Pi Is 0021925817499200
Pi Is 0021925817499200
Pi Is 0021925817499200
The NLR family pyrin domain containing 3 (NLRP3) inflam- teleost fish, providing a cross-species understanding of the evo-
masome is one of the best-characterized inflammasomes in lutionary history of inflammasomes. Our findings also indicate
humans and other mammals. However, knowledge about the that the NLRP3 inflammasome may coordinate inflammatory
NLRP3 inflammasome in nonmammalian species remains lim- cytokine processing and secretion through a GSDME-mediated
ited. Here, we report the molecular and functional identification pyroptotic pathway, uncovering a previously unrecognized reg-
of an NLRP3 homolog (DrNLRP3) in a zebrafish (Danio rerio) ulatory function of NLRP3 in both inflammation and cell
model. We found that DrNLRP3’s overall structural architec- pyroptosis.
ture was shared with mammalian NLRP3s. It initiates a classical
inflammasome assembly for zebrafish inflammatory caspase
(DrCaspase-A/-B) activation and interleukin 1 (DrIL-1) mat- Inflammasomes are cytosolic multiprotein complexes that
uration in an apoptosis-associated speck-like protein contain- assemble in response to exogenous microbial invasions and
ing a caspase-recruitment domain (ASC)-dependent manner, in endogenous damage signals (1, 2). Inflammasomes act as a
which DrNLRP3 organizes DrASC into a filament that recruits central platform to activate inflammatory caspases such as
DrCaspase-A/-B by homotypic pyrin domain (PYD)–PYD inter- Caspase-1, which processes the proinflammatory cytokines
actions. DrCaspase-A/-B activation in the DrNLRP3 inflam- interleukin 1 (IL-1)3 and IL-18 for their maturation, and
masome occurred in two steps, with DrCaspase-A being acti- hydrolyze gasdermin D to generate an N-terminal fragment to
vated first and DrCaspase-B second. DrNLRP3 also directly induce membrane perforation, cytokine release, and cell pyrop-
activated full-length DrCaspase-B and elicited cell pyroptosis tosis (3, 4). Among numerous inflammasomes identified in
in a gasdermin E (GSDME)-dependent but ASC-indepen- mammals, NLRP3 inflammasome is the most extensively stud-
dent manner. These two events were tightly coordinated by ied because of its crucial functional roles in innate immunity
DrNLRP3 to ensure efficient IL-1 secretion for the initiation of and pathogenesis of various diseases (5, 6). NLRP3 is a family
host innate immunity. By knocking down DrNLRP3 in zebrafish member of nucleotide-binding domain and leucine-rich repeat
embryos and generating a DrASC-knockout (DrASCⴚ/ⴚ) fish (LRR)-containing proteins (NLRs) (7). The mammalian NLRP3
clone, we characterized the function of the DrNLRP3 inflam- is structurally characterized by the presence of an N-terminal
masome in anti-bacterial immunity in vivo. The results of our pyrin domain (PYD), a central adenosine triphosphatase
study disclosed the origin of the NLRP3 inflammasome in (ATPase) domain known as NACHT, a domain associated with
NACHT in fish and other vertebrates (FISNA) (8), and a C-ter-
This work was supported by National Natural Science Foundation of China minal LRR domain (9). Once activated by a spectrum of stimuli,
Grants 31630083 and 31572641; National Key Research and Development
Program of China Grants 2018YFD0900503 and 2018YFD0900505; Stem
Cell and Translational Research; National Key Research and Development 3
The abbreviations used are: IL-1, interleukin 1; ASC, apoptosis-associated
Program of China Grant 2016YFA0101001; the Open Fund of the Labora- speck-like protein containing a caspase-recruitment domain; CaspaseB-
tory for Marine Biology and Biotechnology; Qingdao National Laboratory 4DA, mutant of DrCaspase-B in which four asparagine acids (Asp130,
for Marine Science and Technology, Qingdao, China, Grant OF2017NO02; Asp137, Asp308, and Asp314) were substituted by alanine; DrASC, D. rerio
the Open Funding Project of the State Key Laboratory of Bioreactor Engi- ASC; DrCaspase, D. rerio caspase; DrIL-1, D. rerio IL-1; DrNLRP3, D. rerio
neering; and the Zhejiang Major Special Program of Breeding Grant NLRP3; DrGSDME, D. rerio GSDME; FCM, flow cytometry; FISNA, domain
2016C02055-4. The authors declare that they have no conflicts of interest associated with NACHT in fish and other vertebrates; FRAP, fluorescence
with the contents of this article. recovery after photobleaching; GSDMD, gasdermin D; GSDME, gasdermin
This article contains Figs. S1–S5 and Table S1. E; gRNA, guide RNA; hpf, hour post-fertilization; HsNLRP3, H. sapiens
The nucleotide sequence(s) reported in this paper has been submitted to the NLRP3; LDH, lactate dehydrogenase; LPS, lipopolysaccharide; LRR,
GenBankTM/EBI Data Bank with accession number(s) MN088121, XM_ leucine-rich repeat; MDP, muramyl dipeptide; MmNLRP3, M. musculus NLRP3;
005170077.3, and NM_001001947.1. MO, morpholino oligonucleotide; NCBI, National Center for Biotechnology
1
To whom correspondence may be addressed: College of Life Sciences, Zhe- Information; NLRP3, NLR family pyrin domain containing 3; NLR, nucleotide-
jiang University, 866 YuHangTang Rd., Hangzhou 310058, China. Tel.: binding domain and leucine-rich repeat-containing proteins ORF, open read-
86-571-88206582; Fax: 86-571-88206582; E-mail: [email protected]. ing frame; PI, propidium iodide; PDB, Protein Data Bank; PYD, pyrin domain;
2
To whom correspondence may be addressed: College of Life Sciences, Zhe- ROI, region of interest; RSR, relative survival rate; LRR, leucine-rich repeat; Ab,
jiang University, 866 YuHangTang Rd., Hangzhou 310058, China. Tel.: antibody; DMEM, Dulbecco’s modified Eagle’s medium; Ac, acetyl; AFC,
86-571-88206582; Fax: 86-571- 88206582; E-mail: [email protected]. amido-4-trifluoromethylcoumarin; gRNA, guide RNA.
Figure 1. Molecular and structural identification of DrNLRP3 and DrGSDME. A, gene synteny and chromosomal location analysis of genes adjacent to
NLRP3 loci on human chromosome 1 (top) and zebrafish chromosome 1 (bottom). Arrows indicate gene orientation. B, schematic of the domain architecture of
HsNLRP3 and DrNLRP3. C, DrNLRP3 and HsNLRP3 tertiary structures predicted by SWISS-MODEL with crystal structures of NACHT and LRR (PDB code 4kxf.3.A)
as models. D, DrNLRP3 domain architecture and tertiary structure modeled by I-TASSER. The top five threading templates are 5irmA, 6npva, 4kxfK, 6b5bA, and
6eqoA. E, schematic of the domain architecture of HsGSDME and DrGSDMEa/b. The predicted cleaved sites in DrGSDMEa and DrGSDMEb are 253SEVD256 and
244
FEID247, respectively. F, tertiary structures of full-length HsGSDME and DrGSDMEa/b protein predicted by SWISS-MODEL with crystal GSDMA3 structures
(PDB code 5b5r.1.A) as model. G, gene synteny and chromosomal location analysis of genes adjacent to HsGSDME and DrGSDMEa/b loci on human chromo-
some 1 (top) and zebrafish chromosome 19/16 (bottom). Arrows indicate gene orientation. H and I, phylogenetic analysis of the relationship of NLRP3 and
GSDME between fish and other species. The phylogenetic tree was constructed by MEGA (version 5.0) by using the maximum likelihood method. Each node
reliability was estimated by bootstrapping with 2000 replications. J, quantitative RT-PCR analysis of the expression patterns of DrNLRP3 and DrGSDMEa/b
genes in adult zebrafish tissues. K, expression patterns of DrNLRP3 and DrGSDMEa/b genes in zebrafish embryos at different developmental stages. The relative
expression levels of the genes was calculated by the 2⫺⌬Ct method with -actin for normalization. Each data point shows the mean ⫾ S.D. with three replicates
representative of three independent experiments.
naturally have minimal expression of inflammasome compo- DrCaspase-B activation. The DrNLRP3-⌬LRR mutants
nents. As expected, DrNLRP3 and DrASC coexpression signif- restrained DrCaspase-A but not DrCaspase-B activation.
icantly augmented (p ⬍ 0.01) the DrCaspase-A/-B activity in a Conversely, DrNLRP3-⌬B30.2 did not influence either
DrNLRP3 dose-dependent manner (Fig. 2, A and B). However, DrCaspase-A or DrCaspase-B (Fig. 2C). Western blot analysis
the DrNLRP3 and DrCaspase-A/-B coexpression in the absence showed the self-cleavage of 45/47-kDa pro-DrCaspase-A/-B
of DrASC dramatically promoted (p ⬍ 0.001) DrCaspase-B but into a 35-kDa hydrolytic product (p35) if pro-DrCaspase-A/-B
not DrCaspase-A activation, with a maximum up-regulation of was coexpressed with DrNLRP3 and DrASC (Fig. 2, D and E).
up to 180% (Fig. 2B). The DrNLRP3-⌬PYD and DrNLRP3- By contrast, no p35 was detected without the coexpression of
⌬NACHT (lacking both NACHT and FISNA) mutant proteins either DrNLRP3 or DrASC. The DrNLRP3 mutants that lacked
significantly impaired (p ⬍ 0.01 or p ⬍ 0.001) DrCaspase-A and PYD, NACHT-FISNA, and LRR domains failed to induce pro-
Figure 2. DrNLRP3 involvement in the activation of DrCaspase-A and DrCaspase-B. A, DrNLRP3 and DrASC activate DrCaspase-A detected by the specific
Ac-YVAD-AFC fluorescent substrate. Each data point shows the mean ⫾ S.D. with three replicates. *, p ⬍ 0.05. B, DrNLRP3 and DrASC activate DrCaspase-B
detected by specific Ac-WEHD-AFC fluorescent substrate. Each data point shows the mean ⫾ S.D. with three replicates (*, p ⬍ 0.05; **, p ⬍ 0.01; ***, p ⬍ 0.001).
C, DrNLRP3 and its domain-lacking mutants simultaneously activate DrCaspase-A and DrCaspase-B detected by Ac-YVAD-AFC or Ac-WEHD-AFC fluorescent
substrates. Data are representative of three independent experiments as mean ⫾ S.D. (*, p ⬍ 0.05; ***, p ⬍ 0.001). D and E, Western blotting assay of
DrCaspase-A (D) and DrCaspase-B (E) auto-hydrolyzation when coexpressed with DrNLRP3 and DrASC. F, Western blotting assay of the DrCaspase-A and
DrCaspase-B auto-hydrolyzation when coexpressed with DrNLRP3 mutants and DrASC. G, IP assay shows the DrNLRP3 interaction with DrASC through the PYD
domain. HEK293T cells were transfected with pCMV-Tag2B–DrNLRP3/DrNLRP3-⌬PYD and pCMV-HA-DrASC for 48 h. Cell lysates were immunoprecipitated
with rabbit anti-Flag Ab and analyzed by Western blotting by using mouse anti-Flag or anti-HA against DrNLRP3 or DrASC, respectively (top panel). Expression
of the transfected plasmids was analyzed with anti-Flag or anti-HA Ab in the whole-cell lysates (bottom panels). H, coIP assay shows the protein–protein
interactions among DrCaspase-A (CasA-5DA), DrASC (DrASC-⌬PYD/⌬CARD), and DrNLRP3 (WT). I, coIP assay reveals the protein–protein interactions among
DrCaspase-B (CasB-4DA), DrASC (DrASC-⌬PYD/⌬CARD), and DrNLRP3 (WT). The results are representative of three independent experiments. Caspase activity
was detected and expressed as the fold induction over the control as described under “Experimental procedures.”
Figure 3. Aggregation of DrASC-dependent DrNLRP3 inflammasome. A, transient transfection of pCMV-Myc-DrASC or pCMV-Tag2B-DrNLRP3 in HEK293T
cells, and diffuse fluorescent signals were detected in the cells. B, DrNLRP3 and DrASC coexpression in HEK293T cells. Flag-DrNLRP3 signal (red) and Myc-DrASC
signal (green) accumulate in the same speck. C, confocal microscopy image of DrNLRP3-DrASC speck in ZF4 cells transfected with pCMV-Myc-DrASC and
pCMV-Tag2B-DrNLRP3 by electroporation. D, statistics of DrASC speck-forming rates induced by DrNLRP3 and its mutants. More than 100 cells with DrASC
speck were counted in each experimental group to quantify DrNLRP3-dependent DrASC nucleation. Fig. S2E shows the original immunofluorescence images.
Data are representative of three independent experiments as mean ⫾ S.D. (**, p ⬍ 0.01). E, immunofluorescence examination of DrASCPYD/CARD filament when
transiently transfected by Myc-tagged DrASC-PYD (DrASC-⌳CARD) or DrASC-CARD (DrASC-⌳PYD) in HEK293T cells. F and G, with the DrNLRP3 and DrASC
coexpression, DrNLRP3 was colocalized with the DrASCPYD filament (F) but not with the DrASC CARD filament (G). H, DrNLRP3 coexpressed with DrASC-⌳CARD
or DrASC-⌳PYD cannot activate DrCaspase-A when being detected by the specific Ac-YVAD-AFC. Each data point shows the mean ⫾ S.D. with three replicates
(*, p ⬍ 0.05). I, DrASC-⌳PYD but not DrASC-⌳CARD interacts with the linker ASC and inhibits the DrCaspase-A activation by DrNLRP3 and DrASC. Images were
captured under a laser-scanning confocal microscopy (Zeiss LSM-710; original magnification, ⫻630; scale bars represent 5 or 10 m). The results are represen-
tative of three independent experiments as mean ⫾ S.D. (*, p ⬍ 0.05).
Figure 4. Recruitment of DrCaspase-A/-B into DrNLRP3-DrASC inflammasome in a sequential manner. A and B, DrNLRP3-HA, DrASC-Myc, and DrCaspase-
A-Flag (A) or DrCaspase-B-Flag (B) coexpression in HEK293T cells elicited the DrCaspase-A/-B colocalization with the DrASC speck. C, DrNLRP3-HA, DrASC-HA,
DrCaspase-A-Myc, and DrCaspase-B-Flag coexpression in HEK293T cells elicited the formation of DrCaspase-B (white arrowheads) or DrCaspase-A (white arrows)
specks in the cells. D, coexpression of DrNLRP3-HA, DrASC-HA, aPYD–CasB-Flag, and bPYD–CasA-Myc in HEK293T cells elicited the formation of aPYDCasB
(white arrowheads) or bPYDCasA (white arrows) specks in the cells. E–G, FRAP of DrNLRP3 inflammasome. Bleaching was performed after HEK293T cells stably
expressing DrNLRP3–GFP, DrASC–GFP, DrCaspase-A–RFP, or DrCaspase-B–RFP. Time-lapse micrographs of DrNLRP3–DrASC–DrCaspase-A (E), DrNLRP3–
DrASC–DrCaspase-B (F), and DrNLRP3–DrASC (G) punctum formation after bleaching. Arrows indicate punctum. Scale bar, 5 or 10 m. These images are
representative of at least 10 photobleached cells mentioned previously. H, fluorescence intensities of DrNLRP3–DrASC and DrCaspase-A/-B specks over the
time course of 5 min after bleaching. Each data point shows the mean ⫾ S.D. with at least three replicates.
Figure 5. DrNLRP3 contribution to the proDrIL-1 maturation in a DrASC-dependent manner. HEK293T cells were transfected with a pcDNA3.1–DrIL-1
construct alone or with pCMV–DrNLRP3, pCMV–DrNLRP1, pCMV–DrASC, pCMV–DrCaspase-A, and pCMV–DrCaspase-B. At 24 h post-transfection, immunoblot
analysis was performed on the cell lysates with mouse anti-Flag or anti-Myc monoclonal Ab. A, ProDrIL-1 cleavage triggered by activated DrCaspase-A and
DrCaspase-B. B, ProDrIL-1 cleavage triggered by DrNLRP3–DrASC-activated DrCaspase-A alone. C, ProDrIL-1 cleavage triggered by DrNLRP3–DrASC-acti-
vated DrCaspase-B alone. Blots were re-probed for glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as a loading control. The results are representative
of three independent experiments, as described under “Experimental procedures.” Bar charts under A–C showed the relative density of the cleavage product
of DrIL-1 in the blots. Each data point shows the mean ⫾ S.D. with three replicates.
(the first cleavage product at the Asp104 residue of proDrIL-1) activated DrCaspase-B alone failed in the proDrIL-1 cleavage
by the activated DrCaspase-A alone (Fig. 5B). This product (Fig. 5C). In the absence of DrNLRP3 and/or DrASC, no any
coexisted with several other mid-forms (25–30 kDa) known as mature/partial forms of proDrIL-1 (18/20 kDa) and
the cleavage products of some other proteases existing in cells, DrCaspase-A/-B (35 kDa) were detected. These findings indi-
including neutrophil elastase, proteinase 3, cathepsins G/D, cated that DrNLRP3-DrASC inflammasome contributed to
granzyme A, and matrix metalloproteinases. By contrast, the pro-DrCaspase-A/-B activation through self-hydrolyzation,
Figure 7. Coordination of DrNLRP3 inflammasome between mature-formed DrIL-1 secretion and cell pyroptosis. A, HEK293T cells were transfected
with a pcDNA3.1-DrIL-1 construct with pCMV-DrNLRP3, pCMV-DrASC, pCMV-DrCaspase-A (pCMV-CasA-5DA), pCMV-DrCaspase-B (pCMV-CasB-4DA), and
pCMV-DrGSDMEb. Immunoblot analysis was performed at 24 h post-transfection on the cell lysates to detect the proDrIL-1 cleavage triggered by DrNLRP3
inflammasome. B, supernatants from the indicated cells were analyzed for cell death, as measured by the LDH release. C, levels of soluble IL-1 in culture
supernatants were analyzed by ELISA by using DrIL-1 polyclonal antibody. Each data point shows the mean ⫾ S.D. with three replicates. **, p ⬍ 0.01;
***, p ⬍ 0.001.
seA-5DA and CaspaseB-4DA together or with the DrASC DrGSDMEb expression did not influence the DrIL-1 matura-
removed from the combination. Without the DrNLRP3 expres- tion but markedly decreased the cytotoxicity and DrIL-1
sion, no detectable cytotoxicity, DrIL-1 maturation, and release. A schematic diagram for the two types of DrNLRP3
secretion were observed. Meanwhile, the absence of inflammasome is presented in Fig. 8.
Figure 6. DrNLRP3 activates DrCaspase-B to cleave DrGSDMEa/b in a DrASC-independent manner. A, HEK293T cells were transfected with different
combinations of plasmids of DrNLRP3, DrASC, DrCaspase-A/-B, and DrGSDMEa/b. Supernatants from the indicated cells were analyzed for cell death, as
measured by LDH release. B, images were taken after 4.5 M PI were added to the indicated cells. The dyed cells indicate the loss of plasma membrane integrity
and exhibit pyroptotic-like features. Scale bar, 10/100 m. C, DrGSDMEa/b cleavage by DrNLRP3 and DrCaspase-B (or the mutant CasB-4DA) was analyzed by
immunoblotting. D, DrNLRP3 activating DrCaspase-B or CasB-4DA was detected by specific Ac-WEHD-AFC fluorescent substrate. E, supernatants from the
indicated cells were analyzed for cell death, as measured by the LDH release. F, supernatants from the indicated cells coexpressed with different combinations
of DrNLRP3 and DrCaspase-B (including CaspB-P35, CaspB-P20, and CaspB-P10) were analyzed for cell death, as measured by LDH release. G, DrGSDMEb
cleavage by DrNLRP3 and DrCaspase-B in different combinations of CaspB-P35, CaspB-P20, and CaspB-P10 was analyzed by immunoblotting. H, supernatants
from the HEK293T cells transfected with PCMV-GSDMEb/Caspase-B/aPYD–CasB/NLRP3 were analyzed for cell death, as measured by LDH release. I, protein–
protein interactions between DrNLRP3 and DrCaspase-B but not their PYD-lacking mutants. J, DrNLRP3 and mutants activate DrCaspase-B detected by specific
Ac-WEHD-AFC fluorescent substrate. K, supernatants from the indicated cells were analyzed for cell death, as measured by the LDH release. All the above results
are representative of at least three independent experiments, and error bars denote the S.D. of triplicate wells. *, p ⬍ 0.05; **, p ⬍ 0.01; ***, p ⬍ 0.001.
Figure 8. Schematic for the functional roles of DrNLRP3 inflammasome in DrASC-dependent and DrASC-independent manners.
In vivo examination of DrNLRP3 inflammasome LPS and H2O2 but not by muramyl dipeptide (MDP), bacte-
DrNLRP3 inflammasome was evaluated in vivo in the rial DNA, and ATP, although the latter exhibited stimulatory
zebrafish embryo model. The DrNLRP3 inflammasome occur- effects on DrCaspase-A/-B activation at varying degrees in
rence is optimized at 6 hpf embryos or 72 hpf larvae with the embryos without DrNLRP3 knockdown (Fig. 9, K and L).
Edwardsiella tarda immersion infection (108 CFU/ml) for 1– 4 This finding suggests the contribution of LPS and cellular
h as determined by significantly increased DrCaspase-A/-B oxidation to DrNLRP3 inflammasome activation. Next, cell
activity at those times (Fig. 9, A and B). DrNLRP3 overexpres- pyroptosis and DrIL-1 maturation, two downstream events
sion alone in embryos with infection only augmented endog- of the DrNLRP3 inflammasome, were examined. The pyrop-
enous DrCaspase-B activation. However, DrNLRP3 coex- tosis occurrence was optimized in 72 hpf embryos with
pression with DrASC promoted both DrCaspase-A and E. tarda infection (108 CFU/ml) for 4 h. Therein, the expres-
DrCaspase-B activation, accompanied by an increased sion of DrNLRP3, DrCaspase-B, and DrGSDMEb genes but
DrIL-1 maturation (Fig. 9C). These observations support not apoptosis-associated genes (such as Fas/FasL and Bcl2)
the existence of DrASC-dependent and DrASC-independent was significantly (p ⬍ 0.001) augmented, and considerable
types for DrCaspase-A/-B activation and DrIL-1 matura- cells presented pyroptotic morphology (Figs. 9M and 10A).
tion. Structurally, DrNLRP3- and DrASC-colocalized specks FCM analysis showed that the percentage of the PI-positive
were clearly observed in the embryos (Fig. 9D). DrNLRP3 cells increased from 8.80 to 34.8% in the embryos upon infec-
knockdown significantly inhibited (p ⬍ 0.05) DrCaspase- tion. This value decreased to 19.7% in DrNLRP3 morphants.
A/-B activation upon E. tarda infection. This inhibition was This result implied the involvement of DrNLRP3 in pyrop-
rescued by MO-resistant mRNAs in a dose-dependent man- tosis (Fig. 10B). However, DrIL-1 maturation was sup-
ner (Fig. 9, E, F, H, and I and Fig. S5A). Correspondingly, pressed in DrCaspase-A and DrCaspase-B morphants (Fig.
DrNLRP3 knockdown increased the mortality of embryos 10C), whereas cell pyroptosis declined in DrCaspase-B mor-
with E. tarda infection, which was restored by MO-resistant phants (13.2%) but not in DrCaspase-A morphants (30.6%)
mRNAs (Fig. 9, G and J). In addition, DrCaspase-A/-B acti- (Fig. 10D). This result indicated that pyroptosis depends on
vation was abrogated in DrNLRP3 morphants stimulated by DrCaspase-B rather than DrCaspase-A. Furthermore, a
Figure 10. In vivo examination of DrNLRP3 inflammasome in DrASC-independent manner. A, images were taken after PI was added to the 72-hpf
zebrafish larval cells infected by E. tarda (108 CFU/ml) for 4 h. The dyed cells exhibit pyroptotic-like features. Scale bar, 10 m. The sample number for each
group was 20 –100 zebrafish larvae, and each image is representative of three independent experiments. B, flow cytometry with PI detected the cell pyroptosis
in 72-hpf zebrafish larvae (n ⫽ 100) with E. tarda immersion infection (108 CFU/ml) for 4 h. C, DrCaspase-A (CasA-MO) or DrCaspase-B (CasB-MO) knockdown
decreased the DrCaspase-A/-B activation and the DrIL-1 maturation in zebrafish embryos (n ⫽ 100) after E. tarda infection. Data are representative of three
independent experiments as mean ⫾ S.D. (**, p ⬍ 0.01). D, flow cytometry with PI detected the cell pyroptosis in CasA-MO or CasB-MO zebrafish larvae (n ⫽
100) with E. tarda immersion infection (108 CFU/ml) for 4 h. E and F, DrASC knockout (ASC-KO) decreased the DrCaspase-A/-B activation (E) and the DrIL-1
maturation (F) in zebrafish embryos (n ⫽ 100) after E. tarda infection. Data are representative of three independent experiments as mean ⫾ S.D. (*, p ⬍ 0.05).
G, flow cytometry with PI detected the cell pyroptosis in ASC-KO zebrafish larvae (n ⫽ 100) with E. tarda immersion infection (108 CFU/ml) for 4 h.
Figure 9. In vivo determination of DrNLRP3 inflammasome. A and B, fluorogenic substrate detection of the DrCaspase-A/-B activation in 6-hpf embryos (A)
or 72 hpf larvae (B) after E. tarda infection at 108 CFU/ml for 0 – 6 h. The sample number for each group was 20 –100 zebrafish embryos or larvae, and each data
point shows the mean ⫾ S.D. with three replicates. C, DrNLRP3 and DrASC coexpression in vivo increased the DrCaspase-A/-B activation level and promoted the
DrIL-1 maturation under E. tarda infection in 72-hpf larvae (n ⫽ 100). Data are representative of three independent experiments as mean ⫾ S.D. (*, p ⬍ 0.05;
**, p ⬍ 0.01; ***, p ⬍ 0.001). D, Flag-tagged DrNLRP3 and Myc-tagged DrASC coexpression in vivo triggers the DrNLRP3-DrASC speck nucleation in 72-hpf
zebrafish larvae (n ⫽ 100). E and H, DrNLRP3 knockdown by DrNLRP3-MO decreased the DrCaspase-A/-B activation in 6-hpf embryos (n ⫽ 50) (E) or 72 hpf larvae
(n ⫽ 50) (H) after E. tarda infection for 40 min or 4 h. Each data point shows the mean ⫾ S.D. with three replicates (*, p ⬍ 0.05; **, p ⬍ 0.01). F and I, MO-resistant
DrNLRP3 mRNA rescued the DrCaspase-A/-B activation in 6-hpf embryos (n ⫽ 50) (F) or 72-hpf larvae (n ⫽ 50) (I) after E. tarda infection for 40 min or 4 h. Each
data point shows the mean ⫾ S.D. with three replicates (*, p ⬍ 0.05; **, p ⬍ 0.01). G and J, RSRs of 6-hpf embryos (G) or 72-hpf larvae (J) after E. tarda infection
at 106 CFU/ml for 12 h. Zebrafish embryos were microinjected with standard MO (Control), DrNLRP3-MO (NLRP3-MO), or both with the corresponding mRNA
(NLRP3-(MO⫹mRNA)). Mortality in each group was monitored during the 1-h period at one interval. The results are performed in triplicate with 100 embryos per
group. K and L, evaluation of bacterial LPS, MDP, and DNA and cellular metabolites H2O2 and ATP for the DrNLRP3 inflammasome activation in 6-hpf embryos
(n ⫽ 50) via in vivo knockdown assay. Each data point shows the mean ⫾ S.D. with three replicates (***, p ⬍ 0.001). M, fold change of mRNA levels of the genes
involved in pyroptosis and apoptosis in zebrafish 72-hpf larvae (n ⫽ 20) after E. tarda infection for 4 h. The fold change of the relative expression levels was
calculated by the 2⫺⌬⌬Ct method with -actin for normalization. Data are representative of three independent experiments as mean ⫾ S.D. (*, p ⬍ 0.05; **, p ⬍
0.01; ***, p ⬍ 0.001).
Figure 11. Functional substitution of mouse NLRP3 (MmNLRP3) to DrNLRP3 in DrASC nucleation and cell pyroptosis. A, DrASC and DrNLRP3 coexpres-
sion in HEK293T cells elicits DrASC speck aggregation. B, DrASC and MmNLRP3 coexpression instead of DrNLRP3 also elicits DrASC speck aggregation. C,
speck-forming rates of DrNLRP3 or MmNLRP3 with DrASC. Data are representative of three independent experiments as mean ⫾ S.D. (**, p ⬍ 0.01). D, DrNLRP3
or MmNLRP3 directly activates DrCaspase-B and triggers DrGSDMEb-dependent cell pyroptosis. Images were captured under a laser-scanning confocal
microscopy (Zeiss LSM-710; original magnification, ⫻630, scale bars represent 50 or 10 m). The DrCaspase-B activity was detected and expressed as the fold
induction over the control as described under “Experimental procedures.” Each data point shows the mean ⫾ S.D. with three replicates. *, p ⬍ 0.05; **, p ⬍ 0.01;
***, p ⬍ 0.001.
diminished by antisense MO-based DrNLRP3 knockdown and independent but DrCaspaseB-dependent pyroptotic cell death
rescued by MO-resistant mRNAs. Meanwhile, the high mortal- (Fig. 11). This result implies the conservation between fish and
ity of DrNLRP3 morphants with E. tarda infection is restored mammalian NLRP3 inflammasomes throughout vertebrate
by administering MO-resistant mRNAs. These findings verify evolution.
the functional roles of DrNLRP3 inflammasome in antibacterial In conclusion, our study demonstrates the origin of NLRP3
immunity in vivo. Furthermore, by generating a DrASC inflammasome in teleost fish and reveals its role in ASC-depen-
CRISPR mutant (DrASC⫺/⫺) with a 2-bp deletion in zebrafish dent IL-1 maturation and GSDME-mediated pyroptosis. This
by using Cas9/gRNA technology, it was found that the finding enriches our current knowledge on NLRP3 inflam-
DrASC⫺/⫺ mutant only abrogates the DrIL-1 maturation but masome biology. Given that the NLRP3 inflammasome is also
maintains the ability of cell pyroptosis. This result confirms the closely-associated with various diseases, such as type II diabetes
existence of a DrASC-independent pyroptosis pathway in vivo. and inflammatory bowel disease (41, 42), teleost fish will
Mechanistically, LPS and H2O2 administration into the become a new research model for these kinds of diseases.
embryos significantly activates the DrNLRP3 inflammasome.
This outcome suggests that pathogen-associated molecular Experimental procedures
patterns and cellular metabolic homeostasis (such as redox
state) can elicit the DrNLRP3 inflammasome activation as Experimental fish and embryo
observed in mammalian NLRP3 inflammasomes (39, 40). Inter- WT AB zebrafish (Danio rerio) with body lengths of 3– 4 cm
estingly, the mouse NLRP3 (MmNLRP3) can replace DrNLRP3 and weights of 0.5–1.0 g were maintained in circulating water at
for DrASC-dependent inflammasome aggregation and DrASC- 28 °C under standard conditions. The fish that exhibited