for the indicated
times, and supernatant and lysates were analyzed by immunoblotting. D. Cells were infected with M.bovis at the indicated MOI at 14 hpi, and supernatant
and lysates were analyzed by immunoblotting.
doi:10.1371/journal.pone.0152853.g002
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 6/13
which is a multiprotein complex that contains ASC and caspase-1. siRNA knockdown experi-
ments also showed that loss of ASC significantly reduced the induction of caspase-1 activity
and IL-1βsecretion following infection with M.bovis (Fig 3B).
Upon specific stimulation, NLRP7 colocalizes and interacts with ASC and caspase-1 to form
the NLRP7 inflammasome. To confirm M.bovis-induced NLRP7 inflammasome activation, we
carried out an immunofluorescence assay. In M.bovis-infected THP-1 macrophages, we
observed that NLRP7 colocalized with ASC and caspase-1 in the perinuclear area, with some
colocalization in the nucleus (Fig 3C). These effects were also observed after stimulation with
Pam3CSK4, an NLRP7 inflammasome inducer (Fig 3D). Taken together, these data suggest
that M.bovis infection induces NLRP7 inflammasome activation, which in turn promotes cas-
pase-1 activation and IL-1βsecretion.
Fig 3. The NLRP7 inflammasome facilitates caspase-1 activation and IL-1βsecretion upon M.bovis infection. AB. THP-1 cells were
transfected with siRNA that targets NLRP7 or ASC, and then infected with M.bovis. Supernatant and cell lysates were analyzed by immunoblotting.
CD. Cells were stimulated with M.bovis or Pam3CSK4 for 6 h. Colocalization of NLRP7 and ASC or caspase-1 was analyzed by confocal microscopy.
Magnification, ×60.
doi:10.1371/journal.pone.0152853.g003
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 7/13
NLRP7 inflammasome activation induces M.bovis-mediated pyroptosis
Inflammasome activation is linked to caspase-1 dependent cell death called pyroptosis [23].
We evaluated cell death by LDH release, and observed that M.bovis infection led to significant
increase of LDH release, and inhibition of caspase-1 bioactivity markedly decreased this effect
(Fig 4). To evaluate whether NLRP7 inflammasome activation relates to pyroptosis, NLRP7-
and ASC-silenced cells were stimulated with M.bovis in the presence or absence of the cas-
pase-1 inhibitor, Z-YVAD-FMK. The results indicated that NLRP7 or ASC silencing attenu-
ated LDH release, but made no difference in the presence of caspase-1 inhibitor compared to
non-targeting control following infection, suggesting that NLRP7 inflammasome activation is
involved in the cell death induced by M.bovis infection which is dependent on caspase-1 (Fig
4). Cell lysis during pyroptosis results from caspase-1-mediated pore formation in the cell
membrane and subsequent influx of extracellular fluid [23]. The cytoprotective agent glycine
inhibits pyroptosis because it nonspecifically prevents ion fluxes and suppresses swelling and
lysis [24]. Addition of glycine to the culture medium substantially decreased LDH release in M.
bovis-infected cells; this was not further enhanced by silencing of NLRP7 or ASC (Fig 4).
Taken together, these data show that NLRP7 inflammasome activation contributes to pyropto-
sis induced by M.bovis infection.
NLRP7 inflammasome activated by M.bovis promotes mRNA
expression of IL-1β, TNF-αand CCL3, but inhibits IL-18 expression
Virulent mycobacteria causes a potent inflammatory response characterized by macrophage
generation of cytokines, including TNF-αand CCL3, which contributes to granuloma forma-
tion through the recruitment of more macrophages and lymphocytes [25]. To investigate
whether the NLRP7 inflammasome plays a role in the production of TNF-αand CCL3 besides
IL-1βand IL-18, we silenced NLRP7 or ASC in THP-1 macrophages prior to infection, which
Fig 4. NLRP7 inflammasome activation promotes induction of pyroptosis in M.bovis-infected THP-1 cells. Cell death was evaluated by LDH release
in NLRP7- or ASC-silenced cells stimulated with M.bovis in the presence or absence of Z-YVAD-FMK or glycine. Abbreviations: siCon, control non-targeting
siRNA; siNLRP7, NLRP7-targeting siRNA; siASC, ASC-targeting siRNA.
doi:10.1371/journal.pone.0152853.g004
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 8/13
significantly attenuated M.bovis-induced upregulation of TNF-α, CCL3 and IL-1βat the
mRNA level (Fig 5A5C). Additionally, M.bovis infection still led to obvious increases in IL-
1β, TNF-αand CCL3 mRNAs in NLRP7- and ASC-silenced cells. However, M.bovis infection
failed to change IL-18 mRNA expression, while NLRP7- or ASC-silencing led to its upregula-
tion (Fig 5D), which may be due to various sample-collection times. These data suggest that
the NLRP7 inflammasome is involved in M.bovis-induced upregulation of the mRNA expres-
sions of TNF-α, CCL3 and IL-1βas well as downregulation of the mRNA expression of IL-18.
Discussion
Previous studies described the interaction between virulent mycobacteria and their hosts. Viru-
lent mycobacteria lead to the activation of NLRP3 inflammasome through recognition of the
ESAT-6 protein [21], However, a later study suggested that NLRP3 inflammasome activation
was dispensable for the control of pulmonary tuberculosis [26]. The AIM2 inflammasome is
activated through the recognition of M.tuberculosis DNA in infected peritoneal macrophages
[20], but Shah et al. demonstrated that M.tuberculosis inhibits AIM2 inflammasome activation
in BMDCs [27]. Here we found that virulent M.bovis also activates the NLRP7 inflammasome
in THP-1 macrophages, and contributes to induction of pyroptosis and expression of TNF-α
Fig 5. NLRP7 inflammasome activation influences the expression of TNF-α, CCL3, IL-1βand IL-18 mRNA. AB. THP-1 macrophages were
transfected with non-targeting siRNA, or NLRP7-targeting siRNA, or ASC-targeting siRNA, and then infected with M.bovis. Lysates were subjected to
quantitative real-time PCR analysis.
doi:10.1371/journal.pone.0152853.g005
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 9/13
and CCL3 due to NLRP7 inflammasome activation, although the precise mechanisms respon-
sible for NLRP7 inflammasome activation in M.bovis-infected macrophages are still unclear.
The effect of NLRP7 on IL-1βsecretion is debatable; the initial in vitro investigation by
Kinoshita et al. linking NLRP7 to IL-1βsecretion was based on inflammasome reconstitution
assays in HEK293 cells, which displayed a negative regulation. NLRP7 inhibits processing of
procaspase-1 and proIL-1βand LPS-induced IL-1βsecretion through interaction with both of
these proteins [28]. Another study by Khare et al. using stably silenced targeted gene in THP-1
cells proved that NLRP7 assembles with procaspase-1 and ASC to form the inflammasome,
and contribute to increased IL-1βsecretion in response to some heat-killed bacteria. Co-
expression of NLRP7 with ASC, procaspase-1 and proIL-1βalso led to increased level of active
IL-1βrelease compared to the level of active IL-1βin the absence of NLRP7 [12], which is
opposite to the results presented by Kinoshita et al [28]. In our study, we found that knock-
down of endogenous NLRP7 in THP-1 macrophages led to reduced IL-1βsecretion in M.
bovis-infected cells. This is consistent with the data of Khare et al.[12], showing that NLRP7
positively regulates IL-1βrelease through inflammasome activation in response to M.bovis
infection.
The mechanisms involved in inflammasome activation are complicated, with some bacteria
and viruses activating more than one type of inflammasome due to their possession of different
PAMPs. For example, Listeria monocytogenes induces activation of the NLRP3, AIM2 [29],
NLRC4 [30], and NLRP7 inflammasomes within 16 hpi [12]. M.bovis infection caused upregu-
lation of NLRP3, NLRP7 and AIM2 mRNAs at different time points, which may reflect the
temporal production of specific bacterial inflammasome stimuli. Thus, the NLRP3 inflamma-
some is activated by M.tuberculosis as early as 4 hpi [26], and the role of AIM2 inflammasome
in M.tuberculosis- and M.bovis-infected macrophages were detected at 24 hpi [19,20].
NLRP7 was upregulated at the mRNA level, but not at the protein level in THP-1 macro-
phages. This may be similar to NLRP3, which is increased at the protein level in activated
RAW 264.7 cells, primary cultured astrocytes of mice, and LPS-primed bone marrow macro-
phages [3133], but not in LPS-treated THP-1 cells [34]. The reason for these differences may
be due to cell type or to the effect of PMA on protein expression [35].
Pyroptosis, a type of programmed cell death which is dependent on caspase-1, is an efficient
mechanism of intracellular bacterial clearance [36]. Here, we found that the NLRP7 inflamma-
some plays a role in the induction of pyroptosis in M.bovis-infected cells. Although proinflam-
matory cytokine release and pyroptosis are both induced by M.bovis infection, and require
caspase-1 activation, these two processes may occur independently, and previous studies dem-
onstrated that Legionella pneumophila,Burkholderia thailandensis and Salmonella typhimur-
ium that persistently expresses the flagellin protein were cleared through the pyroptosis
pathway and independently of cytokine release [36].
Granulomas are typical histopathological changes in tuberculosis, which represent a stale-
matebetween the host and the bacteria, benefitting both parties [37]. TNF-αand chemokines
play a vital role in granuloma formation and facilitate restriction of M.tuberculosis infection
[38,39]. In this study, M.bovis-induced activation of the NLRP7 inflammasome promoted the
expression of the TNF-αand CCL3. Silencing of NLRP7 or ASC did not block the induction of
TNF-αin M.bovis-infected THP-1 cells, which may be due to incomplete silencing, and/or the
existence of alternate pathways such as NF-κB signaling [40]. For example, avirulent M.bovis
BCG is unable to activate the inflammasome in the infected macrophages [26], but it still
increases the transcription of TNF-α[41].
In conclusion, we demonstrate that M.bovis infection activates the NLRP7 inflammasome,
which in turn facilitates IL-1βsecretion, induction of pyroptosis, and upregulation of TNF-α,
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 10 / 13
CCL3 and IL-1βat the mRNA level. Our study contributes to a better understanding of innate
immune response to mycobacterial infection.
Author Contributions
Conceived and designed the experiments: YZ SZAS LY ZZ XZ DZ. Performed the experiments:
YZ SZAS. Analyzed the data: YZ. Contributed reagents/materials/analysis tools: YZ SZAS LY
ZZ XZ DZ. Wrote the paper: YZ SZAS XZ.
References
1. Fend R, Geddes R, Lesellier S, Vordermeier HM, Corner LA, Gormley E, et al. Use of an electronic
nose to diagnose Mycobacterium bovis infection in badgers and cattle.J Clin Microbiol. 2005; 43
(4):174551. doi: 10.1128/JCM.43.4.1745-1751.2005 PMID: 15814995; PubMed Central PMCID:
PMC1081320.
2. Muller B, Durr S, Alonso S, Hattendorf J, Laisse CJ, Parsons SD, et al. Zoonotic Mycobacterium bovis-
induced tuberculosis in humans. Emerg Infect Dis. 2013; 19(6):899908. doi: 10.3201/eid1906.120543
PMID: 23735540.
3. Ayele WY, Neill SD, Zinsstag J, Weiss MG, Pavlik I. Bovine tuberculosis: an old disease but a new
threat to Africa. The international journal of tuberculosis and lung disease: the official journal of the
International Union against Tuberculosis and Lung Disease. 2004; 8(8):92437. PMID: 15305473.
4. Baker MG, Lopez LD, Cannon MC, De Lisle GW, Collins DM. Continuing Mycobacterium bovis trans-
mission from animals to humans in New Zealand. Epidemiol Infect. 2006; 134(5):106873. doi: 10.
1017/S0950268806005930 PMID: 16569268; PubMed Central PMCID: PMC2870481.
5. Master SS, Rampini SK, Davis AS, Keller C, Ehlers S, Springer B, et al. Mycobacterium tuberculosis
prevents inflammasome activation. Cell Host Microbe. 2008; 3(4):22432. doi: 10.1016/j.chom.2008.
03.003 PMID: 18407066; PubMed Central PMCID: PMC3657562.
6. Chen CC, Tsai SH, Lu CC, Hu ST, Wu TS, Huang TT, et al. Activation of an NLRP3 inflammasome
restricts Mycobacterium kansasii infection. PLoS One. 2012; 7(4):e36292. doi: 10.1371/journal.pone.
0036292 PMID: 22558425; PubMed Central PMCID: PMC3340363.
7. Schroder K, Tschopp J. The inflammasomes. Cell. 2010; 140(6):82132. doi: 10.1016/j.cell.2010.01.
040 WOS:000275746600007. PMID: 20303873
8. Meylan E, Tschopp J, Karin M. Intracellular pattern recognition receptors in the host response. Nature.
2006; 442(7098):3944. doi: 10.1038/nature04946 PMID: 16823444.
9. Manji GA, Wang L, Geddes BJ, Brown M, Merriam S, Al-Garawi A, et al. PYPAF1, a PYRIN-containing
Apaf1-like protein that assembles with ASC and regulates activation of NF-kappa B. The Journal of bio-
logical chemistry. 2002; 277(13):115705. doi: 10.1074/jbc.M112208200 PMID: 11786556.
10. Roberts TL, Idris A, Dunn JA, Kelly GM, Burnton CM, Hodgson S, et al. HIN-200 proteins regulate cas-
pase activation in response to foreign cytoplasmic DNA. Science. 2009; 323(5917):105760. doi: 10.
1126/science.1169841 PMID: 19131592.
11. Mariathasan S, Newton K, Monack DM, Vucic D, French DM, Lee WP, et al. Differential activation of
the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature. 2004; 430(6996):2138. doi: 10.
1038/nature02664 PMID: 15190255.
12. Khare S, Dorfleutner A, Bryan NB, Yun C, Radian AD, de Almeida L, et al. An NLRP7-containing inflam-
masome mediates recognition of microbial lipopeptides in human macrophages. Immunity. 2012; 36
(3):46476. doi: 10.1016/j.immuni.2012.02.001 WOS:000302048400017. PMID: 22361007
13. Murdoch S, Djuric U, Mazhar B, Seoud M, Khan R, Kuick R, et al. Mutations in NALP7 cause recurrent
hydatidiform moles and reproductive wastage in humans. Nat Genet. 2006; 38(3):3002. doi: 10.1038/
ng1740 PMID: 16462743.
14. Bigi F, Alito A, Romano MI, Zumarraga M, Caimi K, Cataldi A. The gene encoding P27 lipoprotein and a
putative antibiotic-resistance gene form an operon in Mycobacterium tuberculosis and Mycobacterium
bovis. Microbiology. 2000; 146 (Pt 4):10118. PMID: 10784059.
15. Bigi F, Espitia C, Alito A, Zumarraga M, Romano MI, Cravero S, et al. A novel 27 kDa lipoprotein antigen
from Mycobacterium bovis. Microbiology. 1997; 143 (Pt 11):3599605. PMID: 9387238.
16. Wiker HG, Lyashchenko KP, Aksoy AM, Lightbody KA, Pollock JM, Komissarenko SV, et al. Immuno-
chemical characterization of the MPB70/80 and MPB83 proteins of Mycobacterium bovis. Infect
Immun. 1998; 66(4):144552. PMID: 9529066; PubMed Central PMCID: PMC108073.
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 11 / 13
17. Zhou Y, Zhao D, Yue R, Khan SH, Shah SZ, Yin X, et al. Inflammasomes-dependent regulation of IL-
1beta secretion induced by the virulent Mycobacterium bovis Beijing strain in THP-1 macrophages.
Antonie van Leeuwenhoek. 2015; 108(1):16371. doi: 10.1007/s10482-015-0475-6 PMID: 25980833.
18. Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, et al. Silica crystals and aluminum
salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008; 9
(8):84756. doi: 10.1038/ni.1631 PMID: 18604214; PubMed Central PMCID: PMC2834784.
19. Yang Y, Zhou X, Kouadir M, Shi F, Ding T, Liu C, et al. the AIM2 inflammasome is involved in macro-
phage activation during infection with virulent Mycobacterium bovis strain. The Journal of infectious dis-
eases. 2013; 208(11):184958. doi: 10.1093/infdis/jit347 PMID: 23901081.
20. Saiga H, Kitada S, Shimada Y, Kamiyama N, Okuyama M, Makino M, et al. Critical role of AIM2 in
Mycobacterium tuberculosis infection. Int Immunol. 2012; 24(10):63744. doi: 10.1093/intimm/dxs062
PMID: 22695634.
21. Mishra BB, Moura-Alves P, Sonawane A, Hacohen N, Griffiths G, Moita LF, et al. Mycobacterium tuber-
culosis protein ESAT-6 is a potent activator of the NLRP3/ASC inflammasome. Cell Microbiol. 2010; 12
(8):104663. doi: 10.1111/j.1462-5822.2010.01450.x PMID: 20148899.
22. Harboe M, Oettinger T, Wiker HG, Rosenkrands I, Andersen P. Evidence for occurrence of the ESAT-6
protein in Mycobacterium tuberculosis and virulent Mycobacterium bovis and for its absence in Myco-
bacterium bovis BCG. Infect Immun. 1996; 64(1):1622. PMID: 8557334; PubMed Central PMCID:
PMC173721.
23. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nature reviews
Microbiology. 2009; 7(2):99109. doi: 10.1038/nrmicro2070 PMID: 19148178; PubMed Central
PMCID: PMC2910423.
24. von Moltke J, Trinidad NJ, Moayeri M, Kintzer AF, Wang SB, van Rooijen N, et al. Rapid induction of
inflammatory lipid mediators by the inflammasome in vivo. Nature. 2012; 490(7418):10711. doi: 10.
1038/nature11351 PMID: 22902502; PubMed Central PMCID: PMC3465483.
25. Saunders BM, Britton WJ. Life and death in the granuloma: immunopathology of tuberculosis. Immunol
Cell Biol. 2007; 85(2):10311. doi: 10.1038/sj.icb.7100027 PMID: 17213830.
26. Dorhoi A, Nouailles G, Jorg S, Hagens K, Heinemann E, Pradl L, et al. Activation of the NLRP3 inflam-
masome by Mycobacterium tuberculosis is uncoupled from susceptibility to active tuberculosis. Eur J
Immunol. 2012; 42(2):37484. doi: 10.1002/eji.201141548 PMID: 22101787.
27. Shah S, Bohsali A, Ahlbrand SE, Srinivasan L, Rathinam VAK, Vogel SN, et al. Cutting Edge: Mycobac-
terium tuberculosis but Not Nonvirulent Mycobacteria Inhibits IFN-beta and AIM2 Inflammasome-
Dependent IL-1 beta Production via Its ESX-1 Secretion System. Journal of Immunology. 2013; 191
(7):35148. doi: 10.4049/jimmunol.1301331 WOS:000324634500006.
28. Kinoshita T, Wang YT, Hasegawa M, Imamura R, Suda T. PYPAF3, a PYRIN-containing APAF-1-like
protein, is a feedback regulator of caspase-1-dependent interleukin-1 beta secretion. Journal of Biologi-
cal Chemistry. 2005; 280(23):217205. doi: 10.1074/jbc.M410057200 PMID: 15817483
29. Kim S, Bauernfeind F, Ablasser A, Hartmann G, Fitzgerald KA, Latz E, et al. Listeria monocytogenes is
sensed by the NLRP3 and AIM2 inflammasome. Eur J Immunol. 2010; 40(6):154551. doi: 10.1002/eji.
201040425 PMID: 20333626; PubMed Central PMCID: PMC3128919.
30. Sauer JD, Pereyre S, Archer KA, Burke TP, Hanson B, Lauer P, et al. Listeria monocytogenes engi-
neered to activate the Nlrc4 inflammasome are severely attenuated and are poor inducers of protective
immunity. Proc Natl Acad Sci U S A. 2011; 108(30):1241924. doi: 10.1073/pnas.1019041108 PMID:
21746921; PubMed Central PMCID: PMC3145703.
31. Loria JRD, Rohmann K, Droemann D, Kujath P, Rupp J, Goldmann T, et al. Nontypeable Haemophilus
Influenzae Infection Upregulates the NLRP3 Inflammasome and Leads to Caspase-1-Dependent
Secretion of Interleukin-1 betaA Possible Pathway of Exacerbations in COPD. Plos One. 2013; 8(6).
doi: 10.1371/journal.pone.0066818 WOS:000321424400042.
32. Alfonso-Loeches S, Urena-Peralta JR, Morillo-Bargues MJ, Oliver-De La Cruz J, Guerri C. Role of mito-
chondria ROS generation in ethanol-induced NLRP3 inflammasome activation and cell death in astro-
glial cells. Front Cell Neurosci. 2014; 8. doi: 10.3389/Fncel.2014.00216 WOS:000339934400001.
33. Schroder K, Sagulenko V, Zamoshnikova A, Richards AA, Cridland JA, Irvine KM, et al. Acute lipopoly-
saccharide priming boosts inflammasome activation independently of inflammasome sensor induction.
Immunobiology. 2012; 217(12):13259. doi: 10.1016/j.imbio.2012.07.020 WOS:000313925100013.
PMID: 22898390
34. Wang JG, Williams JC, Davis BK, Jacobson K, Doerschuk CM, Ting JPY, et al. Monocytic microparti-
cles activate endothelial cells in an IL-1 beta-dependent manner. Blood. 2011; 118(8):236674. doi: 10.
1182/blood-2011-01-330878 WOS:000294258000041. PMID: 21700772
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 12 / 13
35. Haneklaus M, Gerlic M, Kurowska-Stolarska M, Rainey AA, Pich D, McInnes IB, et al. Cutting edge:
miR-223 and EBV miR-BART15 regulate the NLRP3 inflammasome and IL-1beta production. J Immu-
nol. 2012; 189(8):37959. doi: 10.4049/jimmunol.1200312 PMID: 22984081.
36. Miao EA, Leaf IA, Treuting PM, Mao DP, Dors M, Sarkar A, et al. Caspase-1-induced pyroptosis is an
innate immune effector mechanism against intracellular bacteria. Nat Immunol. 2010; 11(12):113642.
doi: 10.1038/ni.1960 PMID: 21057511; PubMed Central PMCID: PMC3058225.
37. Philips JA, Ernst JD. Tuberculosis pathogenesis and immunity. Annual review of pathology. 2012;
7:35384. doi: 10.1146/annurev-pathol-011811-132458 PMID: 22054143.
38. Algood HM, Chan J, Flynn JL. Chemokines and tuberculosis. Cytokine Growth Factor Rev. 2003; 14
(6):46777. PMID: 14563349.
39. Saunders BM, Tran S, Ruuls S, Sedgwick JD, Briscoe H, Britton WJ. Transmembrane TNF is sufficient
to initiate cell migration and granuloma formation and provide acute, but not long-term, control of Myco-
bacterium tuberculosis infection. Journal of Immunology. 2005; 174(8):48529.
WOS:000228234600052.
40. Lee HM, Kang J, Lee SJ, Jo EK. Microglial activation of the NLRP3 inflammasome by the priming sig-
nals derived from macrophages infected with mycobacteria. Glia. 2013; 61(3):44152. doi: 10.1002/
glia.22448 PMID: 23280493.
41. Segueni N, Vigne S, Palmer G, Bourigault ML, Olleros ML, Vesin D, et al. Limited Contribution of IL-36
versus IL-1 and TNF Pathways in Host Response to Mycobacterial Infection. Plos One. 2015; 10(5).
doi: 10.1371/journal.pone.0126058 WOS:000354214400064.
Mycobacterium bovis Activates the NLRP7 Inflammasome
PLOS ONE | DOI:10.1371/journal.pone.0152853 April 4, 2016 13 / 13
... In terms of the pyroptosis regulator NLRP7, our results suggested that NLRP7 may play a prognostic protective role in CRC. NLR containing NLRP7 was found to assemble and activate the inflammasome complex, including caspase-1, to promote the expression of tumor necrosis factor-α and interleukin-1β and induce pyroptosis [62][63][64]. This regulatory mechanism can be observed in human macrophages [65], and the macrophage infiltration is believed to differ markedly between left-and right-sided CRC and affect the tumor microenvironment of CRC. ...
Article
Full-text available
Background Colorectal cancer (CRC) is one of the most common malignancies, and pyroptosis exerts an immunoregulatory role in CRC. Although the location of the primary tumor is a prognostic factor for patients with CRC, the mechanisms of pyroptosis in left- and right-sided CRC remain unclear. Methods Expression and clinical data were collected from The Cancer Genome Atlas and Gene Expression Omnibus databases. Differences in clinical characteristics, immune cell infiltration, and somatic mutations between left- and right-sided CRC were then compared. After screening for differentially expressed genes, Pearson correlation analysis was performed to select pyroptosis-related genes, followed by a gene set enrichment analysis. Univariate and multivariate Cox regression analyses were used to construct and validate the prognostic model and nomogram for predicting prognosis. Collected left- and right-sided CRC samples were subjected to reverse transcription-quantitative polymerase chain reaction (RT-qPCR) to validate the expression of key pyroptosis-related genes. Results Left- and right-sided CRC exhibited significant differences in clinical features and immune cell infiltration. Five prognostic signatures were identified from among 134 pyroptosis-related differentially expressed genes to construct a risk score-based prognostic model, and adverse outcomes for high-risk patients were further verified using an external cohort. A nomogram was also generated based on three independent prognostic factors to predict survival probabilities, while calibration curves confirmed the consistency between the predicted and actual survival. Experiment data confirmed the significant differential expression of five genes between left- and right-sided CRC. Conclusion The five identified pyroptosis-related gene signatures may be potential biomarkers for predicting prognosis in left- and right-sided CRC and may help improve the clinical outcomes of patients with CRC.
... Several other NLRPs are reported to form inflammasomes, including NLRP7, NLRP9, and NLRP12. Human NLRP7 was previously reported to interact with ASC to assemble an inflammasome (41)(42)(43) but failed to do so in our screen of NLRPs. The murine homolog of human NLRP9, NLRP9B, is proposed to form an ASC inflammasome (44,45), but in humans, the NLRP9 PYD is incapable of inducing ASC polymerization (46). ...
Article
Inflammasomes are multiprotein complexes that drive inflammation and contribute to protective immunity against pathogens and immune pathology in autoinflammatory diseases. Inflammasomes assemble when an inflammasome scaffold protein senses an activating signal and forms a signaling platform with the inflammasome adaptor protein ASC. The NLRP subfamily of NOD-like receptors (NLRs) includes inflammasome nucleators (such as NLRP3) and also NLRP12, which is genetically linked to familial autoinflammatory disorders that resemble diseases caused by gain-of-function NLRP3 mutants that generate a hyperactive NLRP3 inflammasome. We performed a screen to identify ASC inflammasome–nucleating proteins among NLRs that have the canonical pyrin-NACHT-LRR domain structure. Only NLRP3 and NLRP6 could initiate ASC polymerization to form “specks,” and NLRP12 failed to nucleate ASC polymerization. However, wild-type NLRP12 inhibited ASC inflammasome assembly induced by wild-type and gain-of-function mutant NLRP3, an effect not seen with disease-associated NLRP12 mutants. The capacity of NLRP12 to suppress NLRP3 inflammasome assembly was limited to human NLRP3 and was not observed for wild-type murine NLRP3. Furthermore, peripheral blood mononuclear cells from patients with an NLRP12 mutant–associated inflammatory disorder produced increased amounts of the inflammatory cytokine IL-1β in response to NLRP3 stimulation. Thus, our findings provide insights into NLRP12 biology and suggest that NLRP3 inhibitors in clinical trials for NLRP3-driven diseases may also be effective in treating NLRP12-associated autoinflammatory diseases.
... 36 Zhou et al reported that NLRP7 inflammasome could promote IL-1β secretion and induce pyroptosis in THP-1 macrophages in response to M. bovis infection. 37 Collectively, the anti-inflammatory or pro-inflammatory role of NLRP7 was observed under specific conditions. Previous studies on genetic polymorphisms of NLRP7 mainly focused on reproductive diseases, such as placental development, early pregnancy, and hydatidiform mole. ...
Article
Full-text available
Background: Cancer development and tumor immune microenvironment remodeling are closely linked to pyroptosis and inflammasome activation. However, little information is available in single nucleotide polymorphisms (SNPs) in pyroptosis and inflammasome-related genes in patients with lung cancer. This study aims to evaluate the associations between pyroptosis-related gene (NLRP3, NLRC4, and NLRP7) polymorphisms and the risk of lung cancer. Methods: The MassARRAY platform was used to genotype six SNPs of the NLRP3, NLRC4, and NLRP7 genes in 660 lung cancer cases and 660 controls. Results: Individuals with rs35829419-A, rs385076-C, and rs775882-T alleles exhibited a higher risk of lung cancer (p < 0.01), while rs212704-T appears protective (p = 0.006). The rs35829419-AA, rs385076-TC/CC, and rs775882-CT/TT genotypes were associated with various degrees of elevated risk of lung cancer (p<0.02), whereas rs212704-TT was associated with a reduced risk of the disease (p=0.014). Genetic models analysis showed that rs35829419, rs385076, and rs775882 was associated with an increased risk of lung cancer, while rs212704 was related to a reduced risk in all three models (p < 0.05). The four SNPs remained significant in smoker and nonsmoker subgroups (p < 0.05). However, rs35829419 was correlated with risk of adenocarcinoma and small cell lung cancer, and rs212704 was only protective for squamous cell carcinoma. The rs385076 and rs775882 were associated with all three pathological types (p < 0.01). Conclusion: Besides providing candidate markers for identification of high-risk populations and early prevention of the disease, our research also provided new insight into anti-tumor strategies targeting inflammasomes and pyroptosis.
... Several inflammasomes can detect mycobacteria and induce the production of IL-1β. Some reports show that non-tuberculosis mycobacteria activate the NLRP3 inflammasome [32][33][34], whereas bacteria from the tuberculosis complex activate the NLPR3, NLRP7, or AIM2 inflammasomes [9,26,27,35]. In this study, we evaluated the participation of the NLRP3 inflammasome because it has previously been shown to be activated by different mycobacteria in different macrophage models. ...
Article
Full-text available
Mycobacterium bovis is a facultative intracellular bacterium that produces cellular necrosis in granulomatous lesions in bovines. Although M. bovis-induced inflammation actively participates in granuloma development, its role in necrotic cell death and in bovine macrophages has not been fully explored. In this study, we evaluate the effect of M. bovis AN5 and its culture filtrate protein extract (CFPE) on inflammasome activation in bovine macrophages and its consequences on cell death. Our results show that both stimuli induce necrotic cell death starting 4 h after incubation. CFPE treatment and M. bovis infection also induce the maturation of IL-1β (>3000 pg/mL), oligomerization of ASC (apoptosis-associated speck-like protein containing CARD), and activation of caspase-1, following the canonical activation pathway of the NLRP3 inflammasome. Inhibiting the oligomerization of NLRP3 and caspase-1 decreases necrosis among the infected or CFPE-stimulated macrophages. Furthermore, histological lymph node sections of bovines naturally infected with M. bovis contained cleaved gasdermin D, mainly in macrophages and giant cells within the granulomas. Finally, the induction of cell death (apoptosis and pyroptosis) decreased the intracellular bacteria count in the infected bovine macrophages, suggesting that cell death helps to control the intracellular growth of the mycobacteria. Our results indicate that M. bovis induces pyroptosis-like cell death that is partially related to the NLRP3 inflammasome activation and that the cell death process could control bacterial growth.
... 87,88 Various mechanisms have been proposed for this regulation of IL-1b, including direct interaction with and inhibition of inflammasome components, and alteration of pro-IL-1b transcription and subsequent trafficking and release of the mature forms, 89 although further validation is required for this, as various reports have also shown a lack of effect of NLRP7 on inflammatory responses. 77,82,84,90 Taken together, these studies illustrate a multifaceted role for the NLRP7 inflammasome in the regulation of inflammatory responses. Taken together, these studies illustrate a multifaceted role for the NLRP7 in the regulation of inflammatory responses, wherein it may negatively regulate the inflammasome in the resting state but assemble an inflammasome following a stimulus such as bacterial infection. ...
Article
Full-text available
Inflammasomes are assembled by innate immune sensors that cells employ to detect a range of danger signals and respond with pro-inflammatory signalling. Inflammasomes activate inflammatory caspases, which trigger a cascade of molecular events with the potential to compromise cellular integrity and release the IL-1β and IL-18 pro-inflammatory cytokines. Several molecular mechanisms, working in concert, ensure that inflammasome activation is tightly regulated; these include NLRP3 post-translational modifications, ubiquitination and phosphorylation, as well as single-domain proteins that competitively bind to key inflammasome components, such as the CARD-only proteins (COPs) and PYD-only proteins (POPs). These diverse regulatory systems ensure that a suitable level of inflammation is initiated to counteract any cellular insult, while simultaneously preserving tissue architecture. When inflammasomes are aberrantly activated can drive excessive production of pro-inflammatory cytokines and cell death, leading to tissue damage. In several autoinflammatory conditions, inflammasomes are aberrantly activated with subsequent development of clinical features that reflect the degree of underlying tissue and organ damage. Several of the resulting disease complications may be successfully controlled by anti-inflammatory drugs and/or specific cytokine inhibitors, in addition to more recently developed small-molecule inhibitors. In this review, we will explore the molecular processes underlying the activation of several inflammasomes and highlight their role during health and disease. We also describe the detrimental effects of these inflammasome complexes, in some pathological conditions, and review current therapeutic approaches as well as future prospective treatments.
... In a more functional view, the NLRP7 inflammasome has been described as being involved in the activation of caspase-1 and IL1β release following infection of THP-1 cells (monocytes cell line) by Mycobacterium bovis. This activation of the NLRP7 inflammasome induced cell death, which the authors suggested to be pyroptosis [148]. Another team focused on the posttranslational modifications regulating NLRP7. ...
Article
Full-text available
Preterm prelabor ruptures of fetal membranes (pPROM) are a pregnancy complication responsible for 30% of all preterm births. This pathology currently appears more as a consequence of early and uncontrolled process runaway activation, which is usually implicated in the physiologic rupture at term: inflammation. This phenomenon can be septic but also sterile. In this latter case, the inflammation depends on some specific molecules called “alarmins” or “damage-associated molecular patterns” (DAMPs) that are recognized by pattern recognition receptors (PRRs), leading to a microbial-free inflammatory response. Recent data clarify how this activation works and which receptor translates this inflammatory signaling into fetal membranes (FM) to manage a successful rupture after 37 weeks of gestation. In this context, this review focused on two PRRs: the receptor for advanced glycation end-products (RAGE) and the NLRP7 inflammasome.
... Western blot analysis was performed as previously described [40]. Briefly, an equal amount of samples was loaded onto SDS-PAGE, and proteins were transferred onto PVDF membranes. ...
Article
Objective: Bovine tuberculosis (bTB) is a chronic respiratory infectious disease of domestic livestock caused by intracellular Mycobacterium bovis infection, which causes ~$3 billion in annual losses to global agriculture. Providing novel tools for bTB management require a comprehensive understanding of the molecular regulatory mechanisms underlying the M. bovis infection. Nevertheless, a combination of different bioinformatics and systems biology methods was used in this study in order to clearly understand the molecular regulatory mechanisms of bTB, especially the immunomodulatory mechanisms of M. bovis infection. Methods: RNA-seq data were retrieved and processed from 78 (39 noninfected control vs. 39 M. bovis-infected samples) bovine alveolar macrophages (bAMs). Next, weighted gene co-expression network analysis (WGCNA) was performed to identify the co-expression modules in non-infected control bAMs as a reference set. The WGCNA module preservation approach was then used to identify non-preserved modules between non-infected controls and M. bovis-infected samples (test set). Additionally, functional enrichment analysis was used to investigate the biological behavior of the non-preserved modules and to identify bTB-specific non-preserved modules. Co-expressed hub genes were identified based on module membership (MM) criteria of WGCNA in the non-preserved modules and then integrated with protein–protein interaction (PPI) networks to identify co-expressed hub genes/transcription factors (TFs) with the highest maximal clique centrality (MCC) score (hub-central genes). Results: As a result, WGCNA analysis led to the identification of 21 modules in the non-infected control bAMs (reference set), among which the topological properties of 14 modules were altered in the M. bovis-infected bAMs (test set). Interestingly, 7 of the 14 non-preserved modules were directly related to the molecular mechanisms underlying the host immune response, immunosuppressive mechanisms of M. bovis, and bTB development. Moreover, among the co-expressed hub genes and TFs of the bTB-specific non-preserved modules, 260 genes/TFs had double centrality in both co-expression and PPI networks and played a crucial role in bAMs-M. bovis interactions. Some of these hub-central genes/TFs, including PSMC4, SRC, BCL2L1, VPS11, MDM2, IRF1, CDKN1A, NLRP3, TLR2, MMP9, ZAP70, LCK, TNF, CCL4, MMP1, CTLA4, ITK, IL6, IL1A, IL1B, CCL20, CD3E, NFKB1, EDN1, STAT1, TIMP1, PTGS2, TNFAIP3, BIRC3, MAPK8, VEGFA, VPS18, ICAM1, TBK1, CTSS, IL10, ACAA1, VPS33B, and HIF1A, had potential targets for inducing immunomodulatory mechanisms by M. bovis to evade the host defense response. Conclusion: The present study provides an in-depth insight into the molecular regulatory mechanisms behind M. bovis infection through biological investigation of the candidate non-preserved modules directly related to bTB development. Furthermore, several hub-central genes/TFs were identified that were significant in determining the fate of M. bovis infection and could be promising targets for developing novel anti-bTB therapies and diagnosis strategies.
Article
Full-text available
Objective: Bovine tuberculosis (bTB) is a chronic respiratory infectious disease of domestic livestock caused by intracellular Mycobacterium bovis infection, which causes approximately $3 billion in annual losses to global agriculture. A combination of different bioinformatics and systems biology methods was used in this study in order to clearly understand the molecular regulatory mechanisms of bTB, especially the immunomodulatory mechanisms of M. bovis infection. Methods: RNA-seq data were retrieved and processed from 78 (39 non-infected control vs 39 M. bovis-infected samples) bovine alveolar macrophages (bAMs). Next, weighted gene co-expression network analysis (WGCNA) was performed to identify the co-expression modules in non-infected control bAMs as reference set. Co-expressed hub genes were identified based on module membership (MM) criteria of WGCNA in the non-preserved modules and then integrated with protein-protein interaction (PPI) networks to identify co-expressed hub genes/transcription factors (TFs) with the highest maximal clique centrality (MCC) score (hub-central genes). Results: As result, WGCNA analysis led to the identification of 21 modules in the non-infected control bAMs (reference set), among which the topological properties of 14 modules were altered in the M. bovis-infected bAMs (test set). Interestingly, 7 of the 14 non-preserved modules were directly related to the molecular mechanisms underlying the host immune response, immunosuppressive mechanisms of M. bovis, and bTB development. Moreover, among the co-expressed hub genes and TFs of the bTB-specific non-preserved modules, 260 genes/TFs had double centrality in both co-expression and PPI networks and played a crucial role in bAMs-M. bovis interactions. Some of these hub-central genes/TFs, including PSMC4, SRC, BCL2L1, VPS11, MDM2, IRF1, CDKN1A, NLRP3, TLR2, MMP9, ZAP70, LCK, TNF, CCL4, MMP1, CTLA4, ITK, IL6, IL1A, IL1B, CCL20, CD3E, NFKB1, EDN1, STAT1, TIMP1, PTGS2, TNFAIP3, BIRC3, MAPK8, VEGFA, VPS18, ICAM1, TBK1, CTSS, IL10, ACAA1, VPS33B, and HIF1A, had potential targets for inducing immunomodulatory mechanisms by M. bovis to evade the host defense response. Conclusion: The present study provides an in-depth insight into the molecular regulatory mechanisms behind M. bovis infection through biological investigation of the candidate non-preserved modules directly related to bTB development.
Article
Nucleotide-binding oligomerization domain (NOD) and leucine-rich repeat (LRR)-containing receptors or NOD-like receptors (NLRs) are cytosolic pattern recognition receptors which sense conserved microbial patterns and host-derived danger signals to elicit innate immune responses. The activation of several prototypic NLRs, including NLR and pyrin domain (PYD) containing (NLRP) 1, NLRP3 and NLR and caspase recruitment domain (CARD) containing (NLRC) 4, results in the assembly of inflammasomes which are large, cytoplasmic multi-protein signaling platforms responsible for the maturation and release of the pro-inflammatory cytokines IL-1β and IL-18, and for the induction of a specialized form of inflammatory cell death called pyroptosis. However, the function of other members of the NLR family, including NLRP7, are less well understood. NLRP7 has been linked to innate immune signaling, but its precise role is still controversial as it has been shown to positively and negatively affect inflammasome responses. Inflammasomes are essential for homeostasis and host defense, but inappropriate inflammasome responses due to hereditary mutations and somatic mosaicism in inflammasome components and defective regulation has been linked to a broad spectrum of human diseases. A compelling connection between NLRP7 mutations and reproductive diseases, and in particular molar pregnancy, has been established. Though, the molecular mechanisms by which NLRP7 mutations contribute to reproductive diseases are largely unknown. In this review, we focus on NLRP7 and discuss the current evidence of its role in inflammasome regulation as well as its implication in human reproductive diseases.
Article
Full-text available
Mycobacterium bovis is the causative agent of tuberculosis in cattle. Infection of macro-phages with M. bovis leads to the activation of the ''nucleotide binding and oligomerization, leucine-rich repeat and pyrin domains-containing protein 3'' (NLRP3) and ''absent in melanoma 2'' (AIM2) inflammasomes, which in turn triggers release of the proinflammatory cytokine interleukin-1b (IL-1b) that contributes to bacterial clearance and plays a crucial role in the host defense. However, NLRP3 and AIM2 inflammasome activation is influenced by several factors and how IL-1b secretion by M. bovis-infected macrophages is regulated via the inflammasome pathway remains unclear. Here we found that IL-1b secretion and pro-IL-1b protein accumulation were inhibited in THP-1 macrophages upon exposure to the virulent M. bovis Beijing strain in the presence of high K ? concentrations, cycloheximide (a protein synthesis inhibitor) and PR-619 (a deubiquitinating enzyme inhibitor). Scavenging reactive oxygen species (ROS) induced by N-acetylcysteine reduced IL-1b release independent of the mitochondrial permeability transition. Collectively, our results suggest that IL-1b secretion by M. bovis-infected THP-1 macrophages is reduced by high extracellular K ? concentration, inhibition of new protein synthesis, deubiquitination, and ROS generation. Keywords Inflammasome Á Mycobacterium bovis Á Cycloheximide Á PR-619 Á ROS
Article
Full-text available
IL-36 cytokines are members of the IL-1 family of cytokines that stimulate dendritic cells and T cells leading to enhanced T helper 1 responses in vitro and in vivo; however, their role in host defense has not been fully addressed thus far. The objective of this study was to examine the role of IL-36R signaling in the control of mycobacterial infection, using models of systemic attenuated M. bovis BCG infection and virulent aerogenic M. tuberculosis infection. IL-36γ expression was increased in the lung of M. bovis BCG infected mice. However, IL-36R deficient mice infected with M. bovis BCG showed similar survival and control of the infection as compared to wild-type mice, although their lung pathology and CXCL1 response were transiently different. While highly susceptible TNF-α deficient mice succumbed with overwhelming M. tuberculosis infection, and IL-1RI deficient mice showed intermediate susceptibility, IL-36R-deficient mice controlled the infection, with bacterial burden, lung inflammation and pathology, similar to wild-type controls. Therefore, IL-36R signaling has only limited influence in the control of mycobacterial infection.
Article
Full-text available
Toll-like receptors (TLRs) and Nod-like receptors (NLRs) are innate immunity sensors that provide an early/effective response to pathogenic or injury conditions. We have reported that ethanol-induced TLR4 activation triggers signaling inflammatory responses in glial cells, causing neuroinflammation and brain damage. However, it is uncertain if ethanol is able to activate NLRs /inflammasome in astroglial cells, which is the mechanism of activation, and whether there is crosstalk between both immune sensors in glial cells. Here we show that chronic ethanol treatment increases the co-localization of caspase-1 with GFAP+ cells, and up-regulates IL-1β and IL-18 in the frontal medial cortex in WT, but not in TLR4 knock-out mice. We further show that cultured cortical astrocytes expressed several inflammasomes (NLRP3, AIM2, NLRP1 and IPAF), although NLRP3 mRNA is the predominant form. Ethanol, as ATP and LPS treatments, up-regulates NLRP3 expression, and causes caspase-1 cleavage and the release of IL-1β and IL-18 in astrocytes supernatant. Ethanol-induced NLRP3/caspase-1 activation is mediated by mitochondrial (m) ROS generation because when using a specific mitochondria ROS scavenger, the mito-TEMPO (500 M) or NLRP3 blocking peptide (4g/ml) or a specific caspase-1 inhibitor, Z-YVAD-FMK (10 M), abrogates mROS release and reduces the up-regulation of IL-1β and IL-18 induced by ethanol or LPS or ATP. Confocal microscopy studies further confirm that ethanol, ATP or LPS promotes NLRP3/caspase-1 complex recruitment within the mitochondria to promote cell death by caspase-1-mediated pyroptosis, which accounts for ≈ 73 % of total cell death (≈22%) and the remaining (≈25%) die by caspase-3-dependent apoptosis. Suppression of the TLR4 function abrogates most ethanol effects on NLRP3 activation and reduces cell death. These findings suggest that NLRP3 participates, in ethanol-induced neuroinflammation and highlight the NLRP3/TLR4 crosstalk in ethanol-induced brain injury.
Article
Full-text available
Mycobacterium tuberculosis extracellular DNA gains access to the host cell cytosol via the ESX-1 secretion system. It is puzzling that this extracellular DNA of M. tuberculosis does not induce activation of the absent in melanoma 2 (AIM2) inflammasome because AIM2 recognizes cytosolic DNA. In this study, we show that nonvirulent mycobacteria such as Mycobacterium smegmatis induce AIM2 inflammasome activation, which is dependent on their strong induction of IFN-β production. In contrast, M. tuberculosis, but not an ESX-1-deficient mutant, inhibits the AIM2 inflammasome activation induced by either M. smegmatis or transfected dsDNA. The inhibition does not involve changes in host cell AIM2 mRNA or protein levels but led to decreased activation of caspase-1. We furthermore demonstrate that M. tuberculosis inhibits IFN-β production and signaling, which was partially responsible for the inhibition of AIM2 activation. In conclusion, we report a novel immune evasion mechanism of M. tuberculosis that involves the ESX-1-dependent, direct or indirect, suppression of the host cell AIM2 inflammasome activation during infection.
Article
Full-text available
Background: Mycobacterium bovis, the causative agent of bovine tuberculosis, infects host macrophages and triggers production of the proinflammatory cytokine interleukin 1β (IL-1β). The mechanism by which macrophages become activated and secrete IL-1β in tuberculosis has not yet been elucidated. Methods: In this study, we investigated the role of the absence in melanoma 2 (AIM2) inflammasome in IL-1β release from macrophages infected with pathogenic M. bovis strain. Results: We found that the AIM2 inflammasome activation is involved in the production of IL-1β in primary and immortalized mouse macrophage upon M. bovis infection; that the activation process requires cytoplasmic potassium efflux, mycobacterial internalization, but not reactive oxygen species (ROS) or IFN-β release; that the AIM2 inflammasome contributes to the synthesis of proinflammatory and chemotatic factors in M. bovis-infected macrophages; and that the activation of the AIM2 inflammasome is due, at least in part, to mycobacterial translocation into the cytosol. Conclusions: We conclude that the AIM2 inflammasome is involved in macrophage activation during infection with virulent M. bovis strain. To our knowledge, this is the first evidences for the involvement of the AIM2 inflammasome in M. bovis infection.
Article
Full-text available
Nontypeable Haemophilus influenzae (NTHi) is the most common cause for bacterial exacerbations in chronic obstructive pulmonary disease (COPD). Recent investigations suggest the participation of the inflammasome in the pathomechanism of airway inflammation. The inflammasome is a cytosolic protein complex important for early inflammatory responses, by processing Interleukin-1β (IL-1β) to its active form. Since inflammasome activation has been described for a variety of inflammatory diseases, we investigated whether this pathway plays a role in NTHi infection of the airways. A murine macrophage cell line (RAW 264.7), human alveolar macrophages and human lung tissue (HLT) were stimulated with viable or non-viable NTHi and/or nigericin, a potassium ionophore. Secreted cytokines were measured with ELISA and participating proteins detected via Western Blot or immunohistochemistry. Western Blot analysis of cells and immunohistochemistry of lung tissue detected the inflammasome key components NLRP3 and caspase-1 after stimulation, leading to a significant induction of IL-1β expression (RAW: control at the lower detection limit vs. NTHi 505±111pg/ml, p<0.01). Inhibition of caspase-1 in human lung tissue led to a significant reduction of IL-1β and IL-18 levels (IL-1β: NTHi 24 h 17423±3198pg/ml vs. NTHi+Z-YVAD-FMK 6961±1751pg/ml, p<0.01). Our data demonstrate the upregulation of the NRLP3-inflammasome during NTHi-induced inflammation in respiratory cells and tissues. Our findings concerning caspase-1 dependent IL-1β release suggest a role for the inflammasome in respiratory tract infections with NTHi which may be relevant for the pathogenesis of bacterial exacerbations in COPD.
Article
Full-text available
We aimed to estimate the global occurrence of zoonotic tuberculosis (TB) caused by Mycobacterium bovis or M. caprae infections in humans by performing a multilingual, systematic review and analysis of relevant scientific literature of the last 2 decades. Although information from many parts of the world was not available, data from 61 countries suggested a low global disease incidence. In regions outside Africa included in this study, overall median proportions of zoonotic TB of ≤1.4% in connection with overall TB incidence rates ≤71/100,000 population/year suggested low incidence rates. For countries of Africa included in the study, we multiplied the observed median proportion of zoonotic TB cases of 2.8% with the continental average overall TB incidence rate of 264/100,000 population/year, which resulted in a crude estimate of 7 zoonotic TB cases/100,000 population/year. These generally low incidence rates notwithstanding, available data indicated substantial consequences of this disease for some population groups and settings.
Article
Full-text available
Although microRNA (miRNA) regulation of TLR signaling is well established, this has not yet been observed for NLR proteins or the inflammasomes they form. We have now validated a highly conserved miR-223 target site in the NLRP3 3'-untranslated region. miR-223 expression decreases as monocytes differentiate into macrophages, whereas NLRP3 protein increases during this time. However, overexpression of miR-223 prevents accumulation of NLRP3 protein and inhibits IL-1β production from the inflammasome. Virus inhibition of the inflammasome is an emerging theme, and we have also identified an EBV miRNA that can target the miR-223 binding site in the NLRP3 3'-untranslated region. Furthermore, this virus miRNA can be secreted from infected B cells via exosomes to inhibit the NLRP3 inflammasome in noninfected cells. Therefore, we have identified both the first endogenous miRNA that limits NLRP3 inflammatory capacity during myeloid cell development and also a viral miRNA that takes advantage of this, limiting inflammation for its own purposes.
Article
The inflammasome is a multimolecular complex that orchestrates the activation of proinflammatory caspases and interleukin (IL)-1β, which is generally increased in the cerebrospinal fluids of patients with tuberculous meningitis. However, it has not been clarified whether mycobacteria can activate the inflammasome and induce IL-1β maturation in microglia. In this study, we found that the priming of primary murine microglial cells with conditioned media from cultures of macrophages infected with Mycobacterium tuberculosis (Mtb) led to robust activation of caspase-1 and IL-1β secretion after Mtb stimulation. Potassium efflux and the lysosomal proteases cathepsin B and cathepsin L were required for the Mtb-induced caspase-1 activation and maturation of IL-1β production in primed microglia. Mtb-induced IL-1β maturation was also found to depend on the nucleotide binding and oligomerization of domain-like receptor family pyrin domain containing 3 protein (NLRP3) and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), as well as the generation of mitochondrial reactive oxygen species (ROS). Notably, the priming of microglia with tumor necrosis factor-α or oncostatin M resulted in caspase-1 cleavage and IL-1β secretion in response to Mtb. Moreover, dexamethasone, as an adjunctive therapy for patients of tuberculous meningitis, significantly reduced the Mtb-induced maturation of IL-1β through inhibition of mitochondrial ROS generation. Collectively, these data suggest that Mtb stimulation induces activation of the microglial NLRP3 inflammasome (composed of NLRP3, ASC, and cysteine protease caspase-1) through microglia-leukocyte interactions as a priming signal, and that dexamethasone decreases inflammasome activation through inhibition of ROS of mitochondrial origin. © 2012 Wiley Periodicals, Inc.