<<

Chen et al. Molecular Neurodegeneration (2020) 15:26 https://doi.org/10.1186/s13024-020-00372-w

RESEARCH ARTICLE Open Access NLRP12 collaborates with NLRP3 and NLRC4 to promote inducing ganglion cell death of acute glaucoma Hui Chen1, Yang Deng1, Xiaoliang Gan1, Yonghao Li1, Wenyong Huang1, Lin Lu1, Lai Wei1, Lishi Su1, Jiawen Luo1, Bin Zou1, Yanhua Hong1, Yihai Cao2, Yizhi Liu1* and Wei Chi1*

Abstract Background: Acute glaucoma, characterized by a sudden elevation in intraocular pressure (IOP) and retinal ganglion cells (RGCs) death, is a major cause of irreversible blindness worldwide that lacks approved effective therapies, validated treatment targets and clear molecular mechanisms. We sought to explore the potential molecular mechanisms underlying the causal link between high IOP and glaucomatous RGCs death. Methods: A murine retinal ischemia/ reperfusion (RIR) model and an in vitro oxygen and glucose deprivation/ reoxygenation (OGDR) model were used to investigate the pathogenic mechanisms of acute glaucoma. Results: Our findings reveal a novel mechanism of microglia-induced pyroptosis-mediated RGCs death associated with glaucomatous vision loss. Genetic deletion of gasdermin D (GSDMD), the of pyroptosis, markedly ameliorated the RGCs death and retinal tissue damage in acute glaucoma. Moreover, GSDMD cleavage of microglial cells was dependent on -8 (CASP8)-hypoxia-inducible factor-1α (HIF-1α) signaling. Mechanistically, the newly identified nucleotide-binding leucine-rich repeat-containing receptor (NLR) family -containing 12 (NLRP12) collaborated with NLR family pyrin domain-containing 3 (NLRP3) and NLR family CARD domain-containing 4 (NLRC4) downstream of the CASP8-HIF-1α axis,toelicitpyroptoticprocesses and interleukin-1β (IL-1β) maturation through caspase-1 activation, facilitating pyroptosis and in acute glaucoma. Interestingly, processing of IL-1β in turn magnified the CASP8-HIF-1α-NLRP12/ NLRP3/NLRC4-pyroptosis circuit to accelerate inflammatory cascades. Conclusions: These data not only indicate that the collaborative effects of NLRP12, NLRP3 and NLRC4 on pyroptosis are responsible for RGCs death, but also shed novel mechanistic insights into microglial pyroptosis, paving novel therapeutic avenues for the treatment of glaucoma-induced irreversible vision loss through simultaneously targeting of pyroptosis. Keywords: Acute glaucoma, pyroptosis, NOD-like receptor 12, caspase-8/ HIF-1α

Background (IOP), severe eye pain, and irreversible vision loss that Acute glaucoma, which remains the most common can progress to permanent blindness [1–3]. The rapid glaucoma type among Asians, is predicted to affect 20 elevation in IOP leads to retinal ischemia/ reperfusion million people worldwide and is characterized by a sud- (RIR) injury and retinal ganglion cells (RGCs) death [4]. den and substantial increase in intraocular pressure The retina becomes vulnerable when disruption- induced ischemia/hypoxia in vessels results in shortages of oxygen and glucose, subsequently triggering a cas- * Correspondence: [email protected]; [email protected] 1State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, cadeofinflammatoryprocesses[5]. Similar to the cen- Sun Yat-sen University, Guangzhou 510060, China tral nervous system (CNS), microglial cells are Full list of author information is available at the end of the article

© The Author(s). 2020 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data. Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 2 of 16

phagocytic sentinels in the retina required for neuronal C57BL/6 mice following systemic homeostasis and innate immune defense, and their acti- (LPS) injection [35, 36]. Furthermore, we have also vation is responsible for neurodegenerative diseases [6]. reported that caspase-8 (CASP8)-induced NLRP3 pro- Recently, we [7], Vargas et al. [8] and many other scien- motes IL-1β processing during innate immune re- tists [9–11] have reported that high IOP-induced ische- sponses in an IOP-induced retinal ischemia model mia initiates microglial neuroinflammation that [7]. Since both CASP8-induced NLRP3 and HIF-1α regulate mediates retinal tissue damage and RGCs death. It is IL-1β, we therefore explored the potential functions and demonstrated that retinal ischemia not only directly in- underlying mechanisms of CASP8, NLRs, and HIF-1α with duces RGCs death but also triggers damage-associated respecttothedevelopmentofRIRinjury,suchasthatin molecular pattern (DAMP)-induced toll-like receptor 4 acute glaucoma. (TLR4) -dependent neuroinflammation In this study, we elucidated novel mechanisms for to activate the microglia, thus inducing further RGCs RGCs death and retinal tissue injury in acute glau- death [7, 12, 13]. coma. We found that genetically deleting GSDMD Traditionally, cell death has been divided into several substantially relieved RGCs death and the severity of distinct subtypes such as , and autoph- retinal ischemic injury, suggesting the crucial func- agy. Pyroptosis is a newly discovered form of pro- tions of pyroptosis in the development of acute glau- grammed lytic cell death that is induced by coma. Our findings further revealed that NLRP12 inflammatory casapse-1 (CASP1), and is characterized cooperated with NLRP3 and NLRC4 to promote pyr- by swelling of the cell, pore formation in unilamellar li- optosis through CASP1-dependent GSDMD cleavage, posomes, membrane rupture and the release of inflam- lactate dehydrogenase (LDH) release and mature IL- matory cytokines [14–18]. Gasdermin D (GSDMD) was 1β processing. Notably, we found that NLRP12-, discovered to be a substrate of CASP1, and cleavage of NLRP3- and NLRC4-induced CASP1 activation and the N-terminal fragment of GSDMD forms membrane pyroptosis were initiated by ischemia-triggered pores and drives pyroptosis [19–21]. Inflammasome CASP8-HIF-1α signaling activation. Strikingly, bio- complexes function as crucial intracellular effectors to active IL-1β served as a key molecule that in turn initiate innate immunity and thus to defend against in- positively accelerated the CASP8-HIF-1α-NLRP12/ fections or tissue damage [22, 23]. It has been demon- NLRP3/NLRC4 axis and promoted pyroptosis by strated that nucleotide-binding leucine-rich repeat cleaving GSDMD, indicating that IL-1β functioned as containing receptor (NLR) family pyrin domain- a pivotal node that magnified pyroptosis and neuroin- containing 3 (NLRP3) and NLR family CARD domain- flammation in the development of acute glaucoma. containing protein 4 (NLRC4) can both can form multi- protein complexes called , to activate Materials and methods CASP1, which subsequently cleaves the pore-forming Antibodies and reagents protein GSDMD [15, 24, 25]. Previous studies have Antibodies targeting the following were pur- shown that the recently discovered NLR family pyrin chased from ABclonal Biotech Co., Ltd. (USA): HIF- domain-containing 12 (NLRP12) plays an important role 1α (#A11945), HIF-1α (#A1544), NLRP3 (#A6345), in regulating the gut microbiota and autoinflammatory NLRP12 (#A6671), NLRC4 (#A7382), CASP1 disease [26, 27]. However, the exact functions of (#A0964), CASP8 (#A0215), IL-1β (#A1112) and NLRP12 in acute glaucoma are poorly studied and GSDMD (#A10164). Anti-cleaved-CASP8 (#8592S) largely unknown. Furthermore, the precise functions and and anti-phospho-NF-kB P65 (#3033S) antibodies complex molecular mechanisms that occur between the were obtained from Cell Signaling Technology. An IL- novel molecule NLRP12 and pyroptosis remain elusive. 1β neutralizing antibody (#14–7012-85) was obtained Scientific research so far has focused on - from eBioscience. The CASP1 inhibitor (CASP1 inh) induced pyroptosis [28, 29], prompting us to further in- Z-YVAD-FMK (YVAD, #ab141388) was obtained from vestigate the involvement and mechanisms of damage- Abcam. The NF-kB P65 inhibitor JSH-23 (#S7351) was induced pyroptosis and the potential roles and mecha- purchased from Selleckchem. Anti-β-actin (#AP0060) nisms of NLRP12 in pyroptosis in an acute glaucoma and secondary antibodies were purchased from Bio- model. world Technology. Lipofectamine 3000 was purchased Hypoxia activates hypoxia-inducible factor-1α (HIF- from Invitrogen. 1α) signaling, which activates a series of cellular pro- cesses including metabolic changes and angiogenesis Animal model of retinal ischemic injury − − to maintain cell viability [30–34]. HIF-1α drives inter- Adult female C57BL/6 mice and NLRP12 / mice and − − leukin (IL)-1β secretion to promote acute inflamma- GSDMD / mice were purchased from the Model Ani- tory responses in mouse primary cell cultures and mal Research Center at Nanjing University. The mice Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 3 of 16

were anesthetized with 100 mg/kg pentobarbital so- control (NC) small interfering RNA (siRNA)(20 μΜ/ dium by intraperitoneal injection and then topically 2 μL), CASP8 siRNA (20 μΜ/2 μL), NLRP3 siRNA (20 treated with 0.5% proparacaine (SomnoSuite; Kent μΜ/2 μL), NLRC4 siRNA (20 μΜ/2 μL), GSDMD siRNA Scientific, Torrington, CT, USA). The corneas were (20 μΜ/2 μL) or a CASP1 inh YVAD (200 μM/2 μL, subsequently treated with 1% tropicamide (Somno- Abcam, USA) into the vitreous cavity. The IOP returned Suite; Kent Scientific) to dilate the pupils. Cannula- to a normal level, and reperfusion was initiated. The tion of the right eye in the anterior chamber was mice were sacrificed 7 days after intravitreal siRNA achievedbyinsertinga30-gaugeneedle(BD,Franklin injection. Lakes, USA) attached to a normal saline reservoir to elevate and maintain an average IOP of 110 mmHg RNA sequence analysis for 90 min, which was monitored by our modified sys- RNA was extracted from retinal tissue samples of ’ tem according to Stockslager MA sstudy[37]. The C57BL/6 mice with and without RIR injury using a Mas- 30-gauge needle and attached normal saline reservoir terPure Complete DNA and RNA Purification Kit were connected to a pressure transducer (Honeywell (MC85200, Epicentre, Madison, WI, USA) according to 142PC01G) attached to a three-way stopcock. Once the manufacturer’s instructions. the needle was inserted into the anterior chamber, we Briefly, 100 ng of RNA was sonicated into 300- to were able to record IOP in real time. We adjusted 400-bp fragments using a Bioruptor (Diagenode, the height of the reservoir to maintain a stable IOP Belgium), and sequencing libraries were prepared using of 110 mmHg. The contralateral eye served as the a VAHTS Total RNA-seq (H/M/R) Library Prep Kit for sham group without an elevated IOP. After 90 min, Illumina® (NR603–02, Vazyme, Nanjing, China) follow- the needle was withdrawn, and the IOP returned to ing the manufacturer’s protocol. The RNA libraries normal levels. Tobramycin ointment was applied to were sequenced on an Illumina HiSeq2500 sequencer the treated eyes to prevent . Eyes with using a HiSeq SR Cluster Kit V4 (GD-401-4001, Illu- cannulation-induced cataracts, iris injury/ bleeding or mina, San Diego, CA, USA) and a HiSeq SBS Kit V4 50 anterior chamber leakage were excluded from this cycle kit (FC-401-4002, Illumina, San Diego, CA, USA). study. No randomization was used. Initial processing was performed with CASAVA (v1.8.2, Illumina, USA). Genotyping of knockout (KO) mice After quality control with FastQC (v0.11.5), we used Mouse tail tissue was collected, and genomic DNA was Cutadapt (v1.9.1) to trim low-quality bases. Trimmed isolated, mixed and incubated with a commercial kit reads were then aligned with TopHat (v2.0.11) to the DNeasy® Blood & Tissue Kit (Qiagen) according to the mouse genome mm9, and unique mapped reads were ’ manufacturer s instructions. A final centrifugation for 1 obtained though filtering of the NH tags of bam files. min at 6000×g was performed and the concentration of We used Cufflinks (v2.1.1) to calculate expression total DNA was measured by a NanoDrop ND-1000 values (as FPKM values) with the RefSeq mm9 reference Spectrophotometer (Thermo Scientific). The primers are transcriptome. Then, we summarized each sample’s ex- listed as below. pression values to detect significant differences in gene NLRP12 primers: transcript expression between groups. Forward: TGGCTTCTATTCAACTCCCT. Reverse: ATCGTTACACTCGGCTTCTC. GSDMD primers: Histological evaluation Forward: CGATGGAACGTAGTGCTGTG. After the mice were sacrificed, their eyes were enucle- Reverse: TCCTTCCCAACCTGCTGTTG. ated at the designated time points, fixed with 4% para- The DNA sequences were verified by PCR and formaldehyde (PFA) and paraffin embedded. Three sequencing. sections across the optic nerve of each eye were pre- μ After PCR amplification, the samples were electropho- pared at 5- m thickness and stained with hematoxylin resed in 2% agarose 1 × TAE (Tris [40 mM Tris], acetic and eosin staining (HE). The observational area was acid [20 mM] and ethylenediaminetetraacetic acid [1 from the internal to the outer limiting membrane mM]) gels, and stained with 1 × SYBR® Safe DNA Gel within a 1-mm distance from the optic nerve. Stain (Life Technologies). Images were collected by an Image Lab system (Bio-Rad). Immunofluorescence staining of the retina The mouse eyes were enucleated and embedded in optimal Treatment regimen cutting temperature (OCT) compound, frozen and sectioned After withdrawal of the needle from the anterior cham- to a thickness of 6 μm. Anti-RBPMS (1:400) (#ab152101, ber, each experimental eye was injected with negative Abcam, USA) was used to identify RGCs in the retina. DAPI Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 4 of 16

was used for nuclear counterstaining. Images were collected using Lipofectamine 3000 (Invitrogen, USA) accord- by Olympus immunofluorescence microscope. ing to the manufacturer’s protocol. Microglial cells were cultured to 70% confluence in 6-well plates and Fluoro-gold (FG) labeling of RGCs transfected with NC siRNA and siRNAs (20 μM) tar- Mice were intraperitoneally anesthetized with a mix- geting NLRP3, NLRP12, or NLRC4 and HIF-1α 12 h ture of 100 mg/kg ketamine and 10 mg/kg xylazine. prior to oxygen and glucose deprivation/reoxygena- Ointment was applied to both eyes to prevent the tion (OGDR) treatment. The cells were harvested 24 corneas from drying. Next, to label RGCs, 4% fluoro- h after reperfusion, and cell lysates were obtained gold (FG) was injected into both sides of the superior for protein assays. colliculi 7 days prior to the establishment of RIR. The mice were sacrificed at the designated time points Culture of murine primary microglia after reperfusion, and their eyes were fixed with 4% The cerebral cortices without meninges and blood ves- PFA on ice for 30 min before retinal flat mounting. sels were separated from P0 – P2 newborn C57BL/6 J FG-positive cells were subsequently identified by two mice, followed by digestion with 0.125% trypsin for 15 independent observers using cellSens Dimension soft- min. Cells were isolated and plated on 25 cm2 flasks ware (Olympus). coated with poly – L – lysine (5 μg/mL). Microglial cells were dissociated from the mixed cul- Preparation of siRNAs tures and collected on the 11th or 12th day by gently siRNAs targeting mouse NLRP3, NLRP12, NLRC4, HIF- shaking and centrifugated. Purification was confirmed by 1α, CASP8, GSDMD were designed by Ribobio Co., Ltd., the microglial marker IBA-1. using the mRNA sequences for these in the NCBI database as follows: mNLRP3 (GTACTTAAATCGTGAA OGDR treatment ACA), mNLRC4 (GCTGGGAGTTTGATGACTA), To mimic the murine model of RIR, BV2 microglial cells mNLRP12 (CCAAATGGAGACCCTCTTT), m HIF-1α were subjected to oxygen and glucose deprivation for 3 h (GGTATGTGGCATTTATTTG), mCASP8 (GTCATG and subsequently returned to their normal environment CTCTATCAGATTT) and mGSDMD (CCATGGCCTC (37 °C, normoxic) and normal medium (DMEM supple- AATGTGCTT). The NC siRNA was purchased from mented with 10% FBS and glucose [4.9 g/L]) for 24 h Ribobio Co., Ltd. as well. prior to harvest. The cells were transferred into serum- and glucose-free medium and placed in a chamber Generation of CASP8 KO cell lines using the CRISPR/Cas9 (ProOx 110, Biospherix, USA) under hypoxic conditions system (5% CO2 and 95% N2) in a 37 °C incubator for 3 h. Re- Stable KO of CASP8 in BV2 microglial cells was estab- perfusion was performed by exposing the cells to nor- lished by BIOCYTOGEN. The sgRNAs targeting exon 3 moxic culture conditions and culturing them in DMEM to exon 5 of the wide-type (WT) allele were designed supplemented with 10% FBS and glucose (4.9 g/L) in a using an online tool to identify the guide sequences. The 37 °C incubator for 24 h. selected sequences were individually inserted into the CRISPR vectors, and the sequences for the sgRNAs used Immunocytochemistry staining in the present study will be provided upon request. Cells WT and CASP8 KO BV2 microglial cells were sub- were transfected with the CRISPR vectors and selected jected to oxygen and glucose deprivation for 3 h for 72 h in medium containing puromycin (0.75 g/mL). followed by 24 h of OGDR. After OGDR, the cells were Subsequently single clones were selected through serial fixed with 4% PFA for 30 min and washed several times dilution. The clones were verified by PCR and western with PBS. The primary antibodies targeted CASP8 (1: blot analysis. 200), HIF-1α (1:50) and p-P65 (1:50). Cell nuclei were stained with DAPI. Fluorescence images were acquired Cell culture and siRNA transfection by two independent observers with an Olympus fluor- The BV2 microglial cell line was purchased from escence microscope. Procell Co., Ltd. (#CL-0493, Wuhan, China) and has been authenticated by Microread Genetics Co., Ltd. Western blot analysis (Beijing, China) using PCR amplification. No myco- Total protein was isolated from either microglial cells or plasma contamination was found in this cell line. retinal tissue and processed for western blotting. Equal The cells were cultured in Dulbecco’smodifiedEa- amounts of protein (50 μg for cell culture and 100 μg for gle’s medium (DMEM) supplemented with 10% fetal retinal tissue) were loaded onto SDS-PAGE gels and bovine serum (FBS) and maintained at 37 °C in an transferred to polyvinylidene difluoride (PVDF) mem- incubator at 5% CO2. The cells were transfected branes. The membranes were blocked for 1 h with 5% Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 5 of 16

nonfat dry milk diluted in TBST and subsequently incu- examined with a scanning electron microscope (#FEI- bated at 4 °C overnight with primary antibodies targeting Quanta200; FEI, Co., Ltd., Czech Republic). the following proteins: HIF-1α (1:200), NLRP3 (1:500), NLRP12 (1:200), NLRC4 (1:500), CASP1 (1:200), CASP8 Statistics (1:200), IL-1β (1:200), GSDMD (1:500) and β-actin (1: The data are shown as the mean ± SD. One-way 1000). The following day, the blots were incubated with ANOVA followed by Bonferroni multiple comparison the appropriate secondary antibody (1:5000) for 1 h at tests was used to compare three or more groups, and room temperature, after which the blots were visualized two-tailed unpaired t-test was used to assess two groups with an enhanced chemiluminescence (ECL) kit, using GraphPad Prism software (version 6.0, GraphPad eBioscience) and recorded with an Image Lab imaging Software, Inc., San Diego, CA, USA). P values less than system (Bio-Rad, USA). 0.05 were considered to indicate statistical significance. The variance was similar among groups that were statis- Quantitative real-time PCR (RT-qPCR) tically compared. Total RNA was extracted from the WT BV2 and CASP8 KO BV2 microglial cells and mouse retinal tis- Results sues using TRIzol reagent (Invitrogen), and cDNA was Pyroptosis plays a key role in elevated IOP-induced synthesized with a PrimeScript RT Master Mix retinal ischemic injury and glaucomatous RGCs death (TaKaRa). Quantitative amplification of murine HIF-1α, Pyroptosis is a novel inflammatory form of cell death NLRP3, NLRP12, NLRC4 and β-actin was performed triggered by CASP1/CASP11 and is characterized by using a Light Cycler 480 Real-Time PCR System with membrane pore formation via the N-terminal fragment software version LCS480 1.5.1.62. The mRNA levels of of GSDMD along with leakage the target genes were normalized to those of β-actin. [19, 38]. Nevertheless, whether and how pyroptosis plays The primers of the target genes were as follows: mβ- its roles in acute glaucoma are still unclear. We there- actin (forward: GGCTGTATTCCCCTCCATCG; re- fore investigated the exact functions of pyroptosis using verse: CCAGTTGGTAACAATGCCATGT), mHIF-1α genetic deletion approaches or pharmacological inhibi- (forward: GGCTCCCTTTTTCAAGCAGC; reverse: tosr targeting pyroptosis-associated components in ele- TGCTCCGTTCCATTCTGTTCA), mNLRP3 (forward: vated IOP-induced RIR. Loss-of-function experiments TTCTGCACCCGGACTGTAAA; reverse: TCGCCA showed that genetic elimination of the pyroptosis- AGATCATTGTTGCC), mNLRP12 (forward: GTTACA effector GSDMD markedly increased total retinal thick- CTCGGCTTCTCCT; reverse: GTTCTTCGTCTGGC ness (136.7 ± 5 μm vs 109.6 ± 5.65 μm) and attenuated TCAA) and mNLRC4 (forward: ACCTGGAAAAGGAT RGCs death after being subjected to RIR injury (Fig. 1a GGGAATGAA; reverse: AAGTTTGGCAAGTC to c). As GSDMD was determined to participate in the CTGGGG). pathogenesis of RIR, we therefore explored its upstream regulator. YVAD, the specific inhibitor targeting CASP1 Cytotoxicity assay, IL-1β ELISA and CASP8 activity assays (referred as CASP1 inh throughout the manuscript), sig- Cellular cytotoxicity was measured with an LDH re- nificantly blocked the cleavage of IL-1β and GSDMD, in- lease kit (#C0016, Beyotime, China) according to the dicating that RIR initiated pyroptosis via the canonical manufacturer’s instructions. Cells were harvested at CASP1 pathway (Fig. 1 d). Furthermore, selective knock- 24 h post-OGDR treatment, washed with cold PBS down of CASP1 through intravitreal injection markedly and centrifuged at low speed for 20 min. The super- attenuated the severity of retinal damage and restored natants were transferred to fresh tubes to measure RGCs numbers during RIR injury (Fig. 1e to g). These the production of mature IL-1β using an IL-1β ELISA findings validated the pivotal roles of pyroptosis in ag- kit (Elabscience, USA). To assess CASP8 activity, the gravating retinal ischemic injury and RGCs death. protein concentrations in BV2 cell lysates were mea- Microglia are the most important type of cells in mediat- sured with a BCA kit, and equal amounts of protein ing immune responses in retinal tissue [6]. In this study, were mixed with the reaction reagent and added to we established an in vitro model using BV2 microglia 96-well plates for measurement with the CASP8 activ- subjected to OGDR to mimic RIR injury. The occur- ity kit (BioVision, USA). rence of pyroptotic cell death in BV2 microglia subjected to ischemic injury was detected by SEM and was charac- Scanning electron microscopy (SEM) terized by cell blebbing, cell swelling and cell flattening. WT and CASP8 KO BV2 microglial cells were fixed in Collectively, these findings reveal that CASP1-mediated 2.5% glutaraldehyde in phosphate buffer at 4 °C over- pyroptosis participates in RIR pathogenesis and is associ- night. After the cells were dehydrated through critical ated with RGCs death and retinal ischemic damage in point drying, they were coated with gold-palladium and acute glaucoma. Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 6 of 16

Fig. 1. CASP1-dependent pyroptosis plays a vital role in elevated IOP-induced retinal ischemic damage and RGCs loss. a HE staining and quantitative analysis of total retinal thickness in retina tissue harvested 7 days post RIR injury (n = 6). Scale bar: 50 μm. b Retrograde FG-labeled images and quantitative assay of RGCs survival in WT and GSDMD KO mice (n = 6). Scale bar: 200 μm. c Representative immunofluorescence images of RGCs in the retina. Anti-RBPMS was used to label RGCs (n = 6). Scale bar: 100 μm. d Western blot analysis of the indicated proteins (n = 6). The protein levels were normalized to β-actin levels. e Representative immunofluorescence images of RGCs in the retina from mice treated with or without CASP1 inhibitor. Anti-RBPMS was used to label RGCs (n = 6). Scale bar: 100 μm. f HE staining and quantitative analysis of total retinal thickness in retinal tissue (n = 6). Scale bar: 50 μm. g Retrograde FG-stained images and quantitative assay of RGCs amount from groups subjected to RIR and RIR concomitant with CASP1 suppression groups (200 μΜ, n = 6). Scale bar: 200 μm. WT: wide type; KO: knockout; RIR: retinal ischemia-reperfusion; GCL: ganglion cell layer; IPL: inner plexiform layer; INL: inner nuclear layer; OPL: outer plexiform layer; ONL: outer nuclear layer; CASP1 inh: caspase-1 inhibitor. All the experiments are representative of at least three independent experiments. Data are represented as the mean ± SD. *P < 0.05, **P < 0.01, one-way ANOVA and two-way ANOVA

NLRP12, NLRP3, and NLRC4 are responsible for RIR injury- NLRP12 collaborates with NLRP3 and NLRC4 to induce induced RGCs death pyroptosis by CASP1-dependent GSDMD cleavage Our previous study has demonstrated that NLRP3 level The activation of NLRP3 recruits pro-CASP1 into inflamma- is elevated in retinal tissue damage following retinal is- somes and cleaves it into its active form, which subsequently chemia [7]. However, the exact roles of novel NLRs, induces GSDMD cleavage to drive pyroptotic cell death as a NLRP12 and NLRC4 in acute glaucoma are still unre- pathogen defense mechanism [39, 40]. However, the effects solved. In the current study, we found that high IOP- of other NLR inflammasomes onpyroptosisinresponseto induced retinal ischemia promoted the upregulation of danger-associated signals have not been reported. Conse- NLRP12 and NLRC4. We established an RIR model in quently, we investigated the effects and regulatory mecha- NLRP12-deficient mice and found significant improve- nisms of NLRs on pyroptosis in vitro and in vivo. First of all, ments in total retinal thickness and RGCs survival in we established OGDR model using murine primary micro- NLRP12 KO mice compared with WT mice (Fig. 2 ato glial cells (Supplementary Fig. 2a) and found that the levels c and supplementary Fig. 1a). Furthermore, inhibition of of NLRP3/ NLRP12/ NLRC4 and the effector of pyroptosis, NLRP3 or NLRC4 also markedly reduced the severity of termed GSDMD, were significantly increased in OGDR retinal ischemic damage and improved RGCs survival group than that in the controls (Supplementary Fig. 2b-e). In (Fig. 2 d to e, and supplementary Fig. 1b). All these find- an In vitro model using BV2 microglia subjected to OGDR, ings confirm that genetic deletion of NLRP12 or block- knocking down NLRP3, NLRP12, or NLRC4 suppressed ade of NLRP3 and NLRC4 protects retinal structure and neuroinflammation and pyroptosis through such effects as RGCs from RIR-induced injury. the downregulation of cleaved-CASP1, reductions in Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 7 of 16

Fig. 2 Genetic deletion of NLRP12 and inhibition of NLRP3 or NLRC4 significantly attenuate retinal damage and improve RGC survival. a Western blot detection of NLRP12 in retinas from NLRP12-deficient mice (n = 6). b HE staining and quantitative measurement of total retinal thickness targeting retinal tissue morphology in WT and NLRP12 KO mice under high IOP (n = 6). Scale bar: 50 μm. c Retrograde FG labeling and RGC amount evaluation in WT and NLRP12 KO mice under high IOP (n = 6). Scale bar: 200 μm. d HE staining and quantitative evaluation of total retinal thickness in retinal tissue subjected to high IOP followed by NLRP3/NLRC4 knockdown (20 μΜ, n = 6). Scale bar: 50 μm. e Retrograde FG labeling and quantitative measurement of RGC survival in retinal tissue subjected to elevated IOP followed by NLRP3/NLRC4 knockdown (20 μΜ, n = 6). Scale bar: 200 μm. WT: wide type; KO: knockout; RIR: retinal ischemia-reperfusion; GCL: ganglion cell layer; IPL: inner plexiform layer; INL: inner nuclear layer; OPL: outer plexiform layer; ONL: outer nuclear layer. All of the data are representative of at least three independent experiments. The data are represented as the mean ± SD. *P < 0.05, **P < 0.01, one-way ANOVA and two-way ANOVA. cleaved-GSDMD and mature IL-1β levels, and reductions in CASP8 is involved in acute glaucoma LDH leakage (Fig. 3 a to c). Similarly, NLRP12 KO and To gain better insight into the pathological molecular knockdown of NLRP3/NLRC4 reduced N-GSDMD and changes initiated by high IOP-induced RIR injury, we per- cleaved-CASP1 levels in the mouse model (Fig. 3gandh). formed RNA sequence analysis and Kyoto Encyclopedia of Furthermore, inhibiting CASP1 significantly reduced Genes and Genomes (KEGG) pathway enrichment analysis pyroptosis-associated LDH release (Fig. 3d) and IL-1β secre- on retinal tissue from untreated and RIR mice. Notably, 385 tion (Fig. 3e) in vitro, as well as N-GSDMD cleavage and IL- genes were upregulated and 595 genes were downregulated 1β production (Fig. 1e) in vivo, suggesting that multiple in the RIR group as compared with the sham group (Fig. 4a). NLRs drove pyroptotic death via CASP1 cleavage. KEGG analyses showed that multiple pathways in re- Here, we discovered the first evidence of reciprocal regula- sponse to inflammatory and hypoxia as well as signaling tion among NLRP3, NLRP12, and NLRC4. Suppression of transduction pathways such as the NF-kappa B and NLRP12 decreased the production of NLRP3 and NLRC4, MAPK signaling pathways were clearly activated during while the knockdown of either NLRP3 or NLRC4 also re- reperfusion (Fig. 4b). As the NF-kB pathway has been re- duced the production of the other two NLRs (Fig. 3iandj), ported to positively involved in neuroinflammation [41, indicating that NLRP12 acted synergistically with NLRP3 42], we investigated the positive regulation of I-kappaB and NLRC4. The elevated IOP-induced retinal ischemic kinase/NF-kappaB signaling in this murine ischemia- murine model also confirmed the in vitro data (Fig. 3 kton). reperfusion model. Among the factors involved in this in- Taken together, these results demonstrated that dicated pathway, CASP8 was markedly upregulated at the NLRP12 collaborates with NLRP3 and NLRC4 to induce mRNA level in the retinas of mice subjected to RIR injury pyroptosis through CASP1-dependent GSDMD cleavage (Fig. 4c), suggesting that CASP8 actively participates in during ischemic injury. the RIR pathogenesis. Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 8 of 16

Fig. 3. NLRP12 collaborates with NLRP3 and NLRC4 to elicit pyroptosis and promote IL-1β production in ischemic injury through CASP1- dependent GSDMD cleavage a-h NLRP12/NLRP3/NLRC4 promoted IL-1β release and induced pyroptosis by CASP1-dependent GSDMD cleavage: a Knockdown of NLRP12/NLRP3/NLRC4 reduced CASP1 and GSDMD cleavage in extracts from BV2 microglia after OGDR injury, as determined by western blot analysis (n = 6). The protein levels were normalized to β-actin levels. b LDH release (n = 6). c IL-1β secretion (n = 6). d-e LDH release (d) and IL-1β production (e) of BV2 microglia treated with the CASP1 inh, YVAD (200 µM) and subjected to OGDR injury (n = 6). f Representative SEM images showed pyroptotic cell death and other morphological changes in BV2 microglia subjected to OGDR combined with or without different additional treatments (n = 5): (i): control; (ii): OGDR; (iii): OGDR plus CASP1 inh (YVAD, 200 µM); (iv): OGDR plus NLRP3 siRNA (si); (v): OGDR plus NLRP12 si; (vi): OGDR plus NLRC4 si; (vii): CASP8 CRISPR plus OGDR; (viii): OGDR plus HIF-1α si ; (ix): OGDR plus IL-1β neutralizing antibody. Scale bar: 20 µm. g-h Immunoblotting analysis for detection of pyroptotic proteins in the retinas of NLRP12-/- or WT mice with or without NLRP3/NLRC4 knockdown under RIR conditions (n = 6). The protein levels were normalized to β-actin levels. i-n Mutual regulatory relationships among NLRP12, NLRP3 and NLRC4: i-j Protein expression and mRNA levels of the indicated molecules, as determined by western blotting and qRT-PCR detection in BV2 microglia exposed to OGDR with or without NLRP3/NLRP12/NLRC4 knockdown (n = 6). The mRNA and protein levels were normalized to β-actin levels. k-n Western blotting and qRT-PCR analyses of NLRP3, NLRP12, and NLRC4 in retinas from WT mice and NLRP12 KO mice that were sacrificed 7 days after elevated IOP injury, combined with or without NLRP3/NLRC4 knockdown (n = 6). The mRNA and protein levels were normalized to β-actin levels. The data shown are representative of at least three independent experiments. The data are represented as the mean ± SD. *P < 0.05, **P < 0.01, one-way ANOVA, two-way ANOVA and two-tailed unpaired t-test.

CASP8 is considered crucial for embryonic development, pathogenesis of retinal hypoxia/ischemia injury [45–48], − − and CASP8 / mice are found abnormal or close to death which has been further confirmed in our study. Intravi- at 10.5 to 12.5 at embryonic days (E10.5 to E12.5) [43, 44]. treally knockdown of HIF-1α significantly alleviated retinal We explored the underlying functions of CASP8 by intravi- damageandRGCsloss(Fig.5 atoc).Thesefindings treally injecting CASP8 siRNA into elevated IOP-induced prompted us to explore whether there is a regulatory rela- RIR model animals. Our findings revealed that knocking tionship between CASP8 and HIF-1α. Therefore, we estab- down CASP8 significantly attenuated retinal damage and lished a CASP8 KO BV2 cell line using the CRISPR-CAS9 reduced RGCs loss (Fig. 4d to f), indicating the pivotal role system (Fig. 5d) to determine the role of CASP8 under ofCASP8inmediatingretinalischemicinjury. OGDR conditions. Our data showed that CASP8 KO signifi- cantly blocked HIF-1α expression under OGDR (Fig. 5eto g), whereas blockade of HIF-1α had no effect on CASP8 ac- Inhibition of CASP8-mediated HIF-1α signaling protects tivity (Fig. 5h), indicating that CASP8 was upstream of HIF- the retina and RGCs from ischemic injury 1α signaling. We next explored the molecular regulatory Research has increasingly shown that HIF-1α plays a mechanisms by which CASP8 controlled HIF-1α.NF-kB critical role in regulating during the Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 9 of 16

Fig. 4 CASP8 is a pivotal participant in RIR injury. a Volcano plot showing a total of 980 genes, of which 385 were upregulated and 595 were downregulated in retinal tissue in the RIR group compared with the sham group. A fold change > 2 or < 0.5 was considered statistically significant (n = 5 mice/group). b KEGG pathway enrichment analysis of differentially expressed genes between the ischemia/reperfusion and sham groups (n = 5 mice/group). The enriched pathways of hypoxia and inflammatory signaling are shown. The positive regulation of I-kappaB kinase/ NF-kappaB signaling was chosen for further analysis. c Heat map showing the contents of the positive regulation of I-kappaB kinase/NF-kappaB signaling in the retinas of mice after the RIR process (n = 5 mice/group). RIR mice were sacrificed 7 days after reperfusion, and retinal tissue was collected for RNA sequence analysis. d HE staining of retinas from mice under RIR with or without CASP8 knockdown (20 μΜ). The retinal tissue was harvested on the seventh day after reperfusion (n = 6). Scale bar: 50 μm. The total retinal thicknesses were quantified for the three groups (n = 6). e Representative immunofluorescence images of RGCs in the retina from mice treated with or without CASP8 knockdown. Primary antibody against RBPMS was used to label RGCs (n = 6). Scale bar: 100 μm. f Retrograde FG staining of RGCs from RIR injury in the absence or presence of CASP8 interference (20 μΜ, n = 6). Scale bar: 200 μm. RGCs survival were analyzed comparing to the controls (n = 6). RIR: retinal ischemia-reperfusion; GCL: ganglion cell layer; IPL: inner plexiform layer; INL: inner nuclear layer; OPL: outer plexiform layer; ONL: outer nuclear layer; CASP8 si: caspase-8 siRNA. All of the data are representative of at least three independent experiments. Data are represented as the mean ± SD. *P < 0.05, **P < 0.01, one-way ANOVA signaling plays a critical role in the pathogenesis of inflam- discovered NLRs remain unknown. Therefore, we explored mation and our results showed that deletion of CASP8 ef- the influence of CASP8 on the newly-discovered NLRC4 and fectively blocked NF-kB translocation (Fig. 5i), while NLRP12 during RIR (Fig. 6a and b). The production of inhibition of NF-κB transcriptional activity significantly NLRP3, NLRC4, and NLRP12 was all significantly lower in downregulated HIF-1α expression (Fig. 5j). Taken together, WT mice treated with CASP8 siRNA than in control mice, these findings revealed that HIF-1α signaling is induced by indicating that CASP8 exerted its inflammatory effect on ret- CASP8 in this OGDR model via NF-kB translocation. inal innate immunity by activating not only the classic NLRP3 but also the novel NLRP12 and NLRC4 in retinal ischemia NLRP12/NLRP3/NLRC4 and CASP1 activation is triggered (Fig. 6a and b). Knocking out CASP8 also likewise suppressed by the CASP8-HIF-1α pathway NLRP12/NLRP3/NLRC4 upregulation and CASP1 cleavage We previously validated that CASP8 mediates retinal neuroin- in microglia subjected to OGDR (Fig. 6candd),indicating flammation by promoting the activation of NLRP1 and that CASP8 activation was at least partially responsible for the NLRP3 [7], whereas the effects of CASP8 on the newly activation of NLRP3/NLRP12/NLRC4 and CASP1. Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 10 of 16

Fig. 5 CASP8-mediated HIF-1α signaling is involved in retinal ischemic injury and RGCs loss. a HE staining and quantitative evaluation of total retinal thickness in retinal tissue subjected to high IOP followed by HIF-1α knockdown (20 μΜ, n = 6). Scale bar: 50 μm. b Retrograde FG labeling and quantitative measurement of RGCs survival from mice subjected to RIR injury in the absence or presence of HIF-1α interference (20 μΜ,n= 6). Scale bar: 200 μm. c Representative immunofluorescence images of RGCs in the retina from mice treated with or without HIF-1α blockage. Primary antibody against RBPMS was used to label RGCs (n = 6). Scale bar: 100 μm. d CRISPR-CAS9 design to knock out CASP8 in BV2 cell line. Targeted vector was designed based on the exon 3 to exon 5 in WT allele. e-f RNA level and protein levels of HIF-1α in WT and CASP8 KO BV2 cell line exposed to OGDR insult (n = 6, both). The protein and mRNA levels were normalized to β-actin levels. g BV2 microglia with the indicated genotypes were subjected to OGDR and stained with antibodies against cleaved-CASP8 and HIF-1α (n = 6). Scale bar: 100 μm. h CASP8 activity (n = 5). i Representative images of immunofluorescence staining targeting phospho-NF-kB P65 translocation in WT BV2 microglia and CASP8- specific KO cell line exposed to OGDR (n = 6). Scale bar: 20 μm. j The protein levels of HIF-1α were assayed by immunoblots in BV2 microglia treated with the NF-kB P65 inhibitor, JSH-23 (40 μM, n = 6). The protein expression was normalized to β-actin expression. RIR: retinal ischemia- reperfusion; GCL: ganglion cell layer; IPL: inner plexiform layer; INL: inner nuclear layer; OPL: outer plexiform layer; ONL: outer nuclear layer; HIF-1α si: HIF-1α siRNA. All of the data are representative of at least three independent experiments. Data are represented as the mean ± SD. *P < 0.05, **P < 0.01, one-way ANOVA and two-tailed unpaired t-test.

Although CASP8 regulated HIF-1α signaling and NLRP3/ studies have revealed the nonapoptotic roles of NLRP12/NLRC4 activation, the details of the mutual regula- CASP8 as initiating cleavage of GSDMD during tion between HIF-1α and NLRs remains obscure. Accord- Yersinia infection [50, 51]. Nevertheless, the poten- ingly, we investigated the precise relationships tial link between CASP8 and pyroptosis in DAMP- between HIF-1α and the NLRP3/NLRP12/NLRC4 induced RIR injury is still unclear. Our current inflammasomes. Our findings revealed that selective study verified that CASP8 inhibition led to a knockdown of HIF-1α was associated with reduced marked reduction in the cleavage of GSDMD (Fig. activation of NLRP3/NLRP12/NLRC4 and CASP1 ac- 7a), implying that CASP8 induced GSDMD- tivation in vivo and in vitro (Fig. 6etoh).In dependent pyroptosis during RIR pathogenesis. addition, knockdown of NLRP12, NLRP3 or NLRC4 In vitro, deletion of CASP8 also markedly suppressed or suppression of CASP1 in turn reduced the expres- CASP1 and GSDMD cleavage, cellular cytotoxicity, IL- sion of HIF-1α (Fig. 6 i to l), suggesting that there 1β release and pyroptotic cell death (Fig. 7 b, c, d and was a positive feedback loop among NLRs, CASP1 Fig. 3f). These data indicated that CASP8 not only par- and HIF-1α. Altogether, these results indicated that ticipated in neuroinflammation via IL-1β secretion, but NLRP12/NLRP3/NLRC4 and CASP1 activation is in- also mediated CASP1-induced pyroptosis under OGDR. duced by CASP8-modulated HIF-1α signaling. Meanwhile, we are the first to report HIF-1α blockage reduced cleavage of GSDMD and IL-1β production as Activation of the CASP8-HIF-1α pathway elicits pyroptosis well as attenuated LDH leakage and pyroptotic cell death via CASP1-dependent GSDMD cleavage in vivo and in vitro (Fig. 7 e, f, g, h and Fig. 3f). Taken Although CASP8 has traditionally been considered together, these results confirmed that the CASP8-HIF- as a pivotal apoptotic initiator [49], accumulating 1α pathway is critical to IL-1β processing and initiation Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 11 of 16

Fig. 6 CASP8 promotes NLRP12/NLRP3/NLRC4 and CASP1 activation upon HIF-1α signaling. a-b The protein and mRNA levels of NLRP12/NLRP3/ NLRC4 and CASP1 was detected in retinas from WT mice with or without CASP8 knockdown (20 μΜ) that were harvested at the seventh day after reperfusion (n = 6). The protein and mRNA levels were normalized to β-actin levels. c-d CASP8 elimination diminished the activation of NLRP12/NLRP3/NLRC4 and CASP1 in BV2 microglia exposed to OGDR (n = 6). The protein and mRNA levels were normalized to β-actin levels. e-h Immunoblot and qRT-PCR analyses of targeting NLRP12/NLRP3/NLRC4 and CASP1 in vivo and in vitro (n = 6) with or without HIF-1α knockdown. The protein and mRNA levels were normalized to β-actin levels. i-j Protein and mRNA levels of HIF-1α upon NLRP12/NLRP3/NLRC4 suppression in vitro (n = 6). The mRNA and protein levels were normalized to β-actin levels. k-l Knockdown of CASP1 suppressed the production of HIF-1α protein and mRNA in vitro (n = 6). The data shown are representative of at least three independent experiments. The data are represented as the mean ± SD. *P < 0.05, **P < 0.01, one-way ANOVA of pyroptosis via NLR-activated CASP1 signaling in re- IOP to an appropriate level is regarded as the principle sponse to ischemic injury. strategy for the treatment of acute glaucoma [53]. Unfor- tunately, there is a lack of approved, effective and vali- Mature IL-1β is the key factor activating NLRP12/NLRP3/ dated treatment targets for acute glaucoma. Therefore, NLRC4-induced pyroptosis through CASP8-HIF-1α prevention and attenuation of acute glaucoma damage pathway are unmet clinical needs. Previously, neuroinflammation HIF-1α production, NLRs expression and CASP8 activity in response to ischemia has been shown to play a pivotal were dramatically reduced when dissociative IL-1β was role in the pathogenesis of elevated IOP-induced retinal neutralized (Fig. 7i and j), indicating that IL-1β could in ischemic injury and RGCs death [7, 8, 54]. A deeper un- turn regulated the CASP8-HIF-1α-NLRs axis. It implied derstanding of the potential neuroinflammation mecha- that CASP8-HIF-1α-NLRs axis-induced IL-1β matur- nisms is urgently needed to support identification of key ation is a positive feedback loop to trigger inflammatory targets for acute glaucoma treatment. cascades. Furthermore, neutralizing IL-1β reduced the In the current study, we demonstrated that pyroptosis release of N-terminal GSDMD to protect against pyrop- plays crucial roles in promoting RGCs death and retinal totic death (Fig. 7i and Fig. 3f). Overall, these results re- ischemic injury in acute glaucoma. Remarkably, our veal a novel pathway whereby the CASP8-HIF-1α-NLRs- findings reveal novel functions of NLRP12, which is in- IL-1β axis mediates pyroptosis and neuroinflammation duced by CASP8 and HIF-1α activation. Furthermore, during the pathogenesis of retinal ischemic injury (Fig. we found that NLRP12 collaborates with NLRP3 and 8). Additionally, these data further extend the functions NLRC4 to initiate pyroptosis via CASP1-dependent of IL-1β in regulating CASP8, HIF-1α and NLRP3/ GSDMD cleavage in vivo and in vitro. We also discov- NLRP12/NLRC4 axis and amplifying pyroptosis. ered a crucial regulatory association between CASP8 and HIF-1α in which CASP8 modulates HIF-1α signal- Discussion ing by driving NF-kB translocation. Furthermore, we Acute glaucoma is one of the most common causes of found that IL-1β magnifies pyroptosis via promoting sharp vision loss worldwide [2]. Elevated IOP induces CASP8-HIF-1α-induced NLRP12/NLRP3/NLRC4 activa- retinal ischemia and RGCs loss, leading to irreversible tion, indicating its role in a key positive feedback loop visual impairment [52]. Medication aimed at lowering that accelerates neuroinflammation and pyroptosis. Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 12 of 16

Fig. 7 Activation of the CASP8-HIF-1α pathway elicits pyroptosis and promotes IL-1β production which in turn magnifies inflammatory cascades via the CASP8-HIF-1α-NLR axis. a The protein levels of cleaved GSDMD were detected in retinas from WT mice with or without CASP8 knockdown (20 μΜ) that were harvested at the seventh day after reperfusion (n = 6). The protein levels were normalized to β-actin levels. b Western blot analysis of cleaved CASP1 and GSDMD in extracts from WT BV2 microglia and CASP8-specific KO cell line after OGDR injury (n = 6). The protein levels were normalized to β-actin levels. c-d Cytotoxicity c and IL-1β production d in BV2 microglia under OGDR injury (n = 6). e-f The protein levels of cleaved GSDMD were detected in vivo and in vitro with or without HIF-1α knockdown. The protein levels were normalized to β-actin levels. g-h Cytotoxicity g and IL-1β processing h were measured in the presence or absence of HIF-1α siRNA treatment in BV2 microglia (n = 6, both). i Western blotting detection of the indicated proteins in BV2 microglia subjected to OGDR and OGDR concomitant with IL-1β neutralizing antibody treatment (n = 6). The protein levels were normalized to β-actin levels. j CASP8 activity (n = 6). The data shown are representative of at least three independent experiments. Data are represented as the mean ± SD. *P < 0.05, **P < 0.01, experiments were assessed by one-way ANOVA, two-way ANOVA or two-tailed unpaired t-test

Pyroptosis is a newly discovered cell death mechanism pyroptosis by eliciting GSDMD cleavage, thus promoting mediated by inflammasome-dependent CASP1 activation IL-1β secretion and LDH release. These findings indicate and is characterized by the presence of multiple pores in that activated microglia-triggered GSDMD-dependent lipid membranes formed by the N-terminus of GSDMD pyroptosis contributes to ischemia-induced RGCs death. [19, 20]. The precise function of GSDMD has attracted One reasonable explanation for this finding is that dis- increasing attention. We therefore elucidated the role of ruption of GSDMD in microglia reduces pore formation GSDMD in DAMP-induced retinal injury using in the membrane and reduces leakage of cytoplasmic GSDMD-deficient mice subjected to substantial elevated components including bioactive IL-1β, thus ameliorating IOP. Interestingly, our study firstly revealed, for the first neuroinflammation and RGCs loss. time, that GSDMD elimination significantly improves To gain better insight into the upstream molecular RGCs survival and reduces the severity of retinal tissue mechanisms of pyroptosis, we next focused on inflam- injury, indicating that GSDMD-predominant pyroptosis masomes. NLRP3 is a classic sensor and has been re- may be a potential therapeutic target for RGCs loss and ported to drive GSDMD cleavage in response to retinal tissue injury. Microglia are gatekeepers of neu- pathogen infection [56]; therefore, we investigated the rons in the CNS, and overactivated microglia harm roles of the newly discovered NLRs NLRP12 and RGCs by releasing inflammatory cytokines and recruit- NLRC4. Unlike that of NLRP3, the effect of NLRP12 has ing inflammatory effector cells to the insult area to mag- been controversial. Several reports state that NLRP12 nify cascade of inflammation [55]. By means of an functions as a negative regulator of inflammation in in vitro model mimicking the physiology of RIR injury, tumorigenesis [57–59], colitis [60, 61] and obesity [62], our study shows that microglial activation initiates while Wang et al.report that NLRP12 can activate Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 13 of 16

Fig. 8 Diagram illustrating the pathway by which the CASP8-HIF-1α- NLRP12/NLRP3/NLRC4-IL1β-pyroptosis circuit contributes to the pathogenesis of acute glaucoma inflammatory signaling pathways by regulating the acti- influences the activation of CASP8 in elevated IOP- vation of NF-kB and CASP1-dependent cytokine pro- induced retinal ischemic injury requires future study. cessing [63]. In current study, we found for the first time HIF-1α has been determined to play vital roles in certain that genetic deletion of NLRP12 strikingly attenuates the ischemic diseases, such as ischemic stroke, liver, kidney is- severity of RGCs loss and retinal damage, validating chemia/reperfusion injury [66–68], and retinal ischemic NLRP12 activation is a trigger for retinal ischemic injury injury [69], but the exact mechanisms in acute glaucoma in acute glaucoma. Moreover, NLRP12 was detected to remain unclear. Previous studies have demonstrated that collaborate with NLRP3 and NLRC4 to promote pyrop- HIF-1α can mediate NLRP3 inflammasome activation and tosis by eliciting CASP1-dependent N-terminal GSDMD IL-1β secretion [70–72]. In additiong, the NLRP3 inflam- cleavage. Together, these findings depict a novel func- masome can be activated via CASP8 stimulation [73]. We tion for NLRP12 in pyroptosis with regard to the patho- therefore hypothesized that there may be a regula- genesis of retinal ischemia and RGCs death, indicating tory link between CASP8 and HIF-1α.First,weval- that NLRP12 coordinates with NLRP3 and NLRC4 to idated the triggering effects of HIF-1α and CASP8 exert both neuroinflammatory and pyroptotic effects on pyroptosis in vivo and in vitro, respectively. Sec- under ischemic conditions. ond, we identified a novel role of CASP8 in indu- Accumulating evidence shows that GSDMD prevents cing HIF-1α signaling via NF-kB translocation: CASP8 activation at the NLRC4 or NLRP3 inflamma- CASP8 promoted NF-kB translocation to drive HIF- somes under conditions of exposure to a bacterial inflam- 1α signaling and thus exacerbate pyroptosis and masome activator [64, 65]. We thus investigated the retinal damage. To elucidate the downstream mo- possible link between CASP8 and GSDMD. Our findings lecular pathway of CASP8-HIF-1α signaling, we reveal the unique effect of CASP8 on pyroptosis: CASP8 employed selective knockdown strategies. CASP8- initiates GSDMD-predominant pyroptosis during retinal HIF-1α signaling inhibition is associated with re- ischemia, in accordance with the results during pathogen duced NLRP12/NLRP3/NLRC4 activation, suggest- infection [50, 51]. However, whether GSDMD in turn ing that CASP8-HIF-1α signaling is the upstream Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 14 of 16

regulator of NLRP3/NLRP12/NLRC4 in high IOP- Supplementary information induced retinal ischemic injury. Together, these Supplementary information accompanies this paper at https://doi.org/10. 1186/s13024-020-00372-w. findings demonstrate that CASP8 induces the HIF- α β 1 -NLRP12/NLRP3/NLRC4-IL-1 axis, exacerbating Additional file 1 Supplementary Figure 1. Suppressing NLRP12/NLRP3/ retinal ischemic neuroinflammation. NLRC4 significantly improves RGCs survival. a Representative IL-1β is an important inflammatory mediator of the immunofluorescence images of retinas from WT and NLRP12 KO mice subjected to RIR injury (n = 6). RBPMS was used to identify RGCs (red). CNS [74]. In the current study, we explore the functions Scale bar: 100 μm. b Representative immunofluorescence images of of IL-1β in mediating neuroinflammation during elevated retinas from mice subjected to RIR injury (n = 6). RBPMS was used to IOP-induced retinal ischemic injury in acute glaucoma. identify RGCs (red). Scale bar: 100 μm. WT: wide type; KO: knockout; RIR: retinal ischemia-reperfusion; GCL: ganglion cell layer; INL: inner nuclear Our findings revealed novel reciprocal interactions be- layer; ONL: outer nuclear layer; si: siRNA. The data shown are representa- tween mature IL-1β and NLRP12/NLRP3/NLRC4, sug- tive of at least three independent experiments. gesting that the NLR-dependent maturation of IL-1β Additional file 2 Supplementary Figure 2. The expression of NLRP3/ positively enhances the activation of multiple proinflam- NLRP12/ NLRC4 and GSDMD in primary microglia. a Representative immunofluorescence images of murine primary microglia, identified by matory NLRs to amplify the inflammatory cascades. We the microglial-specific marker IBA-1 (n = 6). Scale bar: 100 μm. b-e Repre- also found that IL-1β boosts CASP8 and HIF-1α activa- sentative immunofluorescence images of expression of NLRP3 /NLRP12/ tion, implicating IL-1β as a crucial mediator that amplifies NLRC4 and GSDMD in murine primary microglia exposed to OGDR treat- ment (n = 6). Scale bar: 100 μm. The data shown are representative of at the neuroinflammatory cascades in elevated IOP-induced least three independent experiments. retinal ischemic injury by driving CASP8-HIF-1α signaling and subsequently activating various NLRs. It has been β Abbreviations demonstrated that GSDMD controls IL-1 secretion [75, ARRIVE: Animal Research: Reporting of In Vivo Experiments; CASP8: Caspase- 76], and our study validates a new role for IL-1β as a pro- 8; CASP1: Caspase-1; CNS: Central nervous system; DAMP: Danger-associated moter of GSDMD cleavage to induce pyroptosis, suggest- molecular pattern; DMEM: Dulbecco’s Modified Eagle’s Medium; FG: Fluoro- β gold; FBS: Fetal bovine serum; GSDMD: Gasdermin D; HIF-1α: Hypoxia ing that IL-1 aggravates pyroptosis during high IOP- activates hypoxia-inducible factor-1α; H & E: Hematoxylin and eosin staining; induced retinal ischemic injury. Given these novel find- IOP: Intraocular pressure; LPS: Lipopolysaccharide; NLR: Nucleotide-binding ings, we have determined that IL-1β is a central factor en- leucine-rich repeat containing receptor; NLRC4: NLR family CARD domain- α containing protein 4; NLRP3: NLR family pyrin domain-containing 3; hancing CASP8-HIF-1 signaling to subsequently increase NLRP12: NLR family pyrin domain-containing 12;; OGDR: Oxygen and glucose NLRP3/NLRP12/NLRC4 activation and to initiate pyrop- deprivation/reoxygenation; PVDF: Polyvinylidene difluoride; tosis during retinal ischemia. PFA: Paraformaldehyde; RIR: Retinal ischemia-reperfusion; RGCs: Retinal ganglion cells; SEM: Scanning electron microscope; TLR4: Toll-like receptor 4

Conclusions Acknowledgements Our study has revealed the novel mechanisms of pyropto- Not applicable. sis and neuroinflammation associated with irreversible RGCs death and vision loss in acute glaucoma. We have Author contributions Study concept and design: W.C. and H.C. Experimental and technical demonstrated that genetic deletion of GSDMD reduces support: W.C., H.C., L.S., X.G., Y.L., J.L. and Y.H. Acquisition, analysis, or RGCs death and the severity of retinal tissue injury. Re- interpretation of data: W.C., H.C., Y.D.,W.H., Y.L., L.L., Y.C. and L.S. RNA markably, our findings reveal for the first time reveal, that sequence analysis: L.W. and B.Z. Drafting of the manuscript: W.C., H.C. and α Y.D. Critical revision of the manuscript for important intellectual content: CASP8 can promote HIF-1 signaling via NF-kB trans- W.C., H.C., Y.D. and Y.L. Statistical analysis: W.C., H.C., L.S., X.G. and Y.H. All location. Moreover, NLRP12 coordinates with NLRP3 and authors read and approved the final manuscript. NLRC4 to induce IL-1β processing and pyroptotic cell death in a manner dependent on the CASP8-HIF-1α path- Funding The Science and Technology Project of Guangzhou of Wei Chi way, and genetic deletion of CASP8 depresses pyroptosis (No.201804010415), the National Natural Science Foundation of Key Projects dependent on CASP1-induced GSDMD cleavage. Further- of China of Yizhi Liu (No.81530028) and the National Natural Science more, mature IL-1β initiates the neuroinflammation cas- Foundation of Xiaoliang Gan (No.81571884). The funding organization had no role in the design or execution of this research. cades and pyroptotic reaction by promoting the CASP8- α HIF-1 -NLRP12/NLRP3/NLRC4 pathway and GSDMD Availability of data and materials cleavage. This process provides a positive feedback circuit The datasets used and/or analyzed during the current study are available which is likely to magnify the inflammatory cascades dur- from the corresponding author on reasonable request. ing retinal ischemic injury. Our study provides novel in- Ethics approval and consent to participate sights into the roles of pyroptosis and neuroinflammation All the animals were treated in strict accordance with Animal Research: in elevated IOP-induced retinal ischemic injury and sug- Reporting of In Vivo Experiments (ARRIVE) guidelines, and this study was gests that targeting constraint of CASP8-HIF-1α- formally reviewed and approved by the Zhongshan Ophthalmic Center Animal Care and Ethics Committee (2014–003). NLRP12/NLRP3/NLRC4-initiated neuroinflammation and pyroptosis may be a promising strategy for innovative Consent for publication treatment of acute glaucoma. Not applicable. Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 15 of 16

Competing interests 20. Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD The authors declare no competing financial interests. by inflammatory determines pyroptotic cell death. Nature. 2015; 526(7575):660–5. Author details 21. Liu Y, Zhang T, Zhou Y, Li J, Liang X, Zhou N, et al. Visualization of perforin/ 1State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, gasdermin/complement-formed pores in real cell membranes using atomic Sun Yat-sen University, Guangzhou 510060, China. 2Department of force microscopy. Cell Mol Immunol. 2019;16(6):611–20. Microbiology, Tumor and Cell Biology, Karoslinska Institute, 17177 Stockholm, 22. Broderick L, De Nardo D, Franklin BS, Hoffman HM, Latz E. The Sweden. inflammasomes and autoinflammatory syndromes. Annu Rev Pathol. 2015; 10:395–424. Received: 23 July 2019 Accepted: 25 March 2020 23. Sagoo P, Garcia Z, Breart B, Lemaitre F, Michonneau D, Albert ML, et al. In vivo imaging of inflammasome activation reveals a subcapsular burst response that mobilizes innate and adaptive immunity. Nat Med. 2016;22(1):64–71. References 24. Place DE, Kanneganti TD. Recent advances in inflammasome biology. Curr 1. Tanner L, Gazzard G, Nolan WP, Foster PJ. Has the EAGLE landed for the use Opin Immunol. 2018;50:32–8. of clear lens extraction in angle-closure glaucoma? And how should 25. Karki R, Kanneganti TD. Diverging inflammasome signals in tumorigenesis primary angle-closure suspects be treated? Eye (London, England). 2020; and potential targeting. Nat Rev . 2019;19(4):197–214. 34(1):40–50. 26. Lukens JR, Gurung P, Shaw PJ, Barr MJ, Zaki MH, Brown SA, et al. The 2. Tham YC, Li X, Wong TY, Quigley HA, Aung T, Cheng CY. Global prevalence NLRP12 sensor negatively regulates autoinflammatory disease by of glaucoma and projections of glaucoma burden through 2040: a modulating Interleukin-4 production in T cells. Immunity. 2015;42(4):654–64. systematic review and meta-analysis. Ophthalmology. 2014;121(11):2081–90. 27. Chen L, Wilson JE, Koenigsknecht MJ, Chou WC, Montgomery SA, Truax AD, 3. Zhu X, Zeng W, Wu S, Chen X, Zheng T, Ke M. Measurement of retinal et al. NLRP12 attenuates colon inflammation by maintaining colonic changes in primary acute angle closure Glaucoma under different durations microbial diversity and promoting protective commensal bacterial growth. of symptoms. J Ophthalmol. 2019;2019:5409837. Nat Immunol. 2017;18(5):541–51. 4. Almasieh M, Wilson AM, Morquette B, Cueva Vargas JL, Di Polo A. The 28. Platnich JM, Chung H, Lau A, Sandall CF, Bondzi-Simpson A, Chen HM, et al. molecular basis of retinal ganglion cell death in glaucoma. Prog Retin Eye Shiga Toxin/Lipopolysaccharide Activates Caspase-4 and Gasdermin D to Res. 2012;31(2):152–81. Trigger Mitochondrial Upstream of the NLRP3 5. Minhas G, Sharma J, Khan N. Cellular stress response and immune signaling Inflammasome. Cell Rep. 2018;25(6):1525–36 e7. in retinal ischemia-reperfusion injury. Front Immunol. 2016;7:444. 29. Kang R, Zeng L, Zhu S, Xie Y, Liu J, Wen Q, et al. Lipid Peroxidation Drives 6. Langmann T. Microglia activation in retinal degeneration. J Leukoc Biol. Gasdermin D-Mediated Pyroptosis in Lethal Polymicrobial . Cell Host 2007;81(6):1345–51. Microbe. 2018;24(1):97–108 e4. 7. Chi W, Li F, Chen H, Wang Y, Zhu Y, Yang X, et al. Caspase-8 promotes 30. Denko NC. Hypoxia, HIF1 and glucose metabolism in the solid tumour. Nat NLRP1/NLRP3 inflammasome activation and IL-1beta production in acute Rev Cancer. 2008;8(9):705–13. glaucoma. Proc Natl Acad Sci U S A. 2014;111(30):11181–6. 31. Keith B, Johnson RS, Simon MC. HIF1alpha and HIF2alpha: sibling rivalry in 8. Cueva Vargas JL, Belforte N, Di Polo A. The glial cell modulator ibudilast hypoxic tumour growth and progression. Nat Rev Cancer. 2011;12(1):9–22. attenuates neuroinflammation and enhances retinal ganglion cell viability in 32. Montagner M, Enzo E, Forcato M, Zanconato F, Parenti A, Rampazzo E, et al. glaucoma through protein kinase a signaling. Neurobiol Dis. 2016;93:156–71. SHARP1 suppresses breast cancer metastasis by promoting degradation of 9. Silverman SM, Kim BJ, Howell GR, Miller J, John SW, Wordinger RJ, et al. C1q hypoxia-inducible factors. Nature. 2012;487(7407):380–4. propagates microglial activation and neurodegeneration in the visual axis 33. Cai Z, Luo W, Zhan H, Semenza GL. Hypoxia-inducible factor 1 is required following retinal ischemia/reperfusion injury. Mol Neurodegener. 2016;11:24. for remote ischemic preconditioning of the heart. Proc Natl Acad Sci U S A. 10. Gong Y, Cao X, Gong L, Li W. Sulforaphane alleviates retinal ganglion cell 2013;110(43):17462–7. death and inflammation by suppressing NLRP3 inflammasome activation in 34. Greer SN, Metcalf JL, Wang Y, Ohh M. The updated biology of hypoxia- a rat model of retinal ischemia/reperfusion injury. Int J Immunopathol inducible factor. EMBO J. 2012;31(11):2448–60. Pharmacol. 2019;33:2058738419861777. 35. Wen H, Ting JP. Agitation by suffocation: how hypoxia activates innate 11. Coucha M, Shanab AY, Sayed M, Vazdarjanova A, El-Remessy AB. Modulating immunity via the Warburg effect. Cell Metab. 2013;17(6):814–5. Expression of Thioredoxin Interacting Protein (TXNIP) Prevents Secondary 36. Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF, Damage and Preserves Visual Function in a Mouse Model of Ischemia/ Goel G, et al. Succinate is an inflammatory signal that induces IL-1beta Reperfusion. Int J Mol Sci. 2019;20(16). through HIF-1alpha. Nature. 2013;496(7444):238–42. 12. Qi Y, Zhao M, Bai Y, Huang L, Yu W, Bian Z, et al. Retinal ischemia/ 37. Stockslager MA, Samuels BC, Allingham RR, Klesmith ZA, Schwaner SA, reperfusion injury is mediated by toll-like receptor 4 activation of NLRP3 Forest CR, et al. System for rapid, precise modulation of intraocular pressure, inflammasomes. Invest Ophthalmol Vis Sci. 2014;55(9):5466–75. toward minimally-invasive in vivo measurement of intracranial pressure. 13. Dvoriantchikova G, Barakat DJ, Hernandez E, Shestopalov VI, Ivanov D. Toll- PLoS One. 2016;11(1):e0147020. like receptor 4 contributes to retinal ischemia/reperfusion injury. Mol Vis. 38. Shi J, Gao W, Shao F. Pyroptosis: Gasdermin-mediated programmed necrotic 2010;16:1907–12. cell death. Trends Biochem Sci. 2017;42(4):245–54. 14. de Gassart A, Martinon F. Pyroptosis: Caspase-11 unlocks the gates of death. 39. He Y, Hara H, Nunez G. Mechanism and regulation of NLRP3 Inflammasome Immunity. 2015;43(5):835–7. activation. Trends Biochem Sci. 2016;41(12):1012–21. 15. Aglietti RA, Dueber EC. Recent insights into the molecular mechanisms 40. Wree A, Eguchi A, McGeough MD, Pena CA, Johnson CD, Canbay A, et al. underlying Pyroptosis and Gasdermin family functions. Trends Immunol. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver 2017;38(4):261–71. inflammation, and fibrosis in mice. Hepatology. 2014;59(3):898–910. 16. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and 41. Frakes AE, Ferraiuolo L, Haidet-Phillips AM, Schmelzer L, Braun L, Miranda CJ, inflammation. Nat Rev Microbiol. 2009;7(2):99–109. et al. Microglia induce motor neuron death via the classical NF-kappaB 17. Julien O, Wells JA. Caspases and their substrates. Cell Death Differ. 2017; pathway in amyotrophic lateral sclerosis. Neuron. 2014;81(5):1009–23. 24(8):1380–9. 42. Baruch K, Kertser A, Porat Z, Schwartz M. Cerebral nitric oxide represses 18. Maltez VI, Tubbs AL, Cook KD, Aachoui Y, Falcone EL, Holland SM, et al. choroid plexus NFkappaB-dependent gateway activity for leukocyte Inflammasomes coordinate Pyroptosis and natural killer cell cytotoxicity to trafficking. EMBO J. 2015;34(13):1816–28. clear infection by a ubiquitous environmental bacterium. Immunity. 2015; 43. Kaiser WJ, Upton JW, Long AB, Livingston-Rosanoff D, Daley-Bauer LP, 43(5):987–97. Hakem R, et al. RIP3 mediates the embryonic lethality of caspase-8-deficient 19. Liu X, Zhang Z, Ruan J, Pan Y, Magupalli VG, Wu H, et al. Inflammasome- mice. Nature. 2011;471(7338):368–72. activated gasdermin D causes pyroptosis by forming membrane pores. 44. Varfolomeev EE, Schuchmann M, Luria V, Chiannilkulchai N, Beckmann JS, Nature. 2016;535(7610):153–8. Mett IL, et al. Targeted disruption of the mouse Caspase 8 gene ablates cell Chen et al. Molecular Neurodegeneration (2020) 15:26 Page 16 of 16

death induction by the TNF receptors, Fas/Apo1, and DR3 and is lethal 67. Guo Y, Feng L, Zhou Y, Sheng J, Long D, Li S, et al. Systematic review with prenatally. Immunity. 1998;9(2):267–76. meta-analysis: HIF-1alpha attenuates liver ischemia-reperfusion injury. 45. Zhu Y, Zhang L, Gidday JM. Role of hypoxia-inducible factor-1alpha in Transplant Rev (Orlando). 2015;29(3):127–34. preconditioning-induced protection of retinal ganglion cells in glaucoma. 68. Davis CK, Jain SA, Bae ON, Majid A, Rajanikant GK. Hypoxia mimetic agents Mol Vis. 2013;19:2360–72. for ischemic stroke. Front Cell Dev Biol. 2018;6:175. 46. Zhou J, Chen F, Yan A, Xia X. Role of mammalian target of rapamycin in 69. Park SW, Lee HS, Sung MS, Kim SJ. The effect of melatonin on retinal regulating HIF-1alpha and vascular endothelial growth factor signals in ganglion cell survival in ischemic retina. Chonnam Med J. 2012;48(2):116–22. glaucoma. Arch Physiol Biochem. 2019:1–7. 70. Martinez-Garcia JJ, Martinez-Banaclocha H, Angosto-Bazarra D, de Torre- 47. Ji YS, Park JW, Heo H, Park JS, Park SW. The neuroprotective effect of Minguela C, Baroja-Mazo A, Alarcon-Vila C, et al. P2X7 receptor induces carnosine (beta-alanyl-L-histidine) on retinal ganglion cell following mitochondrial failure in monocytes and compromises NLRP3 inflammasome ischemia-reperfusion injury. Curr Eye Res. 2014;39(6):634–41. activation during sepsis. Nat Commun. 2019;10(1):2711. 48. Cheng L, Yu H, Yan N, Lai K, Xiang M. Hypoxia-inducible factor-1alpha target 71. Gupta N, Sahu A, Prabhakar A, Chatterjee T, Tyagi T, Kumari B, et al. genes contribute to retinal Neuroprotection. Front Cell Neurosci. 2017;11:20. Activation of NLRP3 inflammasome complex potentiates venous thrombosis 49. Seong H, Ryu J, Yoo WS, Kim SJ, Han YS, Park JM, et al. Resveratrol in response to hypoxia. Proc Natl Acad Sci U S A. 2017;114(18):4763–8. ameliorates retinal ischemia/reperfusion injury in C57BL/6J mice via 72. Talreja J, Talwar H, Bauerfeld C, Grossman LI, Zhang K, Tranchida P, et al. Downregulation of Caspase-3. Curr Eye Res. 2017:1–9. HIF-1alpha regulates IL-1beta and IL-17 in sarcoidosis. eLife. 2019;8. 50. Sarhan J, Liu BC, Muendlein HI, Li P, Nilson R, Tang AY, et al. Caspase-8 73. Antonopoulos C, Russo HM, El Sanadi C, Martin BN, Li X, Kaiser WJ, et al. induces cleavage of gasdermin D to elicit pyroptosis during Yersinia Caspase-8 as an effector and regulator of NLRP3 Inflammasome signaling. J infection. Proc Natl Acad Sci U S A. 2018;115(46):E10888–E97. Biol Chem. 2015;290(33):20167–84. 51. Orning P, Weng D, Starheim K, Ratner D, Best Z, Lee B, et al. Pathogen 74. Yamasaki Y, Matsuura N, Shozuhara H, Onodera H, Itoyama Y, Kogure K. blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D Interleukin-1 as a pathogenetic mediator of ischemic brain damage in rats. and cell death. Science. 2018;362(6418):1064–9. Stroke. 1995;26(4):676–80 discussion 81. 52. Della Santina L, Inman DM, Lupien CB, Horner PJ, Wong RO. Differential 75. Evavold CL, Ruan J, Tan Y, Xia S, Wu H, Kagan JC. The pore-forming protein progression of structural and functional alterations in distinct retinal ganglion Gasdermin D regulates Interleukin-1 secretion from living . cell types in a mouse model of glaucoma. J Neurosci. 2013;33(44):17444–57. Immunity. 2017. 53. See JLS, Aquino MCD, Aduan J, Chew PTK. Management of angle closure 76. He WT, Wan H, Hu L, Chen P, Wang X, Huang Z, et al. Gasdermin D is an glaucoma. Indian J Ophthalmol. 2011;59 Suppl (Suppl1):S82-SS7. executor of pyroptosis and required for interleukin-1beta secretion. Cell Res. – 54. Wang K, Peng B, Lin B. Fractalkine receptor regulates microglial neurotoxicity in 2015;25(12):1285 98. an experimental mouse glaucoma model. Glia. 2014;62(12):1943–54. 55. Liu X, Huang P, Wang J, Yang Z, Huang S, Luo X, et al. The effect of A2A Publisher’sNote receptor antagonist on microglial activation in experimental Glaucoma. Springer Nature remains neutral with regard to jurisdictional claims in – Invest Ophthalmol Vis Sci. 2016;57(3):776 86. published maps and institutional affiliations. 56. Dubois H, Sorgeloos F, Sarvestani ST, Martens L, Saeys Y, Mackenzie JM, et al. Nlrp3 inflammasome activation and Gasdermin D-driven pyroptosis are immunopathogenic upon gastrointestinal norovirus infection. PLoS Pathog. 2019;15(4):e1007709. 57. Udden SN, Kwak YT, Godfrey V, Khan MAW, Khan S, Loof N, et al. NLRP12 suppresses via downregulation of cJun N-terminal kinase activation in the hepatocyte. eLife. 2019;8. 58. Zaki MH, Vogel P, Malireddi RK, Body-Malapel M, Anand PK, Bertin J, et al. The NOD-like receptor NLRP12 attenuates colon inflammation and tumorigenesis. Cancer Cell. 2011;20(5):649–60. 59. Tuncer S, Fiorillo MT, Sorrentino R. The multifaceted nature of NLRP12. J Leukoc Biol. 2014;96(6):991–1000. 60. Shi F, Yang Y, Kouadir M, Xu W, Hu S, Wang T. Inflammasome-independent role of NLRP12 in suppressing colonic inflammation regulated by Blimp-1. Oncotarget. 2016;7(21):30575–84. 61. Allen IC, Wilson JE, Schneider M, Lich JD, Roberts RA, Arthur JC, et al. NLRP12 suppresses colon inflammation and tumorigenesis through the negative regulation of noncanonical NF-kappaB signaling. Immunity. 2012; 36(5):742–54. 62. Truax AD, Chen L, Tam JW, Cheng N, Guo H, Koblansky AA, et al. The Inhibitory Innate Immune Sensor NLRP12 Maintains a Threshold against Obesity by Regulating Gut Microbiota Homeostasis. Cell Host Microbe. 2018; 24(3):364–78 e6. 63. Wang L, Manji GA, Grenier JM, Al-Garawi A, Merriam S, Lora JM, et al. PYPAF7, a novel PYRIN-containing Apaf1-like protein that regulates activation of NF-kappa B and caspase-1-dependent cytokine processing. J Biol Chem. 2002;277(33):29874–80. 64. Mascarenhas DPA, Cerqueira DM, Pereira MSF, Castanheira FVS, Fernandes TD, Manin GZ, et al. Inhibition of caspase-1 or gasdermin-D enable caspase- 8 activation in the Naip5/NLRC4/ASC inflammasome. PLoS Pathog. 2017; 13(8):e1006502. 65. Schneider KS, Gross CJ, Dreier RF, Saller BS, Mishra R, Gorka O, et al. The Inflammasome drives GSDMD-independent secondary Pyroptosis and IL-1 release in the absence of Caspase-1 protease activity. Cell Rep. 2017;21(13): 3846–59. 66. Xie Y, Jiang D, Xiao J, Fu C, Zhang Z, Ye Z, et al. Ischemic preconditioning attenuates ischemia/reperfusion-induced kidney injury by activating autophagy via the SGK1 signaling pathway. Cell Death Dis. 2018;9(3):338.