You are on page 1of 10

Ecotoxicology and Environmental Safety 258 (2023) 114963

Contents lists available at ScienceDirect

Ecotoxicology and Environmental Safety


journal homepage: www.elsevier.com/locate/ecoenv

Toxicological mechanism of triptolide-induced liver injury:


Caspase3-GSDME-mediated pyroptosis of Kupffer cell
Chenyang Han a, Hongyan Pei b, Yongjia Sheng a, Jin Wang a, Xiaohong Zhou a, Wenyan Li a,
Li Guo c, Yun Kong a, *, Yi Yang a, *
a
Department of pharmacy,The Second Affiliated Hospital of Jiaxing University, 314001, China
b
College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
c
Department of Center Laboratory, The Second Affiliated Hospital of Jiaxing University, China

A R T I C L E I N F O A B S T R A C T

Keywords: Aim: Triptolide (TRI) is an active diterpenoid lactone compound isolated from Tripterygium wilfordii,We focused
Triptolide on investigating the effect and mechanism of Triptolide (TRI) on liver injury.
Liver injury Methods: The toxic dose (LD50 = 100 μM) of TRI on liver Kupffer cells was explored, and network pharmaco­
Kupffer cell
logical analysis was performed to identify Caspase-3 as the target of TRI-induced liver injury. Regarding the
Pyroptosis
pyroptosis research, we examined the level of TRI-induced pyroptosis in Kupffer cells, including inflammatory
Caspase-3
cytokine detection, protein assay, microscopic cell observation and LDH toxicity test. The effect of TRI on
pyroptosis was assessed after knocking out GSDMD, GSDME and Caspase-3 in cells, respectively. We also
investigated the liver injury-inducing action of TRI at the animal level.
Results: Our experimental results were consistent with those predicted by network pharmacology, indicating that
TRI could bind to Caspase-3-VAL27 site to promote the cleavage of Caspase-3, and Cleaved-Caspase-3 induced
pyroptosis of Kupffer cells through GSDME cleavage. GSDMD was not involved in TRI’s action. TRI could pro­
mote Kupffer cell pyroptosis, elevate the inflammatory cytokine levels, and facilitate the expressions of N-GSDME
and Cleaved-Capase 3. After the mutation of VAL27, TRI could not bind to Caspase-3. Animal-level results
showed that TRI could induce liver injury in mice, while Caspase-3 knockout or Caspase-3 inhibitors could
antagonize the action of TRI.
Conclusion: We find that the TRI-induced liver injury occurs primarily through the Caspase-3-GSDME pyroptosis
signal. TRI can promote Caspase − 3 maturation and regulate kupffer cell pyroptosis. The present findings offer a
new idea for the safe use of TRI.

1. Introduction elicitation of immune injury, among which immunological liver injury


has been a research focus in recent years (Mei et al., 2005; Huo et al.,
Triptolide (TRI) is an active diterpenoid lactone compound isolated 2019). A single intake of 300–1000 μG/kg TRI can cause evident liver
from Tripterygium wilfordii (Guan et al., 2021), which possesses signifi­ injury (Huang et al., 2019). In animal experiments, serum alanine
cant immunomodulatory and antiinflammatory activities and is widely aminotransferase (ALT) and aspartate aminotransferase (AST) were
used in the clinical treatment of arthritis and immune diseases (Zhou significantly elevated in the TRI model group, and the intrahepatic
et al., 2021; Liu et al., 2021),TRI can also cause tissue damage in high release of TNF-α and NO was increased, suggesting the occurrence of
doses and long-term use, such as liver damage, which is common. immune injury (Huang et al., 2020a). Additionally, TRI can promote the
However, its toxicity cannot be underestimated, especially hepatotox­ occurrence of inflammation by regulating interleukin (IL)− 17, IL-6 and
icity, which is the main cause of death from it (Yuan et al., 2021). nuclear factor-kappa B (NF-κB) (Nan et al., 2021).
Studies have shown that TRI can cause liver injury through a variety of Although TRI has previously been found to promote inflammation to
mechanisms, such as apoptosis induction, oxidative stress induction, cause liver injury, its exact target and mechanism of action have yet to
induction of inducible nitric oxide synthase (iNOS) expression and be further clarified. Pyroptosis is a way of inflammatory cell death,

* Corresponding authors.
E-mail addresses: kongyun@jxey.com (Y. Kong), wasd911@126.com (Y. Yang).

https://doi.org/10.1016/j.ecoenv.2023.114963
Received 26 December 2022; Received in revised form 22 April 2023; Accepted 25 April 2023
Available online 30 April 2023
0147-6513/© 2023 Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
C. Han et al. Ecotoxicology and Environmental Safety 258 (2023) 114963

which leads to inflammatory response through the GSDM protein- 2.5. ELISA
mediated maturation of inflammatory cytokines and the development
of cell membrane pores (Bergsbaken et al., 2009). Classical pyroptosis is The Kupffer cells were dynamically detected for inflammatory cy­
mediated by Caspase 1, which cleaves GSDMD and cytokines and is tokines. After TRI treatment, the culture medium was sampled at time
activated by proteolysis prior to pro-IL-1β and pro-IL-18 to produce their points of 1–4–6–12–24 h, centrifuged at 3000 g for 30 min, and then the
proinflammatory forms, IL-1β and IL-18. Nonclassical signals can be supernatant was taken for detection of IL-1β, IL-6 and TNF-α using ELISA
mediated by Caspase-3/4/5/11, GSDMA, GSDMB, GSDE, etc (Bergs­ kit (Jiangcheng Bioengineering Institute, Nanjing, China). After incu­
baken and Cookson, 2007). Our preliminary study found that TRI could bation by adding reagent as per the kit instructions, the absorbance was
promote pyroptosis of Kupffer cells. To further explore the liver measured at 450 nm with an ELISA reader (BioTek, USA), and the results
injury-causing mechanism and target of TRI, we performed network were expressed as pg/ml.
pharmacology and basic experiments to further reveal its target, with a The IL-1β, IL-6 and TNF-α levels in mouse liver tissues were assayed
view to providing support for its safe use. by ELISA kit (Jiangcheng Bioengineering Institute, Nanjing, China). The
liver tissues were cut into pieces with sterile surgical scissors, ground
2. Materials and methods with liquid nitrogen, and lysed on ice with 1.0 ml of RIPA lysate for 30
min. Then, the supernatant was collected for protein quantification as
2.1. Cell culture per the kit instructions, and the results were expressed as pg/ml.

Mouse liver Kupffer cells (Procell Life Science & Technology, Wuhan, 2.6. CCK-8
China) were cultured using a special medium in a three gas incubator set
at 37 ◦ C with 5% CO2. Cell viability was assayed using Trypan blue. The Kupffer cells were seeded into 96-well plates and, 12 h later,
Kupffer cells were subcultured and used for experiments after the treated with TRI. The cellular viability was assayed at 0–1–4–6–12–24 h.
viability reached above 90%. We separately constructed GSDMD-, Next, 10 μl of CCK-8 reagent (Solarbio, Beijing, China) was added per
GSDME- and Caspase-3- knockout Kupffer cells(JIMA Gene,Suzhou, well, and incubation continued for an additional 4 h, followed by
China) by CRISPR-Cas9 technology and verified them by Western-Blot, measurement of OD value at 450 nm.
where sgRNA-GSDMD-CAGGACCAGCCCAGCATTGGAA;sgRNA-GSDME
-GCAAGAGAGAGAACCACAATT; sgRNA-Caspase3-GAGTCTGACTGGA 2.7. Mouse experiment
AAGCCGAA.
In the TRI intervention experiment, we treated cells with gradient SPF C57BL/6 mice (WT) and Caspase-3-knockout mice (KO, C57BL/
concentrations of TRI (0–20–40–60–80–100–120–140–160–180–200 6 J-Casp3em1Cya) were purchased from Cyagen Biotechnology, which
μM) for 24 h, and detected their viability by CCK-8 assay. The results were divided into 10 mice per group. To establish a mouse model of liver
showed that LD50 = 100 μM at 24 h. During the research, the TRI injury, 1000 μg/kg TRI was intragastrically administered once daily for
concentration of Kupffer cells was set to 100 μM and the time was set to 7 d. During the intervention experiment of Caspase-3 inhibitor, 160 ng
24 h. of Z-DEVD-FMK was intragastrically administered once daily in combi­
nation with TRI. The mice were killed 7 d later for detection.

2.2. Flow cytometry (FCM) 2.8. ALT/AST

After 24 h of TRI treatment, the kuppffer cells were trypsinized and The mouse peripheral bloods were extracted after 7 d of consecutive
adherent/suspended cells were collected. The collected cells were then TRI administration. Tail venous bloods were collected, centrifuged, and
merged, washed with precooled PBS for several times and subjected to then the supernatants were taken for detection of ALT and AST by ul­
Annexin V-FITC/PI staining as per the kit (BD Biosciences, New Jersey, traviolet colorimetry (Jiangcheng Bioengineering Institute, Nanjing,
USA) instructions. FCM was used to detect the level of apoptosis, and the China) as per the kit instructions. The AST and ALT results were
results were expressed as %. expressed as U/L.

2.3. LDH assay 2.9. H&E

The cytotoxicity of TRI was detected via the LDH assay kit (Solarbio, The mice were killed after 7 d of consecutive TRI administration.
Beijing, China). After 24 h of Kupffer cell treatment with TRI, the LDH Their liver tissues were embedded in paraffin, and prepared into 4-μm-
release rate was determined as per the kit instructions, and the results thick serial sections for staining as per following procedure: Xylene
were expressed as %. deparaffinization, dehydration with gradient concentrations (100%,
95%, 80%) of alcohol, tap water rinsing for 2 min, hematoxylin staining
for 3 min, tap water rinsing for 2 min, treatment with 1% hydrochloric
2.4. PI staining acid-alcohol for 2 s, tap water rinsing for 2 min, treatment with 1%
ammonia for 20 s, staining with 0.5% eosin alcohol for 10 s, gradient
The relative PI uptake rate of Kupffer cells was detected at alcohol dehydration, xylene permeabilization, neutral gum sealing.
0–1–4–6–12–24 h. The cells were treated with 1 μg/ml PI, 120 nm NaCl, Finally, the histopathological changes of liver tissues were observed
5 mM Glucose, 1.5 mM CaCl2, 1 mm magnesium chloride and 0.1% under light microscope.
bovine serum albumin (BSA). The cellular absorbance was measured at
533/617 nm. PI absorption rate (%) = (ODSample-ODbackground)/ 2.10. IHC and TUNEL
(ODmaximum-ODbackground).
PI staining of cells was performed following 24 h of TRI treatment. The paraffin-embedded liver tissue sections were deparaffinized in
After discarding the medium, the cells were washed twice with PBS, xylene thrice, and soaked in anhydrous alcohol for 5 min and subse­
stained using 1 μg/ml PI reagent (Beyotime Biotechnology, Shanghai, quently in 95–85% ethanol twice. Then, the sections were placed in
China) for 30 min and washed twice with PBS. Positive cells, which 0.01 mol/L citrate buffer (PH= 6.0) and retrieved by microwave at 98 ◦ C
showed red fluorescence, were subjected to DAPI restaining and then for 20 min, followed by cooling at room temperature for 30 min and
observed. rinsing with distilled water. A 10-min incubation with 3% hydrogen

2
C. Han et al. Ecotoxicology and Environmental Safety 258 (2023) 114963

peroxide proceeded at room temperature to eliminate endogenous (2) Construction of "drug-action target" network: Cytoscape 3.7.1
peroxidase. After blocking with 2% BSA, the sections were incubated at was utilized to visualize the "drug-action target" interaction
4 ◦ C with GSDMD and GSDME monoclonal antibodies (1:300 dilutions; network diagram.
Abcam, USA), and further with peroxisase-labeled streptomycin (3) Taking the intersection of drug and disease targets: Disease tar­
(Abcam, USA) for 15 min. After rinsing with phosphate buffer for 5 min gets were collected via the Gene Card database. To eliminate the
thrice, each section was added with freshly prepared DAB solution interference of subsequent experiments by targets lowly associ­
(DAKO, Denmark) for visualization. The chromogenic reaction was ated with disease, those whose relevance scores ≥ median were
observed under a microscope, and then the sections were restained with selected as the disease targets. The potential targets of the drug
hematoxylin, sealed and observed under an Olympus-BX51 microscope. for treating acute hepatic injury were acquired by taking the
During TUNEL staining, the tissue sections were added dropwise with intersection of drug action targets with screened disease targets.
TUNEL reagent, incubated for 30 min, washed with TBST, and then (4) PPI network analysis: The action targets of drug for disease
observed microscopically. treatment were input into the STRING database for PPI network
analysis, where the confidence level was set at 0.7. Target
2.11. Western-blot and pull-down screening was performed, and data were derived. The results
were imported into Cytoscape 3.7.1 to reflect the intensity of
Following 24 h of treatment with TRI, the suspended and adherent target interaction according to the Degree value, and the corre­
Kupffer cells were collected, lysed on ice with 1.0 ml of RIPA buffer sponding targets with Degree values of ≥ twice the median were
(Beyotime Biotechnology, Shanghai, China) for 30 min, and then defined as the core targets. The PPI network diagram was created
centrifuged at 10,000 g for 15 min. The supernatant was taken for by deciding the edge thickness according to the magnitude of
protein quantification using BCA kit (Beyotime Biotechnology, combined score.
Shanghai, China). The volume of protein solution was replenished with (5) Construction of "drug-disease-target" network: The "drug-disease-
5x loading buffer to 20 μl. After boiling for 8 min, electrophoresis was target" interaction network diagram was created via Cytoscape
performed at 80 V and subsequently at 120 V. Next, gel was removed, 3.7.1, and the degree of "drug-disease-target" interaction was
and the proteins were electrotransferred onto PVDF membranes at a 300 obtained via the network analyzer function, thereby clarifying
mA constant current for 0.5–2 h. Thereafter, the PVDF membranes were potential targets through which can the drug treat acute hepatic
blocked with 5% skimmed milk powder for 2 h, incubated with TBST- injury, as well as the intensity of their interaction.
diluted monoclonal primary antibody, washed twice in TBST, and (6) GO enrichment analysis: Utilizing the Metascape database, a
further incubated with horseradish peroxidase-labeled goat anti-rabbit comprehensive enrichment analysis was conducted on gene
secondary antibody (Abcam, USA), followed by chemiluminescence ontology (GO) terms, including GO Biological Process, GO Mo­
detection. OD was analyzed via Image Pro-Plus 6.0, and the results were lecular Function and GO Cellular Component.
expressed as OD comparisons between the target protein and the in­
ternal reference GAPDH. The proteins in tissues were detected by
consulting the aforementioned method. 2.12.1. Statistical methods
Pull-down assay for TRI-Biotin binding to Caspase-3. The SUMO- All measurement data were expressed as x ±SD, and analyzed and
labeled Caspase-3 protein was expressed in E. coli overnight. After in­ processed via SPSS 17.0. After homogeneity test for variance, compar­
duction with β-D-thiogalactopyranoside, the OD600 was 0.7–0.8. Cells ison between two sets of data was made by two independent samples t-
were suspended in Tris-HCl and NaCl, and then collected, ultra­ test. Three or more sets of data were analyzed by one-way ANOVA, and
sonicated, centrifuged and purified on Ni column. On the column, His- the subsequent pairwise comparison between groups was made by LSD
SUMO-Caspase-3 protein was washed twice with 500 mM of imid­ method. All of the above tests were two-sided, and the differences were
azole, while His-SUMO tag was cleaved with SUMO protease (ULP-1) considered statistically significant when P < 0.05.
and subjected to Ni column purification. The purified protein was
separated by SDS-Page and verified by Coomassie blue staining. Re­ 3. Results
combinant protein (15 μg) was bound to Biotin-labeled TRI for pull-
down with streptavidin beads (Sigma, Massachusetts, USA). The beads 3.1. TRI induced pyroptosis of Kupffer cells
were blocked in 5% BSA in Tris buffer, while protein was precleared
with beads that were subsequently discarded. Following incubation with We treated Kupffer cells with gradient concentrations of TRI, and
50 μM of Biotin-MAF, the magnetic beads were washed thrice in Tris found that cell viability was weakened significantly as the concentration
buffer, and then boiled using the buffer and β-mercaptoethanol. The rose, with LD50 = 100 μM during a 24-h treatment duration [Fig. 1A].
recombinant G protein magnetic beads were incubated with Caspase-3 Accordingly, we adopted a TRI amount of 100 μM and a processing time
antibody (Abcam, Massachusetts, USA). After washing with Tris of 24 h during the experiment. TRI could induce Kupffer cell damage, as
buffer, Caspase-3 was detected according to the aforementioned manifested by significantly higher cell mortality than that in the DMSO
Western-Blot procedure, while biotin was detected using the horseradish group [Fig. 1B]. TRI induced pyroptosis of Kupffer cells, and the LDH
peroxidase-conjugated antibiotin antibody (CST, Boston, USA). release rate was significantly higher than that in the DMSO group
[Fig. 1C]. Meanwhile, the rate of PI uptake rose significantly over time
2.12. Network pharmacology [Fig. 1D]. Detection of inflammatory cytokines also revealed that TRI
could induce the release of Kupffer inflammatory cytokines. In DMSO
During the analysis of TRI and liver injury targets, a combination of group, the cytokine levels did not change significantly over time, while
PubChem database (https://pubchem.ncbi.nlm.nih.gov/), HERB data­ in TRI group, the IL-6, IL-1β and TNF-α levels were significantly upre­
base (http://herb.ac.cn/), Gene Card database (www.genecards.org/), gulated, showing higher values than those in the DMSO group [Fig. 1E-
STRING (https://string-db.org/), Cytoscape 3.7.1 software and Meta­ G]. TRI could also promote the opening of Kupffer cell membrane pores,
scape Database (https://metascape.org/) were used. as manifested by the significantly stronger fluorescence intensity in PI
staining compared to the DMSO group [Fig. 1H]. I detected the
(1) Acquisition of active constituent and action targets: The CAS pyroptosis-related proteins, finding that N-GSDMD and Cleaved-Caspase
number of active constituent was obtained based on PubChem 1 were almost unexpressed, while N-GSDME and Cleaved-Caspase-3
database, and the targets of its action were derived based on were significantly upregulated, TRI group, in particular, exhibited
HERB database. significantly higher levels of expression compared to the DMSO group

3
C. Han et al. Ecotoxicology and Environmental Safety 258 (2023) 114963

Fig. 1. TRI induced pyroptosis of Kupffer cells A: TRILD50 experimental results. LC50 = 100 μM B: FCM results (n = 3). TRI induced apoptosis, as manifested by
higher cell mortality than that in the DMSO group. Inter-group comparison, **P < 0.05. C: LDH release rate (n = 3). TRI group exhibited significantly higher rate of
LDH release compared to the DMSO group. Inter-group comparison, **P < 0.05. D: Relative uptake rate of PI (n = 3). With the prolongation of time, the PI uptake rate
was significantly elevated, which was higher than that in the DMSO group. Inter-group comparison, **P < 0.05. E-G: ELISA (n = 3). The inflammatory cytokine levels
in TRI medium increased significantly in a time-dependent manner. Inter-group comparison, **P < 0.05. H: PI staining (n = 3). The PI fluorescence intensity in the
TRI group was significantly stronger than that in the DMSO group. I-J: Relative protein expression (n = 3). N-GSDMD and Cleaved-Caspase 1 were almost unex­
pressed, while N-GSDME and Cleaved-Caspase-3 were significantly upregulated. TRI group, in particular, exhibited significantly higher expression levels than the
DMSO group. Inter-group comparison, **P < 0.05.

Fig. 2. Network pharmacological analysis of TRI and liver injury A: Interaction analysis of TRI and liver injury targets. B, E, F: Analysis and scoring of targets, among
which Caspase-3 was highly likely to interact with TRI. C-D: Construction of PPI and drug-target networks.

4
C. Han et al. Ecotoxicology and Environmental Safety 258 (2023) 114963

[Fig. 1I-J]. Thus, I speculated that TRI mediated the Kupffer cell F]. IL-1 β Maturity may not depend on GSDME signals and may be
pyroptosis through Caspase-3-GSDME. related to the activation of inflammasome. TRI could facilitate the
opening of cell membrane pores, as manifested by the significantly
3.2. Network pharmacological analysis of TRI and liver injury stronger fluorescence intensity of PI than that in the NC and GSDMD-/-
[Fig. 3G]. Assay of pyroptosis-related proteins found knockout of
The interaction between TRI and liver injury targets revealed the GSDMD, which affected neither the N-GSDME nor the Cleaved-Caspase-
presence of 55 coincidental protein targets between the two [Fig. 2A]. 3 expression [Fig. 3H-I].
Further, targets were bound to TRI and scored. The results showed that
nine proteins, including AKT1, Caspase-3 and CTNNB1, had strong 3.4. Knockout of GSDME or Caspase-3 could inhibit the TRI-induced
binding to TRI, especially Caspase-3, which exhibited a high possibility pyroptosis
of binding to small molecules [Fig. 2B, G]. After constructing the PPI
network, I considered that Caspase-3 was highly likely to be the action I performed GSDME knockout on Kupffer cells, and found that the
target of TRI [Fig. 2C-E]. Hence, through network pharmacological TRI-induced pyroptosis was inhibited. TRI could still induce the decline
analysis, I considered Caspase-3 as the action target of TRI. of cell viability, while insignificant viability downregulation was noted
in GSDME-/- and NC cells [Fig. S1A]. However, the LDH release rate in
3.3. GSDMD knockout did not affect the TRI-induced pyroptosis TRI-GSDME-/- differed insignificantly from that in NC and GSDME-/-
[Fig. S1B]. The relative uptake rate of PI in TRI-GSDME-/- and NC
I first knocked out GSDMD, a classical pyroptosis executive protein, differed insignificantly from that in GSDME-/- [Fig. S1C]. No significant
in Kupffer cells, and found that the cell viability was unaffected after changes were observed in the levels of inflammatory cytokines in TRI-
GSDMD knockout. Cell viability differed insignificantly between NC and GSDME-/-, which differed insignificantly from those in NC and GSDME-/-
GSDMD-/-, although TRI could still induce cell damage. Meanwhile, the [Fig. S1D-F]. After PI staining, insignificant alterations were observed in
cell viability in TRI+GSDMD-/- was significantly downregulated the PI fluorescence intensity between the three groups of cells, and TRI
[Fig. 3A]. The LDH release rate in TRI+GSDMD-/- was also higher than could induce neither the cell membrane pore opening nor the PI uptake
that in NC and GSDMD-/- [Fig. 3B]. The relative uptake rate of PI [Fig. S1G]. Protein assay revealed that the GSDME knockout did not
increased significantly over time in TRI+GSDMD-/-, while changed influence the cleavage or expression of Cleaved-Caspase-3 [Fig. S1H-I].
insignificantly in NC and GSDMD-/-, showing low PI uptake levels TRI acted on Caspase-3 instead of on GSDME.
[Fig. 3C]. Detection of inflammatory cytokines also revealed that TRI Caspase-3 knockout was performed on Kupffer cells, and the results
could promote the upregulation of IL-6, IL-1β and TNF-α in a time- showed that the TRI-induced pyroptosis could also be inhibited after the
dependent manner and was unaffected by GSDMD. Meanwhile, insig­ knockout of Caspase-3. TRI could still induce the decline of cell viability,
nificant cytokine expressions were noted in NC and GSDMD-/- [Fig. 3D- while insignificant viability downregulation was noted in GSDME-/- and

Fig. 3. Knockout of GSDMD did not affect the TRI-induced pyroptosis A: CCK-8 assay (n = 3). TRI could induce cell damage, so that the cell viability in
TRI+GSDMD-/- was significantly downregulated, while the GSDMD knockout did not affect the cell viability. *P < 0.05 vs. NC; #P < 0.05 vs. GSDMD-/-. B: LDH
release rate (n = 3). TRI induced pyroptosis of Kupffer cells, leading to significantly higher rate of LDH release than that in the DMSO group. *P < 0.05 vs. NC;
#
P < 0.05 vs. GSDMD-/-. C: Relative uptake rate of PI (n = 3). TRI could induce the opening of cell membrane pores and promote the uptake of PI, as manifested by
the significantly increased rate of PI uptake over time. *P < 0.05 vs. NC; #P < 0.05 vs. GSDMD-/-. F: ELISA assay (n = 3). TRI could induce the release of Kupffer
inflammatory cytokines, so that the IL-6, IL-1β and TNF-α levels were significantly upregulated over time. *P < 0.05 vs. NC; #P < 0.05 vs. GSDMD-/-. G: PI staining
(n = 3). The fluorescence intensity of PI in TRI was significantly stronger than that in NC and GSDMD-/-. H-I: Relative protein expression (n = 3). GSDMD knockout
affected the expression of neither N-GSDME nor Cleaved-Caspase-3, and TRI could still induce the expression of these two proteins. *P < 0.05 vs. NC; #P < 0.05
vs. GSDMD-/-.

5
C. Han et al. Ecotoxicology and Environmental Safety 258 (2023) 114963

NC cells [Fig. S2A]. The LDH release rate in TRI-GSDME-/- differed 3.5. Caspase-3-KO could inhibit the TRI-induced liver injury in mice
insignificantly from that in NC and GSDME-/- [Fig. S2B]. The relative
uptake rate of PI in TRI-GSDME-/- and NC differed insignificantly from Wild-type (WT) and Caspase-3-knockout (KO) mice were used for
that in GSDME-/- [Fig. S2C]. No significant changes were observed in the research. After intragastric TRI administration for 7 d, I found that the
levels of inflammatory cytokines in TRI-GSDME-/-, which differed ALT and AST levels differed insignificantly between WT and KO mice.
insignificantly from those in NC and GSDME-/- [Fig. S2D-F]. After PI TRI-WT mice exhibited significantly upregulated levels of ALT and AST,
staining, insignificant alterations were observed in the fluorescence in­ which were higher than those in WT and KO mice. TRI-KO mice
tensity of PI between the three groups of cells, and TRI could induce exhibited lower levels compared to the TRI-WT mice [Fig. 4A-B]. Ac­
neither the cell membrane pore opening nor the PI uptake [Fig. S2G]. cording to the tissue damage observations, evident cell damage and le­
Protein assay also revealed that the Caspase-3 knockout influenced the sions appeared in the TRI-WT mouse livers during H&E staining assay,
cleavage and expression of GSDME [Fig. S2H-I]. the current result, I and the TUNEL-positive rate increased significantly. In KO and WT mice,
could further prove that TRI acted on Caspase-3, rather than on GSDME. TUNEL was negatively expressed, while in TRI-KO mice, cell damage
Although our results demonstrate that TRI works through GSDME was markedly ameliorated and the TUNEL-positive rate was reduced
pyroptosis, when GSDME or Caspase-3 is knocked out, TRI is likely to [Fig. 4C].
inhibit cell viability through other signaling pathways. This requires Protein assay revealed that TRI promoted the expression of N-
further exploration. GSDME, although producing insignificant effect on N-GSDMD and
Cleaved-Caspase 1. The Tri-KO mice exhibited lower N-GSDME level
compared to the Tri-WT mice [Fig. 4D-E]. IHC results showed that TIR

Fig. 4. Caspase-3-KO could inhibit the TRI-induced liver injury A-B: ALT and AST (n = 10). Insignificant differences in ALT and AST levels were noted between WT
and KO mice. TRI-WT mice exhibited significantly upregulated levels of AST and AST, which were higher than those in WT and KO mice. Compared to the TRI-WT
mice, these levels were lower in the TRI-KO mice. *P < 0.05 vs. WT; #P < 0.05 vs. KO. C: H&E and TUNEL (n = 5). H&E staining revealed evident cell damage and
lesions in the livers of TRI-WT mice, as well as significantly elevated TUNEL-positive rate. In KO and WT mice, TUNEL was negatively expressed while in TRI-KO
mice, cell damage was markedly ameliorated and the TUNEL-positive rate decreased. D-E: Relative protein expression (n = 5). TRI promoted the expression of N-
GSDME, although producing insignificant effect on N-GSDMD and Cleaved-Caspase 1. The N-GSDME level in TRI-KO was lower than that in TRI-WT. *P < 0.05 vs.
WT; #P < 0.05 vs. KO. F: IHC (n = 5). TIR could promote the expression of GSDME, although producing insignificant effect on GSDMD. Caspase-3-KO could lower the
GSDME expression. G-H: ELISA (n = 5). TRI could promote the IL-6, IL-1β and TNF-α expression in tissues and peripheral bloods. These cytokine levels in TRI-KO
were lower than those in TRI-WT. *P < 0.05 vs. WT; #P < 0.05 vs. KO.

6
C. Han et al. Ecotoxicology and Environmental Safety 258 (2023) 114963

could promote the expression of GSDME, although producing insignifi­ The AST and ALT results showed that TRI could induce liver injury in
cant effect on GSDMD. Caspase-3-KO could lower the GSDME expression WT mice, leading to significantly upregulated ALT and AST levels
[Fig. 4F]. Assay of inflammatory cytokines in liver tissues and peripheral compared to those in WT mice. FMK could antagonize the action of TRI,
bloods found that TRI could promote the IL-6, IL-1β and TNF-α expres­ as manifested by significantly lower AST and ALT levels in TRI+FMK
sion in these sites. The cytokine levels in TRI-KO were lower than those mice than those in TRI mice [Fig. 5A-B]. H&E and TUNEL staining
in TRI-WT [Fig. 4G-H]. revealed that FMK could relieve the severity of liver injury in mice, so
that the TUNEL-positive rate was downregulated, and the degree of cell
3.6. Caspase-3 inhibitor could inhibit the TRI-induced liver injury in mice damage was reduced [Fig. 5C]. Protein assay revealed that FMK
inhibited the expression of N-GSDME and Cleaved-Caspase-3, although
We applied FMK, a Caspase-3 inhibitor, to WT mice, in order to producing insignificant effect on N-GSDMD or Cleaved-Caspase 1
clarify whether inhibiting Caspase-3 could antagonize the action of TRI. [Fig. 5D-E]. However, the irreversible binding of FMK to Caspase-3

Fig. 5. Caspase-3 inhibitor could inhibit the TRI-induced liver injury in mice A-B: ALT and AST (n = 10). TRI could induce liver injury in WT mice, leading to
significantly upregulated ALT and AST levels compared to those in WT mice. FMK could antagonize the action of TRI, as manifested by significantly lower AST and
ALT levels in TRI+FMK mice than those in TRI mice. *P < 0.05 vs. WT; #P < 0.05 vs. TRI. C: H&E and TUNEL (n = 5). FMK could relieve the severity of liver injury
in mice, so that the TUNEL-positive rate was downregulated, and the degree of cell damage was reduced. D-E: FMK inhibited the expression of N-GSDME and
Cleaved-Caspase-3, although producing insignificant effect on N-GSDMD or Cleaved-Caspase 1. *P < 0.05 vs. WT; #P < 0.05 vs. TRI. F: IHC (n = 5). Although
having no effect on GSDMD, FMK could lower the GSDME expression, showing significant difference from the TRI group.G-H: ELISA (n = 5). FMK could lower the
inflammatory cytokine levels in liver tissues and peripheral bloods. *P < 0.05 vs. WT; #P < 0.05 vs. TRI.

7
C. Han et al. Ecotoxicology and Environmental Safety 258 (2023) 114963

inhibits the cutting of GSDME, as Caspase-3 is necessary for GSDME both Triptolide and Caspase-3 had certain binding potential. VAL27 was
cutting.According to IHC results, FMK could lower the expression of the site for Pro-Caspase-3 cleavage, and Cleaved-Caspase-3 was formed
GSDME, although having no effect on GSDMD, with significant differ­ after cleavaging Caspase-3. The binding of TRI to VAL27 promoted the
ence from TRI group [Fig. 5F]. Assay of inflammatory cytokines found maturation and cleavage of Caspase-3 [Fig. 6A-B]. Our Pull-down assay
that FMK could significantly lower the cytokine levels in liver tissues and results showed that TRI-Biotin bound to Caspase-3, rather than to
peripheral bloods compared to the TRI group [Fig. 5G-H]. GSDME. After the mutation of VAL27, TRI could no longer bind to
Caspase-3, proving that Caspase-3-VAL27 was the binding site of TRI
[Fig. 6C-G].
3.7. Validation of targeting relationship between TRI and Caspase-3
RMSF (Root Mean Square Fluctuation) measures the average
movement of atom positions in a molecular structure within a period of
The 3D structure of Caspase-3 protein used for docking was acquired
time, which is the root mean square of distance between a target
from UniProt database (UniProt ID: P70677), while the 3D structure of
structure’s specific atom and the reference structure over the entire time
small-molecule Triptolide used was acquired from PubChem database √̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅
∑ ref 2
(PubChem ID: 107985). Prior to the docking, AVOGADR 1.2.0 was used range. Its specific formula is: RMSFI = T1 N t=1 (ri (t) − ri ) , where ri
ref

to minimize the energy for the 3D structure of small-molecule Triptolide stands for the position coordinates of ith atom in the reference structure,
under the MMFF94 field. Molecular docking herein was performed via ri (t) refers to the position coordinates of ith atom in the target structure
the AutoDock Vina 1.1.2. All receptor proteins were hydrogenated using at time t, and T denotes the time range under computation. The units of
Open Source PyMol before the docking. Then, ADFRsuite 1.0 was RMSF are length units, usually nm (10− 9 m) or Å (10− 10 m). The larger
employed to convert all the processed small molecules and receptor the RMSF value, the greater the change of structural flexibility in the
proteins into the PDBQT format that was necessary for docking via corresponding area, and vice versa. As shown in the Figure, the amino
AutoDock Vina 1.1.2. During the Vina-based docking, the grid box and acid structural flexibility in regions 1–30 and 179–180 of Caspase-3
the PDBQT file of processed proteins and small molecules were used as changed greatly, attributable perhaps to the presence of excessively
the input file. The level of detail of global search for the docking was set many loop zones therein [Fig. 6H].
to 32, while other parameter settings were kept default. Finally, the RMSD (Root Mean Square Distance/Deviation) measures the differ­
docking conformation with highest output score was considered as the ence between varying conformations of the same molecular structure,
bound conformation, which was subjected to molecular dynamics which is the distance root mean square of all corresponding atoms
simulation and analysis. following position overlap of two conformations. Its specific formula is:
The docking between small-molecule Triptolide and Caspase-3 pro­ √̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅̅
∑N ref 2 ref
1
tein was explored with the aid of AutoDock Vina 1.1.2, and their binding RMSD(t) = N i=1 (ri (t) − ri ) , where ri stands for the position
energy scores were derived. Negative binding energy value indicated the coordinates of ith atom in the reference structure, ri (t) refers to the
possibility of binding. As is clear from the Table, the binding affinity position coordinates of ith atom in the target structure at time t,
between Caspase-3 and Triptolide was − 4.4 kcal/mol, implying that

Fig. 6. Validation of the TRI binding to Caspase-3 A-B: Binding mode diagram of TRI and Caspase-3. The binding affinity between Caspase-3 and Triptolide was
− 4.4 kcal/mol, implying that both of them had certain binding potential. VAL27 was the site for Pro-Caspase-3 cleavage, and Cleaved-Caspase-3 was formed after
cleavaging Caspase-3. Meanwhile, the binding of TRI to VAL27 promoted the maturation and cleavage of Caspase-3. C-G: Pull-down assay. TRI-Biotin bound to
Caspase-3, rather than to GSDME. After the mutation of VAL27, TRI could no longer bind to Caspase-3, proving that Caspase-3-VAL27 was the binding site of TRI. H-I:
Model diagram of RMSD and RMSF fitting results.

8
C. Han et al. Ecotoxicology and Environmental Safety 258 (2023) 114963

ref
ri (t) − ri is defined as the distance between two conformations of ith Contrastively, after GSDME knockout, TRI could no longer induce the
atom, and N denotes the total atom number of molecule or the total atom Kupffer cell pyroptosis, which lowered the levels of inflammatory cy­
number of RMSD under computation. The distances for all correspond­ tokines and inhibited the opening of cell pores. However, noteworthy
ing atoms were defined identically and then the root mean square was was that TRI still promoted the expression of Cleaved-Caspase-3. This
calculated, thereby deriving the RMSD value, which measured the indicated that GSDME knockout failed to affect the Caspase-3-
structural difference of the same molecule in different conformations. Its cleavaging effect of TRI, which were independent of each other; be­
units are length units, usually nm (10− 9 m) or Å (10− 10 m). RMSD is sides, GSDME was also not the action target of TRI. Combined with the
commonly used for analyzing the stability of overall molecular structure network pharmacological results, I identified Caspase-3 as the major
of the system. The decrease of RMSD indicated that the structure tended action target of TRI. VAL27 was the cleavaging site of Caspase-3, and
to be stable, and vice versa. It is clear from the results that after 4 ns, the after cleavage of Caspase-3, Cleaved-Caspase-3 was formed, which
RMSD curve of Caspase-3-Triptolide was regionally stabilized at 0.7 nm, further interacted with GSDME to form N-GSDME (Huang et al., 2020b).
the structure of complex changed little, and the simulation process In our study, N-GSDME was also insignificantly expressed after knocking
already reached equilibrium [Fig. 6I]. out Caspase-3, and TRI was unable to enhance the cleavage of GSDME.
Through Pull-down assay, I verified that Caspase-3 was the major action
4. Discussion target of TRI, and that VAL27 was the binding site. After the mutation of
VAL27, TRI could no longer bind to Caspase-3. The fitting results of
As a novel mechanism of programmed cell death, pyroptosis is based RMSD and RMSF also indicated that the TRI-Caspase-3 binding model
structurally on the formation of cell membrane pores, swelling of cells had low energy and high stability.
and rupture of cell membranes, which is characterized mainly by ulti­ During animal experiments, we used WT and Caspase-3-KO mice.
mate expression and release of inflammatory cytokines (Kepp et al., The results showed that TRI could induce liver injury in mice and elevate
2010). Excessive inflammatory response can activate pyroptosis. ALT and AST levels. However, in Caspase-3-KO mice, the action of TRI
Numerous immune cells in the liver tissues are involved in pyroptosis, was antagonized to a certain extent and liver damage was ameliorated.
which initiates an immunological defense response, and the liver plays a Notably, TRI activated Caspase-3-GSDME in tissues and elevated the
significant role in preserving body metabolism and immune homeostasis levels of N-GSDME and Cleaved-Caspase-3, although producing insig­
(Aglietti et al., 2016; Rayamajhi et al., 2013). Pyroptosis exerts its role nificant effect on GSDMD-Casapase 1. These findings were consistent
primarily by relying on caspase protease, and activation of Caspases with the results of Kupffer cell experiments. When Caspase-3 inhibitor
1/4/5 can promote the interleukin maturation and activation (Yang was used, the action of TRI was also inhibited and the severity of liver
et al., 2015). Casapase proteins can also promote the cleavage of GSDM injury in mice was relieved.
proteins (Bibo-Verdugo et al., 2020; Naji et al., 2016), which form
N-GSDMs to play a role in opening the cell membrane pores (Teng et al., 5. Conclusion
2020). Kupffer cells, as a kind of liver-specific macrophages, function
crucially in liver injury. A study by Pelegrin found that LPS and ATP can Through network pharmacology and other means, we revealed the
induce pyroptosis of J774a.1 macrophages, activate P2×7 receptor, and mechanism and targets whereby TRI induced liver injury. The binding of
exert their effects after substantial IL-1β expression (Pelegrin and Sur­ TRI, a Caspase-3 activator, to VAL27 site promoted the maturation and
prenant, 2007). Rare earth oxides like Mirshafiee can promote the cleavage of Cleaved-Caspase-3, which further promoted the cleavage of
activation of NLRP3 bodies and Caspase 1 in Kupffer cells, thereby GSDME to form N-GSDME, ultimately resulting in Kupffer cell pyrop­
facilitating the occurrence of pyroptosis (Mirshafiee et al., 2018). Recent tosis. Caspase-3-GSDME is the major signal of TRI-induced liver injury.
research has shown that the Caspase-3 activation can induce The findings of this study provide a new reference for the prevention and
GSDME-dependent pyroptosis in addition to apoptosis. When cells treatment of TRI-induced liver injury, which also offer a new idea for the
highly express GSDME, activation of Caspase-3 can induce rational use of TRI in clinical settings.
GSDME-dependent pyroptosis, but when cells lowly express GSDME,
apoptosis is induced instead (Zheng et al., 2019). As a member of the CRediT authorship contribution statement
Gasdermin protein family, GSDME is composed of cytotoxic GSDME-N
and GSDME-C (Xixi, 2018). Upon stimulation by external pathogenic Chenyang Han, Hongyan Pei: Experimental operation and data
factors or death-inducing signals, cells promote cleavage of GSDME into acquisition. Yongjia Sheng, Jin Wang, Xiaohong Zhou: Experiment
GSDME-N fragments by activating Caspase-3, which then bind to design and data statistical analysis. Wenyan Li, Li Guo: Project fund
phospholipid molecules on the cell membranes to form GSDME pores, support. Yun Kong, Yi Yang: Article writing and revision.
thereby leading to the release of inflammatory IL-1β and IL-18 cytokines
until swelling and rupture of cells (Huang et al., 2020b). Declaration of Competing Interest
For many years, it has been a clinical consensus that long-term or
large-dose application of TRI, a major active constituent of Tripterygium The authors declare that they have no known competing financial
wilfordii, can induce liver injury. I further explored the role and mech­ interests or personal relationships that could have appeared to influence
anism of TRI in liver injury. Our results demonstrated that TRI could the work reported in this paper.
induce pyroptosis of Kupffer cells and promote the maturation and
release of inflammatory cytokines. According to TEM observations, TRI Data availability
did cause the opening of Kupffer cell membrane pores. However, note­
worthy was that TRI primarily promoted the expression of N-GSDME No data was used for the research described in the article.
and Cleaved-Caspase-3, while producing no effect on N-GSDMD or
Cleaved-Caspase 1. Suggestively, TRI was associated chiefly with the Acknowledgement
activation of nonclassical Caspase-3-GSDME signal. Network pharma­
cological analysis revealed that Caspase-3 had a strong targeting rela­ Thank you for the support of ZheJiang Provincial Natural Science
tionship with TRI. To further clarify the involvement of GSDMD and Foundation.
GSDME in TRI-induced liver injury, I separately knocked out GSDMD
and GSDME. The results showed that the knockout of GSDMD did not Data availability statement
affect the action of TRI, with Kupffer cells still being pyroptotic.
The data that support the findings of this study are available from the

9
C. Han et al. Ecotoxicology and Environmental Safety 258 (2023) 114963

corresponding author upon reasonable request. Huang, Z., Zhang, Q., Wang, Y., et al., 2020b. Inhibition of caspase-3-mediated GSDME-
derived pyroptosis aids in noncancerous tissue protection of squamous cell
carcinoma patients during cisplatin-based chemotherapy[J]. Am. J. Cancer Res. 10
Fund (12), 4287–4307.
Huo, J., Yu, Q., Zhang, Y., et al., 2019. Triptolide-induced hepatotoxicity via apoptosis
ZheJiang provincial Natural Science Foundation [LYY20H280005]. and autophagy in zebrafish[J]. J. Appl. Toxicol. 39 (11), 1532–1540.
Kepp, O., Galluzzi, L., Zitvogel, L., et al., 2010. Pyroptosis - a cell death modality of its
kind?[J]. Eur. J. Immunol. 40 (3), 627–630.
Ethical approval and consent to participate Liu, X., Hu, C., Li, H., et al., 2021. Metabolic profiling of fatty acids in Tripterygium
wilfordii multiglucoside- and triptolide-induced liver-injured rats[J]. Open. Life Sci.
16 (1), 184–197.
The study approvaled with Ethics Committee. Mei, Z., Li, X., Wu, Q., et al., 2005. The research on the anti-inflammatory activity and
hepatotoxicity of triptolide-loaded solid lipid nanoparticle[J]. Pharmacol. Res. 51
Consent for publication (4), 345–351.
Mirshafiee, V., Sun, B., Chang, C.H., et al., 2018. Toxicological profiling of metal oxide
nanoparticles in liver context reveals pyroptosis in Kupffer cells and macrophages
All authors approval published the article. versus apoptosis in hepatocytes[J]. Acs Nano acsnano.8b01086.
Naji, A., Muzembo, B.A., Yagyu, K.I., et al., 2016. Endocytosis of indium-tin-oxide
nanoparticles by macrophages provokes pyroptosis requiring NLRP3-ASC-Caspase1
Appendix A. Supporting information axis that can be prevented by mesenchymal stem cells[J]. Sci. Rep. 6, 26162.
Nan, Z., Aw, A., Tong, W.A., et al., 2021. Triptolide and atorvastatin synergistically
Supplementary data associated with this article can be found in the promote hepatotoxicity in cultured hepatocytes and female Sprague-Dawley rats by
inhibiting pregnane X receptor-mediated transcriptional activation of CYP3A4[J].
online version at doi:10.1016/j.ecoenv.2023.114963.
Toxicol. Lett. 342, 85–94.
Pelegrin, P., Surprenant, A., 2007. Pannexin-1 couples to maitotoxin- and nigericin-
References induced interleukin-1beta release through a dye uptake-independent pathway.[J].
J. Biol. Chem. 282 (4), 2386–2394.
Aglietti, R.A., Estevez, A., Gupta, A., et al., 2016. GsdmD p30 elicited by caspase-11 Rayamajhi, M., Yue, Z., Miao, E.A., 2013. Detection of pyroptosis by measuring released
during pyroptosis forms pores in membranes[J]. Proc. Natl. Acad. ences 113 (28), lactate dehydrogenase activity[J]. Methods Mol. Biol. 1040, 85–90.
7858–7863. Teng, J.F., Mei, Q.B., Zhou, X.G., et al., 2020. Polyphyllin VI Induces Caspase-1-Mediated
Bergsbaken, T., Cookson, B.T., 2007. Macrophage activation redirects yersinia-infected Pyroptosis via the Induction of ROS/NF-κB/NLRP3/GSDMD Signal Axis in Non-
host cell death from apoptosis to caspase-1-dependent pyroptosis[J]. PLoS Pathog. 3 Small Cell Lung Cancer[J]. Cancers 12 (1), 193–202.
(11), e161. Xixi, Zhang, 2018. Haibing, et al. Chemotherapy drugs induce pyroptosis through
Bergsbaken, T., Fink, S.L., Cookson, B.T., 2009. Pyroptosis: host cell death and caspase-3-dependent cleavage of GSDME[J]. Sci. China(Life Sci. ) v.61 (06),
inflammation. Nat. Rev. Microbiol. 7 (2), 99–109. 129–130.
Bibo-Verdugo, B., Snipas, S.J., Kolt, S., et al., 2020. Extended subsite profiling of the Yang, D., He, Y., Mu?Oz-Planillo, R., et al., 2015. Caspase-11 Requires the Pannexin-1
pyroptosis effector protein gasdermin D reveals a region recognized by inflammatory Channel and the Purinergic P2×7 Pore to Mediate Pyroptosis and Endotoxic Shock
caspase-11[J]. J. Biol. Chem. 295 (32) jbc.RA120.014259. [J]. Immunity 43 (5), 1–10.
Guan, H., Xie, L., Ji, Z., et al., 2021. Triptolide inhibits neutrophil extracellular trap Yuan, Z., Zhang, H., Hasnat, M., et al., 2021. A new perspective of triptolide-associated
formation.[J]. Ann. Transl. Med. 9 (17), 1384. hepatotoxicity: Liver hypersensitivity upon LPS stimulation (vol 414, pg 45, 2019)
Huang, C., Zeng, T., Li, J., et al., 2019. Folate Receptor-Mediated Renal-Targeting [J]. Toxicol.: Int. J. Concern. Eff. Chem. Living Syst. (463), 463.
Nanoplatform for the Specific Delivery of Triptolide to Treat Renal Ischemia/ Zheng, X., Zhong, T., Ma, Y., et al., 2019. Bnip3 mediates doxorubicin-induced
Reperfusion Injury[J]. ACS Biomater. Sci. Eng. 5 (6), 129–136. cardiomyocyte pyroptosis via caspase-3/GSDME[J]. Life Sci. 242, 117186.
Huang, J.F., Zhao, Q., Dai, M.Y., et al., 2020a. Gut microbiota protects from triptolide- Zhou, K., Chang, Y., Han, B., et al., 2021. MicroRNAs as crucial mediators in the
induced hepatotoxicity: key role of propionate and its downstream signalling events pharmacological activities of triptolide (Review).[J]. Experimental and Therapeutic.
[J]. Pharmacol. Res., 104752 Medicine 5, 115–123.

10

You might also like