ArticlePDF Available

Cutting Edge: Alum Adjuvant Stimulates Inflammatory Dendritic Cells through Activation of the NALP3 Inflammasome

Authors:

Abstract and Figures

Adjuvants are vaccine additives that stimulate the immune system without having any specific antigenic effect of itself. In this study we show that alum adjuvant induces the release of IL-1beta from macrophages and dendritic cells and that this is abrogated in cells lacking various NALP3 inflammasome components. The NALP3 inflammasome is also required in vivo for the innate immune response to OVA in alum. The early production of IL-1beta and the influx of inflammatory cells into the peritoneal cavity is strongly reduced in NALP3-deficient mice. The activation of adaptive cellular immunity to OVA-alum is initiated by monocytic dendritic cell precursors that induce the expansion of Ag-specific T cells in a NALP3-dependent way. We propose that, in addition to TLR stimulators, agonists of the NALP3 inflammasome should also be considered as vaccine adjuvants.
Content may be subject to copyright.
Cutting Edge
Cutting Edge
Cutting Edge: Alum Adjuvant Stimulates Inflammatory
Dendritic Cells through Activation of the NALP3
Inflammasome
Mirjam Kool,* Virginie Pe´trilli,
Thibaut De Smedt,
Aline Rolaz,
Hamida Hammad,*
§
Menno van Nimwegen,* Ingrid M. Bergen,* Rosa Castillo,
Bart N. Lambrecht,
1,2
*
§
and
Ju¨rg Tschopp
1,2†
Adjuvants are vaccine additives that stimulate the im-
mune system without having any specific antigenic ef-
fect of itself. In this study we show that alum adjuvant
induces the release of IL-1
from macrophages and den-
dritic cells and that this is abrogated in cells lacking var-
ious NALP3 inflammasome components. The NALP3
inflammasome is also required in vivo for the innate im-
mune response to OVA in alum. The early production
of IL-1
and the influx of inflammatory cells into the
peritoneal cavity is strongly reduced in NALP3-defi-
cient mice. The activation of adaptive cellular immunity
to OVA-alum is initiated by monocytic dendritic cell
precursors that induce the expansion of Ag-specific T
cells in a NALP3-dependent way. We propose that, in
addition to TLR stimulators, agonists of the NALP3 in-
flammasome should also be considered as vaccine
adjuvants. The Journal of Immunology, 2008, 181:
3755–3759.
Activationof dendritic cells (DCs)
3
is a crucial mecha-
nism by which vaccine adjuvants stimulate protec-
tive adaptive immunity to cancer or microbial in-
fection. Agonists to several TLRs are in clinical development
as adjuvants (1). The most promising TLR ligand is an un-
methylated CpG dinucleotide containing DNA that stimu-
lates TLR9 in a pathway requiring the adaptor MyD88, lead-
ing to the activation of DCs (2, 3). Nonetheless, there is a
debate as to whether TLR stimulation is essential for the Ab-
enhancing effect of vaccine adjuvants like alum. Although
initial studies showed that MyD88- deficient mice are defec-
tive in mounting Th1 and B cell responses (4), this notion
has been recently challenged (5). It has also become clear
that an alternative pathogen detection system exists, in ad-
dition to TLRs, that relies on a family of intracellular recep-
tors called nucleotide-binding oligomerization domain-like
receptors (NLRs) (6 8). In addition to pathogen-associated
molecular patterns, some of these NLRs also sense the pres-
ence of endogenous danger signals. The most studied NLR
member is NALP3, which, together with Cardinal and apo-
ptosis-associated speck-like protein containing a caspase re-
cruitment domain (ASC), forms a caspase-1 activating com-
plex, the so-called inflammasome (9). This complex is
activated by multiple agonists including a bacterially derived
muramyl dipeptide or endogenous ATP and uric acid, lead-
ing to the processing and release of IL-1
(10, 11). We there-
fore considered the possibility that NLRs, in addition to
TLRs, are implicated in the effect of some adjuvants.
Materials and Methods
Mice
Wild-type (WT), ASC
/
, IPAF
/
(gifts of V. Dixit, Genetech; IPAF is IL-
1
-converting enzyme protease activating factor), MyD88
/
, and
NALP3
/
mice (12) (all mice in C57BL/6 background) were bred at the Uni-
versity of Lausanne, Lausanne, Switzerland and at the University Hospital, Gh-
ent, Belgium. Male and female mice were used between the ages of 8 and 14 wk.
All in vivo experiments were approved by the animal ethics committee of Ghent
University.
Isolation and activation of mouse primary cells
Peritoneal macrophages were isolated 3 days after an i.p. injection with 10%
thioglycollate and primed in vitro with LPS as previously described (13). Uric
acid, uricase, and cytochalasin D were bought from Sigma-Aldrich and crystals
were generated as previously described (12).
Ags and adjuvant
OVA was purchased from Worthington Biochemical. In Ag-tracking experi-
ments, OVA-Alexa Fluor 647 (Molecular Probes) was used. Imject Alum
(Pierce Biochemicals; hereafter simply called alum) is a mixture of aluminum
hydroxide and magnesium hydroxide and was mixed at a 1:20 ratio with a
*Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam,
The Netherlands;
Department of Biochemistry, University of Lausanne, Epalinges, Swit-
zerland;
TopoTarget Switzerland, Lausanne, Switzerland; and
§
Laboratory of Immuno-
regulation, University Hospital Ghent, Ghent, Belgium
Received for publication May 22, 2008. Accepted for publication July 14, 2008.
The costs of publication of this article were defrayed in part by the payment of page charges.
This article must therefore be hereby marked advertisement in accordance with 18 U.S.C.
Section 1734 solely to indicate this fact.
1
B.N.L. and J.T. shared supervision over the work.
2
Address correspondence and reprint requests to Dr. Ju¨rg Tschopp, Department of Bio-
chemistry, Chemin des Boveresses 155, 1066 Epalinges, Switzerland. E-mail address:
jurg.tschopp@unil.ch or Dr. Bart Lambrecht, Laboratory of Immunoregulation, Univer-
sity Hospital Ghent, Ghent, Belgium. E-mail address: bart.lambrecht@ugent.be
3
Abbreviations used in this paper: DC, dendritic cell; ASC, apoptosis-associated speck-
like protein containing a caspase recruitment domain; IPAF, IL-1
-converting enzyme
protease activating factor; MLN, mediastinal lymph node; MSU, monosodium urate;
NLR, nucleotide-binding oligomerization domain-like receptor; ROS, reactive oxygen
species; WT, wild type.
Copyright © 2008 by The American Association of Immunologists, Inc. 0022-1767/08/$2.00
www.jimmunol.org
solution of OVA Ag in saline, followed by stirring for at least 1 h. For im-
munization, 500
l of alum (crystalline magnesium hydroxide and amor-
phous aluminum hydroxide) suspension (1 mg) containing 10
gofOVA
(OVA-alum) was injected i.p. or, alternatively, 10
g of OVA in 500
l
saline was injected.
Detection of cellular influx in vivo after OVA-alum injection
In WT and NALP3
/
mice, 2, 6, and 24 h after injection the peritoneal cavity
was washed with 3 ml of PBS containing EDTA (peritoneal lavage) and the
mediastinal lymph nodes were harvested and used for flow cytometry as previ-
ously described (14).
In experiments where the role of IL-1
was examined, mice were treated
with anti-IL-1
mAb (200
g/ms i.p.; BioLegend) 5 min before OVA-alum
injection and sacrificed 24 h later. Detection of Ag-specific T cell responses was
done as previously described (14).
Statistical analysis
For all experiments, the difference between groups was calculated using the
Mann-Whitney Utest for unpaired data (GraphPad Prism version 4.0). Dif-
ferences were considered significant when p0.05.
Results
Alum adjuvant activates the NALP3 inflammasome
We first exposed peritoneal macrophages obtained from WT,
NALP3
/
, and ASC
/
mice to various amounts of alum (see
Materials and Methods for composition). Even at low doses, a
substantial amount of processed IL-1
was detected in the cel-
lular supernatant, which compared well with the quantities of
this cytokine observed with crystalline monosodium urate
(MSU), one of the most potent activators of the NALP3 inflam-
masome (12) (Fig. 1A). IL-1
was almost completely absent in
supernatants of macrophages from NALP3- and ASC-deficient
mice, indicating a crucial involvement of NALP3 in caspase-1-
mediated, pro-IL-1
processing.
Activation of caspase-1 occurs not only upon assembly of the
NALP3 but also upon that of the IPAF inflammasome (15).
However, a contribution of this complex in alum-triggered
caspase-1 activation is unlikely, because alum-induced IL-1
secretion from macrophages derived from IPAF-deficient mice
was still observed (data not shown). Also, alum adjuvant con-
tinued to activate caspase-1 in the absence of MyD88, exclud-
ing a major role of TLRs in alum-mediated inflammasome ac-
tivation (Ref. 16 and data not shown).
Mechanisms of NALP3 inflammasome activation by alum
and uric acid crystals are similar
We next investigated the mechanisms by which alum acti-
vates the NALP3 inflammasome. Recent studies have shown
that NALP3 inflammasome activation is dependent on K
efflux (17, 18) and the generation of reactive oxygen species
(ROS) (17). K
efflux can be blocked by the addition of high
concentrations of K
(130 mM) to the extracellular me-
dium. The blockade of K
efflux resulted in the inhibition of
alum-induced caspase-1 activation and IL-1
secretion (Fig.
1B). Activation of the inflammasome was also reduced when
the ROS inhibitor (2R,4R)-4-aminopyrrolidine-2, 4-dicar-
boxylate (APDC) was added (Fig. 1B).
We could not exclude the possibility that activation of the
inflammasome by alum occurred indirectly through one of
the two endogenous danger signals known to activate
NALP3, namely ATP and uric acid (12, 19). ATP, which is
thought to be released from necrotic cells, potently activates
the inflammasome via binding to the P2X7 receptor (19),
whereas uric acid can form crystals that activate the inflam-
masome by an as yet poorly understood mechanism. We
have recently reported that extracellular ATP triggers in-
flammation by activating DCs (20) and that in vivo injection
of alum into the peritoneal cavity leads to uric acid (MSU)
release (14). It was therefore possible that MSU or ATP or
both, were released from alum-treated cells, leading to the
subsequent activation of the NALP3 inflammasome. How-
ever, we obtained no evidence for this hypothesis. Uricase,
which catalyzes the conversion of uric acid to allantoin (14),
had only a minor effect on IL-1
processing in vitro (Fig.
1B). Alum also still activated the inflammasome in macro-
phages of P2X7-deficient mice (data not shown).
Despite the fact that both ATP and MSU activate the
NALP3 inflammasome, the initial pathways leading to inflam-
masome activation are not identical. MSU is a particulate mat-
ter and requires endocytosis (12). In fact, endocytosis seems to
be required for particles in general, including asbestos and silica,
to activate the inflammasome (13). It was therefore expected
that alum also required endocytosis. The effect of alum, but not
ATP (data not shown), was abrogated by the actin-destabilizing
drug cytochalasin D (Fig. 1B). Taken together, NALP3 inflam-
masome activation in vitro by alum requires endocytosis of the
particle, K
efflux, and generation of ROS, but not release of
ATP or uric acid.
Recruitment of inflammatory cells to the site of alum injection is
decreased in the absence of NALP3
Intraperitoneal injection of alum is frequently used in mice to
induce cellular Th2 immunity and humoral immunity to ad-
sorbed protein Ags (21). To assess the role of the NALP3 in-
flammasome in alum adjuvanticity in vivo, we examined the
FIGURE 1. Role of NALP3 and ASC in alum-mediated IL-1
production.
A, Peritoneal macrophages from WT, NALP3
/
, and ASC
/
mice were
primed overnight with ultrapure LPS and then stimulated with different
amount of Imject Alum (Pierce Biochemicals). MSU (150
g/ml) was used as
a control. Production of mature IL-1
was measured by Western blotting in
supernatants (SN) and cell extracts (XT). B, Alum-induced IL-
secretion is
dependent on phagocytosis, K
efflux, and ROS. Peritoneal macrophages from
WZ mice were primed as described in Aand treated with alum in the presence
and absence of high concentrations of extracellular K
(130 mM), the ROS
inhibitor (2R,4R)-4-aminopyrrolidine-2, 4-dicarboxylate (APDC) (100
M),
cytochalasin D (2
M), and uricase (0.1 U/ml).
3756 CUTTING EDGE: INFLAMMASOME AND ALUM ADJUVANT
innate inflammatory response caused by alum upon i.p. injec-
tion of OVA adsorbed to alum. Twenty-four hours after injec-
tion there was a massive recruitment of neutrophils (Ly6G
,
F4/80
, and CD11b
cells), eosinophils (F4/80
dim
, Ly6G
,
and CD11b
cells), and inflammatory mononuclear cells
(CD11b
, F4/80
, and Ly6C
cells) into the peritoneal cavity
that was not seen when OVA was injected without alum (Fig.
2A). This innate response was severely reduced in NALP3-de-
ficient animals but nevertheless still present. Compared with in-
jection of saline or OVA alone, injection of OVA-alum induced
a rapid increase in the peritoneal cavity of the innate cytokines
IL-6 and IL-1
(Fig. 2B). Strikingly, in NALP3
/
mice the
increase in IL-1
was completely reduced to the level seen in
mice receiving saline or OVA, whereas the level of IL-6 was re-
duced but not abolished (Fig. 2B).
Activation of DCs is IL-1
and NALP3 dependent
DCs are seen as nature’s adjuvant and have a unique potential to
induce adaptive immunity. We have recently reported that the
induction of T cell adaptive immunity in vivo following the in-
jection of alum depends on uptake and transport of Ag from the
peritoneal cavity to the mediastinal LN by both resident
CD11c
DCs and inflammatory CD11b
Ly6C
monocytes
that become CD11c
DCs upon migration to the draining me-
diastinal lymph node (MLN) (14). To address the contribution
of inflammasome activation to this process, we measured the
uptake of fluorescent OVA in resident Ly6C
CD11c
DCs
following the injection of OVA or OVA-alum. Adsorption of
FIGURE 2. Innate inflammatory response induced by alum is NALP3 de-
pendent. NALP3
/
or WT mice were injected i.p. with OVA or OVA-alum.
A, Twenty-four hours after injection, the peritoneal lavage was taken and the
number of neutrophils (CD11b
Ly6C
Ly6G
high
F4/80
), eosinophils
(CD11b
Ly6C
int
Ly6G
int
F4/80
int
), and monocytes (CD11b
Ly6C
high
Ly6G
F4/80
int
) (were “int” is “intermediate”) was determined by flow cytometry. B,
Two hours after OVA or OVA-alum injection the peritoneal lavage was taken
and cytokine levels were determined in the supernatant by ELISA. Data shown
are mean SEM; ,p0.05; ⴱⴱ,p0.01; and ⴱⴱⴱ,p0.001; n4–6
mice per group.
FIGURE 3. Activation of DCs by alum is NALP3 dependent. A,
NALP3
/
or WT mice were injected i.p. with OVA-Alexa Fluor 647 or OVA-
AF647-alum. Twenty-four hours after injection the number of OVA
resident
Ly6C
CD11c
DCs was determined in the peritoneal lavage by flow cytom-
etry. B, Twenty-four hours after injection the number of CD11c
OVA
monocytes in the MLN was measured. C, Mice were treated with anti-IL-1
mAb before OVA-alum injection. The influx of CD11c
OVA
monocytes
into the MLN was measured 24 h after injection. D, CD86 and MHC II ex-
pressions were measured on the OVA
and OVA
monocytes in the MLN
24 h after the injection of OVA-alum in WT or NALP3
/
mice. Data shown
are mean SEM; ,p0.05; n46 mice per group.
3757The Journal of Immunology
OVA-Alexa Fluor 647 in alum led to a strong increase in DCs
carrying antigenic cargo in WT mice but not in NALP3
/
mice (Fig. 3A). We also followed the appearance of OVA-laden
Ly6C
inflammatory monocytes in the MLN (Fig. 3B). In WT
mice there was a time-dependent increase in the number of in-
flammatory monocytes in the MLN at 24 h, and a majority of
these carried OVA. In contrast, in NALP3
/
mice this in-
crease was severely diminished. This deficiency was most likely
explained by a defective generation of bioactive IL-1
in
NALP3
/
mice, as the alum-induced increase in OVA-laden
CD11c
monocytes in the MLN was severely reduced when
IL-1
was neutralized in vivo, implying an important func-
tional role for this cytokine in promoting monocyte migra-
tion and differentiation (Fig. 3C). Provision of costimula-
tion is a necessary requisite for the induction of adaptive
immunity. Injection of OVA-alum induced the up-regula-
tion of the costimulatory molecule CD86 on inflammatory
OVA
CD11c
Ly6C
monocytes in the MLN of WT mice,
but to a much lesser extent in NALP3
/
mice (Fig. 3D). A
similar effect was seen for MHC class II (Fig. 3D).
The reduced migration of OVA-laden Ly6C
monocytic
cells with DC characteristics to the mediastinal nodes of
NALP3
/
mice prompted us to study the activation of naive
OVA-specific CD4
T cells. For this, mice first received a co-
hort of CFSE-labeled T cells obtained from OT-II TCR Tg
mice followed by injection of OVA or OVA-alum, and accu-
mulation of OVA-specific T cells was measured in the lymph
nodes. In WT mice OVA-alum caused a marked increase in the
number of Ag-reactive T cells in the mediastinal LN at days 4
and 10 postimmunization (Fig. 4A) compared with injection of
OVA alone. In NALP3
/
mice, the number of T cells induced
by OVA-alum was severely reduced but still higher than that
obtained by the injection of OVA alone. These changes in the
accumulation of T cells were mainly due to altered degrees of T
cell division, as measured using CFSE dilution (Fig. 4B).
We finally measured the effect of NALP3 deficiency on the
type of immunoglobulins induced by OVA-alum cells (Fig.
4C). Mice receiving OVA alone have generally low Ab titers to
OVA (data not shown). In WT mice, OVA-alum induced the
production of IgE, IgG1, and somewhat lower levels of IgG2c.
In NALP3
/
mice there was a significant decrease in OVA-
specific IgE Abs and an increase in OVA-specific IgG2c Abs.
Strikingly, IgG1 levels were unaffected.
Discussion
In 1926, the adjuvant effect of aluminum compounds was first
described by Glenny et al. (22). Despite this early discovery, to
this day little is known about the mode of action of alum adju-
vant. Adjuvants containing a pathogen-associated molecular
patterns act as ligands for the TLRs. For example, TLR9 was
shown to be essential for the adjuvant effect of CpG oligode-
oxynucleotides (3). However, incubation of DCs with alum ad-
juvant fails to activate the DCs as indicated by the increased
expression of MHC class II and costimulatory molecules (23);
yet, several authors described an increased production of IL-1
secretion (24, 25). Mice lacking TLR signaling components
still mount a robust Ab responses if alum is given as an adjuvant
(5). Thus, alum adjuvants, in contrast to bacteria-derived adju-
vants, do not activate TLRs.
Entirely in agreement with two recent studies by Eisenbarth
and Li and colleagues (16, 26), our data show that alum adju-
vant triggers activation of the NALP3 inflammasome. The po-
tential of alum to trigger the NALP3 inflammasome leads to
early activation of the innate cytokine IL-1
and an innate cel-
lular immune response at the site of injection. Activation of the
NALP3 inflammasome and the subsequent release of IL-1
leads to the recruitment of immature monocytes and DCs. Pro-
duction of IL-1
also leads to the activation of inflammatory
monocytes and their migration to the lymph nodes draining the
peritoneum. This situation resembles the migration of skin
Langerhans cells to the lymph nodes, which also requires IL-1
for functional migration and maturation to occur (27). In
OVA-alum-immunized mice, OVA-laden DCs and inflamma-
tory monocytes take their cargo to the MLN and up-regulate
the expression of critical costimulatory molecules like CD86
with the subsequent expansion of Ag-specific T cells (14). All of
these steps are highly impaired in NALP3-deficient mice, indi-
cating that activation of the NALP3 inflammasome contributes
FIGURE 4. Induction of adaptive cellular immunity by alum is NALP3 de-
pendent. A, NALP3
/
or WT mice were injected with CFSE-labeled OT-2
OVA-TCR Tg cells 1 day before the i.p. injection of OVA or OVA-alum. Four
and 10 days after the injection the MLN was analyzed for T cell proliferation
with flow cytometry (n4 mice; experiment was performed two times). B,
Representative CFSE profiles of adoptively transferred T cells on day 10 after
injection. Data are shown as mean SEM; ,p0.05; n5–11 mice per
group. C, On day 0, NALP3
/
or WT mice were injected i.p. with OVA-alum
and boosted i.p. on day 7 with OVA. At day 14, serum samples were taken and
OVA-specific IgE, IgG1, and IgG2c levels were determined by ELISA. Data are
shown as mean SEM; ,p0.05; n5–11 mice per group.
3758 CUTTING EDGE: INFLAMMASOME AND ALUM ADJUVANT
not only to the immediate inflammatory innate response at the
site of injection of alum but also to the generation of adaptive
cellular immunity.
Levels of serum Igs are also influenced in the NALP3-defi-
cient mice, as a decreased level of OVA-IgE and increased levels
of OVA-IgG2c are found. This pattern of Ig synthesis is most
likely the result of a shift toward IFN-
-secreting Th1-like
helper cells necessary for Ig class switching to IgG2c and equally
able to suppress IgE synthesis. Interestingly, we found that
NALP3 does not influence IgG1 levels, whereas decreased levels
of this subclass were found in the Eisenbarth report (16). This
striking-difference may be due to a different experimental
setup, as in their study Ab generation was determined after an
intranasal challenge with OVA (16) whereas we boosted mice
by injecting OVA i.p. The data are most consistent with the
notion that NALP3-dependent IL-1
production by DCs
skews the adaptive cellular immune response toward a Th2 type
of response, as recently proposed (25).
Strikingly, alum adjuvant activates the NALP3 inflamma-
some directly in vitro, in agreement with two reports showing
IL-1
secretion from alum-treated DCs (24, 25). We have re-
cently found that alum also induces high-level production of
uric acid in vivo and that this increased level of uric acid was
required for the infiltration of inflammatory cells (14). Al-
though how this is done and which cells generate or release uric
acid upon alum administration are open questions, it suggests
that the increased level of uric acid leads to an amplification of
NALP3 inflammasome activation and, thus, IL-1
secretion.
Interestingly, uric acid was identified not only as one of the
most potent danger signals released from dying cells, but also as
an excellent adjuvant (28). Thus, we propose NALP3 as the
newest member of a list of innate receptors that could be ex-
ploited for the design of new adjuvants.
Acknowledgments
We thank V. Dixit (Genentech, San Francisco, CA) for the ASC
/
and
Ipaf
/
mice and S. Akira (Osaka University, Osaka, Japan) for the
MyD88
/
mice.
Disclosures
The authors have no financial conflict of interest.
References
1. Parkinson, T. 2008. The future of Toll-like receptor therapeutics. Curr. Opin. Mol.
Ther. 10: 21–31.
2. Daubenberger, C. A. 2007. TLR9 agonists as adjuvants for prophylactic and thera-
peutic vaccines. Curr. Opin. Mol. Ther. 9: 45–52.
3. Akira, S., and K. Takeda. 2004. Toll-like receptor signalling. Nat. Rev. Immunol. 4:
499–511.
4. Schnare, M., G. M. Barton, A. C. Holt, K. Takeda, S. Akira, and R. Medzhitov. 2001.
Toll-like receptors control activation of adaptive immune responses. Nat. Immunol. 2:
947–950.
5. Gavin, A. L., K. Hoebe, B. Duong, T. Ota, C. Martin, B. Beutler, and D. Nemazee.
2006. Adjuvant-enhanced antibody responses in the absence of Toll-like receptor sig-
naling. Science 314: 1936–1938.
6. Martinon, F., and J. Tschopp. 2007. Inflammatory caspases and inflammasomes:
master switches of inflammation. Cell Death Differ. 14: 10–22.
7. Franchi, L., J. H. Park, M. H. Shaw, N. Marina-Garcia, G. Chen, Y. G. Kim, and
G. Nunez. 2008. Intracellular NOD-like receptors in innate immunity, infection and
disease. Cell. Microbiol. 10: 1–8.
8. Ting, J., and B. Davis. 2005. CATERPILLER: a novel gene family important in im-
munity, cell death, and diseases. Annu. Rev. Immunol. 23: 387–414.
9. Agostini, L., F. Martinon, K. Burns, E. M. McDermott, P. N. Hawkins, and
J. Tschopp. 2004. NALP3 forms an IL-1
processing inflammasome with increased
activity in Muckle-Wells auto-inflammatory disorder. Immunity 20: 319–325.
10. Fritz, J. H., R. L. Ferrero, D. J. Philpott, and S. E. Girardin. 2006. Nod-like proteins
in immunity, inflammation and disease. Nat. Immunol. 7: 1250–1257.
11. Petrilli, V., C. Dostert, D. A. Muruve, and J. Tschopp. 2007. The inflammasome: a
danger sensing complex triggering innate immunity. Curr. Opin. Immunol. 19:
615–622.
12. Martinon, F., V. Petrilli, A. Mayor, A. Tardivel, and J. Tschopp. 2006. Gout-associ-
ated uric acid crystals activate the NALP3 inflammasome. Nature 440: 237–241.
13. Dostert, C., V. Petrilli, R. Van Bruggen, C. Steele, B. T. Mossman, and J. Tschopp.
2008. Induction of innate immune responses through Nalp3 inflammasome sensing
of asbestos and silica. Science 320: 674–677.
14. Kool, M., T. Soullie, M. van Nimwegen, M. A. Willart, F. Muskens, S. Jung,
H. C. Hoogsteden, H. Hammad, and B. N. Lambrecht. 2008. Alum adjuvant boosts
adaptive immunity by inducing uric acid and activating inflammatory dendritic cells.
J. Exp. Med. 205: 869–882.
15. Mariathasan, S., K. Newton, D. M. Monack, D. Vucic, D. M. French, W. P. Lee,
M. Roose-Girma, S. Erickson, and M. V. Dixit. 2004. Differential activation of the
inflammasome by caspase-1adaptors ASC and Ipaf. Nature 430: 213–218.
16. Eisenbarth, S. C., O. R. Colegio, W. O’Connor, F. S. Sutterwala, and R. A. Flavell.
2008. Crucial role for the Nalp3 inflammasome in the immunostimulatory properties
of aluminium adjuvants. Nature 453: 1122–1126.
17. Petrilli, V., S. Papin, C. Dostert, A. Mayor, F. Martinon, and J. Tschopp. 2007. Ac-
tivation of the NALP3 inflammasome is triggered by low intracellular potassium con-
centration. Cell Death Differ. 14: 1583–1589.
18. Franchi, L., T. D. Kanneganti, G. R. Dubyak, and G. Nunez. 2007. Differential re-
quirement of P2X7 receptor and intracellular K
for caspase-1 activation induced by
intracellular and extracellular bacteria. J. Biol. Chem. 282: 18810–18818.
19. Mariathasan, S., D. S. Weiss, K. Newton, J. McBride, K. O’Rourke, M. Roose-Girma,
W. P. Lee, Y. Weinrauch, D. M. Monack, and V. M. Dixit. 2006. Cryopyrin activates
the inflammasome in response to toxins and ATP. Nature 440: 228–232.
20. Idzko, M., H. Hammad, M. van Nimwegen, M. Kool, M. A. Willart, F. Muskens,
H. C. Hoogsteden, W. Luttmann, D. Ferrari, F. Di Virgilio, et al. 2007. Extracellular
ATP triggers and maintains asthmatic airway inflammation by activating dendritic
cells. Nat. Med. 13: 913–919.
21. Lindblad, E. B. 2004. Aluminium adjuvants–in retrospect and prospect. Vaccine 22:
3658–3668.
22. Glenny, A. T., C. G. Pope, H. Waddington, and U. Wallace. 1926. Immunological
notes XVII to XXIV. J. Pathol. 29: 31–40.
23. Sun, H., K. G. Pollock, and J. M. Brewer. 2003. Analysis of the role of vaccine adju-
vants in modulating dendritic cell activation and antigen presentation in vitro. Vaccine
21: 849–855.
24. Li, H., S. Nookala, and F. Re. 2007. Aluminum hydroxide adjuvants activate
caspase-1 and induce IL-1
and IL-18 release. J. Immunol. 178: 5271–5276.
25. Sokolovska, A., S. L. Hem, and H. HogenEsch. 2007. Activation of dendritic cells and
induction of CD4
T cell differentiation by aluminum-containing adjuvants. Vaccine
25: 4575–4585.
26. Li, H., S. B. Willingham, J. P. Ting, and F. Re. 2008. Cutting Edge: Inflammasome
activation by alum and alum’s adjuvant effect are mediated by NLRP3. J. Immunol.
181: 17–21.
27. Antonopoulos, C., M. Cumberbatch, R. J. Dearman, R. J. Daniel, I. Kimber, and
R. W. Groves. 2001. Functional caspase-1 is required for Langerhans cell migration
and optimal contact sensitization in mice. J. Immunol. 166: 3672–3677.
28. Shi, Y., J. E. Evans, and K. L. Rock. 2003. Molecular identification of a danger signal
that alerts the immune system to dying cells. Nature 425: 516–521.
3759The Journal of Immunology
... While these convenient procedures are adequate to examine allergic responses in general, they are insufficient for investigating the sensitization process to inhaled allergens such as BP. Furthermore, the fact that alum mediates its adjuvant function via activation of the NLRP3 inflammasome (29,30) makes it impossible to clearly distinguish alum-induced immune responses from direct responses to the allergen or allergen extract. Therefore, we aimed to develop a physiologic and adjuvant-free mouse model to study the involvement of NLRP3 in BP allergy. ...
Article
Full-text available
Introduction & Objective Allergic sensitization is an essential step in the development of allergic airway inflammation to birch pollen (BP); however, this process remains to be fully elucidated. Recent scientific advances have highlighted the importance of the allergen context. In this regard, microbial patterns (PAMPs) present on BP have attracted increasing interest. As these PAMPs are recognized by specialized pattern recognition receptors (PRRs), this study aims at investigating the roles of intracellular PRRs and the inflammasome regulator NLRP3. Methods We established a physiologically relevant intranasal and adjuvant-free sensitization procedure to study BP-induced systemic and local lung inflammation. Results Strikingly, BP-sensitized Nlrp3-deficient mice showed significantly lower IgE levels, Th2-associated cytokines, cell infiltration into the lung, mucin production and epithelial thickening than their wild-type counterparts, which appears to be independent of inflammasome formation. Intriguingly, bone-marrow chimera revealed that expression of NLRP3 in the hematopoietic system is required to trigger an allergic response. Conclusion Overall, this study identifies NLRP3 as an important driver of BP-induced allergic immune responses.
... Aluminum adjuvant, the unique adjuvant approved by the U. S. Food and Drug Administration (FDA), and the most used adjuvant in licensed human vaccines, may divert antigens from their typical trafcking route toward endolysosomal compartments by its adsorption, which can sharply augment antigen persistency, antigen uptake, and local immunostimulatory response [15,16]. Te aluminum adjuvant is able to adsorb antigens in solution and generates immunogenic particles, and it triggers a T2-biased immune response by activating the NALP3 infammasome, followed by the secretion of IL-1β, IL-18, and IL-33 [15,[17][18][19]; however, poor T1-type immune response induced by aluminum adjuvant makes it inappropriate as a single component in some vaccines. ...
Article
Full-text available
Introduction The development of combinatorial adjuvants is a promising strategy to boost vaccination efficiency. Accumulating evidence indicates that manganese exerts strong immunocompetence and will become an enormous potential adjuvant. Here, we described a novel combination of Mn²⁺ plus aluminum hydroxide (AH) adjuvant that significantly exhibited the synergistic immune effect. Methodology. Initially, IsdB3 proteins as the immune-dominant fragment of IsdB proteins derived from Staphylococcus aureus (S. aureus) were prepared. IsdB3 proteins were identified by western blotting. Furthermore, we immunized C57/B6 mice with IsdB3 proteins plus Mn²⁺ and AH adjuvant. After the second immunization, the proliferation of lymphocytes was measured by the cell counting kit-8 (CCK-8) and the level of IFN-γ, IL-4, IL-10, and IL-17 cytokine from spleen lymphocytes in mice and generation of the antibodies against IsdB3 in serum was detected with ELISA, and the protective immune response was assessed through S. aureus challenge. Results IsdB3 proteins plus Mn²⁺ and AH obviously stimulated the proliferation of spleen lymphocytes and increased the secretion of IFN-γ, IL-4, IL-10, and IL-17 cytokine in mice, markedly enhanced the generation of the antibodies against IsdB3 in serum, observably decreased bacterial load in organs, and greatly improved the survival rate of mice. Conclusion These data showed that the combination of Mn²⁺ and AH significantly acted a synergistic effect, reinforced the immunogenicity of IsdB3, and offered a new strategy to increase vaccine efficiency.
... The adjuvantantigen aggregates activate immune cells, leading to the secretion of cytokines and chemokines as well as the formation of nucleotide oligomerization domain (NOD)-, leucine rich repeat (LRR)-and pyrin domain-containing protein 3 (NLRP3) inflammasomes. Subsequently, antigen-presenting cells (APCs) are recruited and activated by the production of strongly pro-inflammatory, bioactive IL-1b and IL-18, cell death, and the release of damage-associated molecular patterns (DAMPs) such as uric acid and self-DNA ( Figure 1) (10)(11)(12). In addition, adjuvant-antigen aggregates can not only be directly taken up by APCs, but also act as depots, causing a slow release and effective presentation of the incorporated antigen(s) to APCs (Figure 1) (12). ...
Article
Full-text available
Type I hypersensitivity, or so-called type I allergy, is caused by Th2-mediated immune responses directed against otherwise harmless environmental antigens. Currently, allergen-specific immunotherapy (AIT) is the only disease-modifying treatment with the potential to re-establish clinical tolerance towards the corresponding allergen(s). However, conventional AIT has certain drawbacks, including long treatment durations, the risk of inducing allergic side effects, and the fact that allergens by themselves have a rather low immunogenicity. To improve AIT, adjuvants can be a powerful tool not only to increase the immunogenicity of co-applied allergens but also to induce the desired immune activation, such as promoting allergen-specific Th1- or regulatory responses. This review summarizes the knowledge on adjuvants currently approved for use in human AIT: aluminum hydroxide, calcium phosphate, microcrystalline tyrosine, and MPLA, as well as novel adjuvants that have been studied in recent years: oil-in-water emulsions, virus-like particles, viral components, carbohydrate-based adjuvants (QS-21, glucans, and mannan) and TLR-ligands (flagellin and CpG-ODN). The investigated adjuvants show distinct properties, such as prolonging allergen release at the injection site, inducing allergen-specific IgG production while also reducing IgE levels, as well as promoting differentiation and activation of different immune cells. In the future, better understanding of the immunological mechanisms underlying the effects of these adjuvants in clinical settings may help us to improve AIT.
... Trong đó, ATP và uric acid cảm ứng con đường tín hiệu viêm của các phân tử NACHT, LRR and PYD domains-containing protein 3 (NLRP3), còn các DNA giải phóng từ tế bào chết sau khi bị tế bào tua thực bào được nhận diện bởi enzyme nội bào cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase (cGAS) để cảm ứng stimulator of interferon gene (STING), là protein thiết yếu của tính miễn dịch bẩm sinh. NLRP3 và STING sau khi được cảm ứng sẽ hoạt hóa các nhân tố phiên mã như NF-κB và IRF3 giúp tăng cường biểu hiện các cytokine giữ vai trò kích Hình 1: Quá trình trình diện kháng nguyên của tế bào tua khi có (A) và không có (B) phân tử đồng kích thích thích hệ miễn dịch thích ứng 16,26,30,31 . Tuy nhiên, nhược điểm lớn nhất của tá dược miễn dịch nhôm là khả năng kích thích tế bào B chuyển lớp kháng thể sang immunoglobulin (Ig) E 26 (dạng kháng thể giữ vai trò ngăn chặn kí sinh trùng và liên quan đến dị ứng). ...
Article
Full-text available
In the emergency that the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has globally spread, vaccine is one of the most indispensable defense lines to repulse the resulting Coronavirus Disease 2019 (COVID-19) pandemic. The effective immunization frequently requires appropriate adjuvants which play a key role to activate the dendritic cell maturation. Indeed, the antigen presentation of dendritic cells indisputably needs the collaboration of co-stimulatory molecules which are only expressed by mature dendritic cells to trigger T cell response. In this review, we collectively summarized well-known adjuvants used for vaccination as well as their characteristics and immunostimulatory effects on both of humoral and cell-mediated immune response. Besides, the applicability of adjuvants in COVID-19 vaccination and accompanying challenges were also discussed. Generally, despite of certain successes of available licensed adjuvants in the vaccination, new generations of novel adjuvants would absolutely be required to produce effective vaccines for other inevitable pathogenic organisms in the future.
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
Introduction: Prostaglandin D2 (PGD2), which is produced mainly by Th2 cells and mast cells, promotes a type-2 immune response by activating Th2 cells, mast cells, eosinophils, and group 2 innate lymphoid cells (ILC2s) via its receptor, chemoattractant receptor-homologous molecules on Th2 cells (CRTH2). However, the role of CRTH2 in models of airway inflammation induced by sensitization without adjuvants, in which both IgE and mast cells may play major roles, remain unclear. Methods: Wild-type (WT) and CRTH2-knockout (KO) mice were sensitized with ovalbumin (OVA) without an adjuvant and then challenged intranasally with OVA. Airway inflammation was assessed based on airway hyperresponsiveness (AHR), lung histology, number of leukocytes, and levels of type-2 cytokines in the bronchoalveolar lavage fluid (BALF). Results: AHR was significantly reduced after OVA challenge in CRTH2 KO mice compared to WT mice. The number of eosinophils, levels of type-2 cytokines (IL-4, IL-5, and IL-13) in BALF, and IgE concentration in serum were decreased in CRTH2 KO mice compared to WT mice. However, lung histological changes were comparable between WT and CRTH2 KO mice. Conclusion: CRTH2 is responsible for the development of asthma responses in a mouse model of airway inflammation that features prominent involvement of both IgE and mast cells.
Article
Full-text available
Langerhans cell (LC) migration from epidermis to draining lymph node is a critical first step in cutaneous immune responses. Both TNF-alpha and IL-1 beta are important signals governing this process, but the potential regulatory role of IL-1 alpha processing by caspase-1 is unknown. In wild-type (WT) mice, application of the contact allergens 2,4-dinitrofluorobenzine and oxazolone lead to a marked reduction in epidermal LC numbers, but in caspase-1-deficient mice this reduction was not observed. Moreover, although intradermal injection of TNF-alpha (50 ng) induced epidermal LC migration in WT mice, this cytokine failed to induce LC migration in caspase-1-deficient mice. Intradermal IL-1 beta (50 ng) caused a similar reduction in epidermal LC numbers in both WT and caspase-1-deficient mice, indicating that, given an appropriate signal, caspase-1-deficient epidermal LC are capable of migration. Contact hypersensitivity to both 2,4-dinitrofluorobenzine and oxazolone was inhibited in caspase-1-deficient mice, indicating a functional consequence of the LC migration defect. In organ culture the caspase-1 inhibitor Ac-YVAD-cmk, but not control peptide, potently inhibited the epidermal LC migration that occurs in this system, and reduced spontaneous migration of LC was observed in skin derived from caspase-1-deficient mice. Moreover, Ac-YVAD-cmk applied to BALB/c mouse skin before application of contact sensitizers inhibited LC migration and contact hypersensitivity in vivo. Taken together, these data indicate that caspase-1 may play a central role in the regulation of LC migration and suggest that the activity of this enzyme is amenable to control by specific inhibitors both in vivo and in vitro.
Article
Full-text available
Mechanisms that control the activation of antigen-specific immune responses in vivo are poorly understood. It has been suggested that the initiation of adaptive immune responses is controlled by innate immune recognition. Mammalian Toll-like receptors play an essential role in innate immunity by recognizing conserved pathogen-associated molecular patterns and initiating the activation of NF-kappaB and other signaling pathways through the adapter protein, MyD88. Here we show that MyD88-deficient mice have a profound defect in the activation of antigen-specific T helper type 1 (TH1) but not TH2 immune responses. These results suggest that distinct pathways of the innate immune system control activation of the two effector arms of adaptive immunity.
Article
Full-text available
In infections, microbial components provide signals that alert the immune system to danger and promote the generation of immunity. In the absence of such signals, there is often no immune response or tolerance may develop. This has led to the concept that the immune system responds only to antigens perceived to be associated with a dangerous situation such as infection. Danger signals are thought to act by stimulating dendritic cells to mature so that they can present foreign antigens and stimulate T lymphocytes. Dying mammalian cells have also been found to release danger signals of unknown identity. Here we show that uric acid is a principal endogenous danger signal released from injured cells. Uric acid stimulates dendritic cell maturation and, when co-injected with antigen in vivo, significantly enhances the generation of responses from CD8+ T cells. Eliminating uric acid in vivo inhibits the immune response to antigens associated with injured cells, but not to antigens presented by activated dendritic cells. Our findings provide a molecular link between cell injury and immunity and have important implications for vaccines, autoimmunity and inflammation.
Article
Aluminium compounds have been used as adjuvants in practical vaccination for more than 60 years to induce an early, an efficient and a long lasting protective immunity and are at present the most widely used adjuvants in both veterinary and human vaccines. Although the last two decades of systematic research into the nature of these adjuvants has contributed significantly to understanding their nature and their limitations as Th2 stimulators the more detailed mode of action of these adjuvants is still not completely understood. We have a comprehensive record of their behaviour and performance in practical vaccination, but an empirical approach to optimising their use in new vaccine formulations is still to some extent a necessity. The aim of the present review is to put the recent findings into a broader perspective to facilitate the application of these adjuvants in general and experimental vaccinology.
Article
Aluminum hydroxide (Alum) is the only adjuvant approved for routine use in humans, although the basis for its adjuvanticity remains poorly understood. In this study, we show that Alum activates caspase-1 and induce secretion of mature IL-1beta and IL-18. Human PBMC or dendritic cells stimulated with pure TLR4 and TLR2 agonists released only traces of IL-1beta or IL-18, despite the fact that the IL-1beta mRNA was readily induced by both TLR agonists. In contrast, cells costimulated with TLR agonists plus Alum released large amount of IL-1beta and IL-18. Alum-induced IL-1beta and IL-18 production was not due to enhancement of TLR signaling but rather reflected caspase-1 activation and in mouse dendritic cells occurred in a MyD88-independent fashion. Secretion of other proinflammatory cytokines such as IL-8 was not affected by Alum treatments. However, TLR-induced production of IL-10 was increased and that of IFN-gamma-inducible protein decreased by Alum cotreatment. Considering the immunostimulatory activities of these cytokines and the ability of IL-1beta to act as adjuvant, our results suggest a mechanism for the adjuvanticity of Alum.
Article
We have studied the effects of adjuvant formulations on the activation and antigen-presenting functions of bone marrow-derived dendritic cells (DCs). While LPS could induce high-level expression of MHC Class II and co-stimulator molecules on DCs, it did not enhance antigen presentation to co-stimulation independent DO11.GFP T hybridoma cells. In contrast, alum, NISV and PLGA formulations failed to activate DCs, but NISV and PLGA could enhance antigen-presentation efficiency by 10-100-fold. Irrespective of the previously described antigen release characteristics of each adjuvant, antigen presentation peaked at 6h and waned thereafter for all formulations. Given the importance of DCs in the activation of nai;ve T cell responses, these studies suggest that as yet undefined pathways of DC activation in vivo may underlie the activity of alum, PLGA and NISV adjuvants. Furthermore, as NISV and PLGA do not appear to act as slow-release systems in DCs, the ability of these particulate systems to induce high levels of antigen presentation by DCs probably has a more significant role in their adjuvant activity.