1. Introduction
The eye challenged with chronic OHT, like in the ocular hypertension type of glaucoma, is exposed to multiple mechanical, ischemic and metabolic stresses that affect essential RGC functions, including axonal transport, energy homeostasis, electrophysiological outputs and glial activation[
1,
2,
3,
4,
5]. Repetitive short-term OHT episodes, caused by intraocular pressure (IOP) “spikes”, are also recognized as important risk factors [
6,
7], though these are much less studied than chronic OHT. Mounting clinical and experimental evidence from animal studies indicate that transient IOP spikes are more damaging to vision when they are recurrent and/or reach the ischemic level, such as with eye rubbing [
8,
9]. Recurrent OHT spikes are typically induced by medications, eye surgeries, intraocular drug injections, head-down postures during post-surgical recovery, or some lifestyle activities, including playing wind instruments, weightlifting or head-down positions in yoga [
10,
11,
12,
13]. Repeated incidence of OHT spikes have been linked to nerve fiber layer thinning and vision loss in human eyes [
8,
14]. For example, repetitive intraocular drug injections were associated with vascular hypoperfusion [
15,
16,
17], nerve fiber layer thinning [
17,
18] and it has also been suggested to play a role in normal tension glaucoma [
9,
13,
19]. Although spOHT and chronic OHT modalities differ dramatically by the type of the initial injury, both could produce similar RGC and axon injuries clinically presented as glaucomatous [
8,
14]. To the best of our knowledge, the present work is the first to compare neuroinflammatory responses and their link to RGC pathology following spOHT and chronic OHT.
Neuroinflammation and accumulation of pro-inflammatory cytokines, the two common end-products of active inflammasomes[
20,
21,
22,
23], have been detected in animal[
24] and human[
25,
26,
27,
28] eyes with glaucoma along with an increase in extracellular ATP, a common inducer of the inflammasome[
29]. At the cellular level, neuroinflammatory responses mediated by the release of IL-1b and tumor necrosis factor (TNF-a) have been confirmed in astrocytes, Muller glia, microglia, and in RGCs in glaucomatous eyes [
25,
30,
31,
32]. Recent reports from us and others have shown very early activation of neuronal NLRP1/3 inflammasome, caspase-1, and the release of IL-1β in a rodent model of ischemic OHT injury [
33,
34,
35], suggesting that inflammasome signaling may serve as the key trigger of glial responses. The key role of inflammation and inflammatory caspases, like Casp8, in the pathogenesis of glaucoma induced by non-ischemic OHT injury has been recently shown [
36,
37].
An active role of inflammasome activation in the pathogenesis of retinal OHT damage [
38,
39] could shift the current paradigm where inflammation is considered secondary to optic nerve damage and RGC pathology [
40]. To gain insight into how the short-term repetitive non-ischemic OHT spikes lead to RGC injury, we investigated the impact of activation of retinal inflammasomes on RGC function and viability using a mouse model of IOP spike-induced (spOHT) injury. Here we report that repetitive episodes of non-ischemic IOP spikes lead to impaired RGC electrophysiological function and cell death. Our results strongly suggest that the effects of spiking OHT are mediated predominantly by acute innate immune responses triggered by the activation of the endogenous neuronal NLRP1 inflammasome. Our results also identify key components of the inflammasome, along with its upstream regulator the mechanosensitive Panx1 channels, as therapeutic targets for OHT-mediated RGC degeneration.
The present work is an important step towards a better understanding of the underlying molecular mechanism through spiking OHT-induced degenerative stress in RGCs. Furthermore, our results from multiple inflammasome knockout mice indicate that acute activation of inflammasomes and their downstream products are mechanistically involved in the OHT-induced RGC dysfunction and loss, while their inactivation protected RGCs.
2. Materials and Methods
Animals. Animal handling, anesthetic procedures, experiments and post-surgical care were performed in compliance with the NIH Guide for the Care and Use of Laboratory Animals and according to the University of Miami, Institutional Animal Care and Use Committee approved protocols #18-025 and #21-036. Wildtype (WT, C57BL/6J) and transgenic mice, including Casp1
-/- (Casp1/Casp4(11)
del strain B6N.129S2-Casp1tm1Flv/J, Jax # 016621); NLRP1b
-/- (B6.129S6-
Nlrp1btm1Bhk/J, Jax # 021301), and GsdmD
-/- (C57BL/6N-Gsdmdem4Fcw/J, Jax # 032410), were obtained from Laboratories Depository (Bar Harbor, ME). Mice expressing ASC-citrine for ASC speck visualization were provided by Dr. D. Golenbock [
41], University of Massachusetts Med. School. All animals were bred in the University of Miami animal facility and housed under standard conditions of temperature and humidity with a 12-h light/dark cycle and free access to food and water.
Reagents. Antibodies were purchased from commercial sources: anti-GFAP (Dako, cat#z0334), anti-AIF1/Iba1 (Wako, cat#019-19741), anti-Casp1 (Novus Biologicals, cat#IMG5028); caspase-1, p-20 (Adipogen, cat# AG-20B-0042); anti-IL-1β (Cell Signaling, cat#8689S), anti-NLRP1 (Novus cat# NB100-561148SS); anti-NLRP3 (R&D, cat#AF7010); anti-RBPMS (GeneTex, cat#118619); anti-Brn3a (SantaCruz, cat# sc31984); anti- Class III β-Tubulin (clone TUJ1, Covance); anti-GSDMDC1 (A7, Santa Cruz, cat# sc-271054); anti-Casp8 monoclonal antibody (1G12) (Enzo, cat# ALX-804-447); anti-ASC-1 (F-9, Santa Cruz, cat# sc-271054); anti-CD11b (Santa Cruz, cat# sc-271050); anti-CD45 (Santa Cruz, cat# sc-271024).
The Spiking OHT injury model. The mean arterial pressure in murine eyes, is 112 mm Hg [
42] and transient non-ischemic IOP elevations to 30-40 mm Hg were shown to be not injurious [
43,
44,
45]. The spiking OHT model, however, was administered by seven consecutive IOP elevations of 40 mmHg. During the procedure animals were under isoflurane (3%) gas anesthesia and topical analgesia was induced with 0.5% proparacaine HCl (Bausch & Lomb Pharmaceuticals, Rochester, NY). Pupils were dilated with 1% tropicamide and 2.5% phenylephrine hydrochloride (NutraMax Products, Inc., Gloucester, MA) to aid in placement of the pressure input needle. IOP was elevated by cannulation of the anterior chamber with a 29G needle connected to a reservoir of normal saline (0.9% NaCl) that was placed 54 cm above the eye level to achieve an H
2O pressure equivalent to 40 mm Hg, as described [
33,
43]. A digital in-line digital mini-pressure gauge (Centurion Compass CUHG; precision ±1.5 mm Hg) was used to monitor IOP elevation and potential loss of pressure from leaks. The spOHT experimental paradigm, shown in
Figure 1A, consisted of seven consecutive 1-minute IOP spikes (spOHT) achieved by a rapid rise and consecutive lowering of the reservoir, with a 1-minute interval of normotension between spikes and quick transition between normal and elevated IOP. In the steady OHT group, IOP was elevated by a gradual 1-minute increase to 40 mmHg, which was maintained for 7 minutes, then returned to baseline gradually over 1 minute. Thus, both models produced the same maximum IOP exposure to 40 mm Hg for a total of 7 min and were induced by a single needle insertion through the peripheral cornea. Sham control procedure was performed by a single cannulation of the anterior chamber with no elevation of the reservoir (i.e. no IOP elevation) under anesthesia for the same duration as the experimental groups.
The Y437H-Myoc-induced OHT model. In this model, IOP was elevated by over expressing the pathogenic Y437H variant of human Myocilin, as described earlier by Grotegut and Kuehn [
46,
47]. To over express Y437H human Myocilin, the anterior chamber of each eye of an animal was injected with 1.4 µl of Ad5-Myoc suspension (5X10
7 pfu/eye) using 33G needle. A gradual IOP increase to 25-35mm was observed at 6-8 weeks in all mouse lines tested. Eyes were harvested and inner retinas collected at 3, 5 and 8 weeks after vector injection.
The retinal ischemia-reperfusion model. Retinal ischemia was achieved by increasing IOP above systolic blood pressure to 110 mm Hg for 45 min by direct cannulation of the anterior chamber with a 29 G needle connected to a normal saline (0.9% NaCl)-filled reservoir placed at 150 cm above the eye to create a pressure of 150 cm H
2O (equivalent to 110 mm Hg), as previously described[
33]. The pressure changes in the tube connected to the needle were calibrated using an in-line Centurion Compass CUHG1 digital pressure transducer (Centurion Medical Products Inc) prior to experiments. The contralateral eyes, cannulated at normal IOP, served as normotensive controls. Complete retinal ischemia was confirmed as the whitening of the anterior segment and blanching of the retinal arteries. Eyes were harvested 24h post-injury, mice were euthanized by CO
2 overdose and eyes were collected. The vitreous body was harvested for cytokine analysis by ELISA, and retinas were dissected out, fixed and processed.
Intravitreal cytokine activity assay. To collect the vitreous, mice were perfused with phosphate-buffered saline (PBS), eyes were collected, placed on ice, and immediately dissected. Vitreous fluid was collected with three consecutive flushes of the vitreous cavity with 20 µl sterile PBS containing a protease inhibitor cocktail. All flush samples were combined, spun for 5 min in a refrigerated centrifuge, and stored at -80ºC. ELISA kits for mouse IL-1b (R&D ID# MBL00C) or Ella SimplePlex for IL1b, TNFa, and MCP1/CCL2 (Protein Simple) as described in [
48] were used to measure cytokine released into the vitreous. Sample aliquots were processed for protein analyses in parallel with standards and controls following the manufacturer’s instructions. A colorimetric assay was done using a FLUOstar Omega plate reader (BMG Labtech) and analyzed using MARS data analysis software (BMG Labtech). Values from the wells containing blank samples were subtracted from the background. To validate the significance of measurements at the lowest reading, the limit of detection (LOD) and limit of quantification (LOQ) ratios were calculated from empirical data obtained in the ‘‘zero’’ wells of each plate, as described[
49]. A minimum of three (N= 3) biological repeats were used for each data point. Significance was calculated using one-way analyses of variance (ANOVA) followed by Tukey’s test for multiple comparisons. To measure the co-release of IL-1b, TNFa and CCL2 cytokines we used the Ella Simple Plex microfluidic technology (Protein Simple) with internal calibration as described in [
48].
In vivo retinal electrophysiology and data analysis. An optimized protocol for PERG (pattern electroretinogram) recording in mice was previously described [
50,
51]. Briefly, animals were anesthetized with ketamine/xylazine (80/10 mg/kg) and gently restrained in an animal holder. PERG signals were recorded simultaneously from both eyes from subdermal electrodes in the snout in response to horizontal bars that maximized PERG amplitude and minimized the noise (spatial frequency 0.05 cycles/deg, temporal frequency 1 Hz, contrast 100%, robust averaging of 2232 sweeps). The PERG signal-to-noise ratio was of the order of 10, and the test–retest variability was of the order of 30% [
52]. Balanced salt solution (BSS) drops were applied to maintain cornea hydration.
RGC loss assessment. To assess RGC loss, retinas were collected at 7 days post-injury 7 dpi, fixed in 4% paraformaldehyde, and flat-mounted. RGCs were identified by RBPMS (RNA binding protein with multiple splicing) immunolabeling, visualized by confocal microscopy. RBPMS-positive cells were counted with ImageJ plugin open-source software after thresholding and manual exclusion of artifacts. Each retina was sampled from 16 fields in 4 retinal quadrants in 3 regions of the same eccentricities (0.5 mm, 1.0 mm, 1.5 mm from the optic disk) as previously described [
33]. RGC loss was calculated as a percentage of RBPMS-positive cells in experimental eyes relative to sham-operated contralateral control eyes. The cell density data (n ≥5) were averaged for each group/genotype; statistical analyses data were analyzed with one-way ANOVA followed by the Tukey test for multiple comparisons; P values ≤0.05 were considered statistically significant.
Inflammasome detection using the citrine-labeled ASC-speck complex in vivo has been described earlier [
34]. ASC-citrine was previously shown to incorporate into oligomerizing inflammasome complexes, thus providing a surrogate inflammasome activation in mouse tissues [
53]. The bioindicator mice expressing ASC fusion protein with a C-terminal citrine protein (fluorescent GFP isoform) that brightly labels the filamentous ASC specks in vivo and allows for visualization in vivo were provided by Dr. D. Golenbock (University of Massachusetts, MA, USA).
Real-time PCR. Gene expression was assessed by real-time PCR using gene-specific primer pairs (primer pairs were validated to span an intron and to amplify only one product (see Supplementary data
Table S1 for details). Total RNA from 2-4 pooled retinas was extracted using Trizol and quality controlled by Nanodrop. cDNA was synthesized with the Reverse Transcription System (Promega, Fitchburg, WI, USA). Real-time PCR was performed in the Rotor-Gene 6000 Cycler (Corbett Research, Mortlake, Australia) using the SYBR GREEN PCR MasterMix (Qiagen, Valencia, CA). Relative expression was calculated by comparison with a standard curve following normalization to the β-actin or Gapdh genes.
Immunohistochemistry. Eyes were enucleated, fixed in 4% paraformaldehyde for 1 h, and cryoprotected with 30% sucrose. Retinas were embedded into the OCT media and frozen-sectioned to a thickness of 10 µm on a microtome (Leica). Slides were washed in PBS, permeabilized in PBS with 0,2% Tween20, and incubated with a primary antibody for 4–16 h. Retinal flat-mounts were incubated with primary antibodies for 3–5 days at 4ºC to ensure even staining. To identify target proteins, specific antibodies were diluted and incubated 4-16h. After washes with PBS-Tween 20, secondary antibodies were applied for 2-4h for frozen sections and for 16h for whole mounts. Secondary AlexaFluor dye-labeled antibodies (Thermo Fisher Scientific, Waltham, MA) were applied for imaging with the Leica TSL AOBS SP5 confocal microscope (Leica Microsystems, Wetzlar, Germany); controls with primary antibodies omitted were used for specificity tests.
Statistical Analysis. Statistical comparisons of PERG data were made using non-parametric Mann–Whitney test and Kruskal-Wallis test followed by post hoc Dunn’s Multiple Comparisons. Protein assay (ELISA, SimplePlex) data were presented as the mean ± standard deviation (SD) or standard error (SEM) for RGC survival data. Real-time PCR data were analyzed with one-way ANOVA followed by the Tukey test for multiple comparisons. For single comparisons, Student’s t-test was applied; one-way ANOVA was used for between-group comparisons. GraphPad Prism software (version 6.07; GraphPad Software, La Jolla, CA, USA) was used for statistical analysis. A minimum of three biological repeats per treatment was used for in vivo IL-1β release assessment and for gene expression analysis by quantitative RT-PCR. Groups of data were compared using ANOVA or two-tailed unpaired Student’s t-tests. Cell density data were analyzed with one-way ANOVA followed by Tukey’s test for multiple comparisons. For two group comparisons, Student’s t-test were carried out. P values < 0.05 were considered statistically significant for all analyses.
Description of common methods, namely immunohistochemistry and Western blot analysis is provided in the Supplementary information.
4. Discussion
In this study, we investigated the impact of immune responses induced by repetitive (spiking) non-ischemic IOP elevations on function and viability of RGCs in mouse eyes. We demonstrated that, in contrast to the stOHT of similar magnitude and duration, the spOHT challenge caused rapid activation of the inflammasome in the retina, resulting in the release of pro-inflammatory cytokines. These events preceded an irreversible dysfunction and caspase-3 dependent loss of RGCs. A lower grade activation of the inflammasome was also observed in 3 weeks post-induction in retinas challenged with chronic OHT in the Ad5-MYOC-induced glaucoma model. Despite major differences in the types of OHT insult in these models, both spOHT and chronic OHT challenges induced RGC dysfunction and death, whereas its inactivation prevented RGC deficits in both models. Our results suggest mechanistic involvement of Casp1, Casp8, NLRP1, NLRP3, GsdmD proteins and inflammatory cytokines expressed by neuronal, glial and infiltrating immune cells in the spOHT-induced RGC pathology.
Acute and progressive inflammasome activation in the spOHT model. In the spOHT model, the production of pro-inflammatory cytokines TNFα and monocyte chemoattractant protein (MCP-1 or CCL2) was dependent on the activity of the Panx1-inflammasome signaling axis. We obtained evidence of mechanistic involvement of the endogenous NLRP1 inflammasome, and its downstream targets GsdmD and Casp1 in the spOHT-induced pathophysiology of RGCs by using gene knockout strains and drug inhibitors. In addition, we also confirmed an essential role of the upstream mechanosensitive regulator of NLRP1 and NLRP3 and the Panx1 channel. RGC dysfunction and loss strongly correlated with activation of NLRP1, NLRP3, and their products, Casp1, IL-1β cytokine, and GsdmD. Importantly, ablation of the
nlrp1b gene was profoundly neuroprotective in the chronic model (
Figure 10D). The alternative IL-1β convertase, Casp8, which is both pro-inflammatory and pro-apoptotic
via activation of Casp3, was also synergistically co-activated with Casp1 and in RGC cells (Supplement
Figure S2). Finally, AIM2, known to drive pyroptotic death and sensitized by intracellular self-DNA [
64,
65], was specifically active in Muller glia, the cell type also implicated in the glaucomatous loss of RGCs [
66,
67]. Significantly, in control stOHT-challenged eyes, we did not detect any significant increase in inflammasome activity (release of cytokines), induction of Casp1, Casp8, and RGC damage, thus allowing us to conclude that the spiking pattern of OHT is a key trigger of retinal inflammation and injury. To summarize, our model provided new mechanistic insights into 1) very early post-spOHT pro-inflammatory events, and 2) mechanisms driving excessive neurotoxicity after repetitive vs. single, non-repetitive IOP spikes. Finally, mechanistic similarities between the acute spOHT and chronic OHT-induced glaucoma, including the inflammasome/Casp1-dependent RGC dysfunction and death, suggest that spOHT can be instrumental in studies of the earliest pathophysiological events in the OHT-challenged retina.
Mechanosensor signaling drives inflammasome activation. Most inflammasomes are induced by a combination of internal (loss of ionic and energetic homeostasis) and external signals via surface receptors. These are integrated into Signal 1 that facilitates transcriptional induction of inflammasome components via TLR, TNF, IL-1, C5a receptors, and alarmins, and Signal 2, which facilitates complex assembly in response to lysosomal damage and extracellular ATP and K
+ elevations via the Panx1-P2X4/7 activation [
68,
69,
70,
71,
72]. Abundant literature evidence supports a model, where spOHT-induced stimulation of mechanosensitive Panx1 [
33,
73,
74], Piezo-1[
75] and transient receptor potential (TRP) vanilloid TRPV1 and TRPV4 [
3,
76,
77] channels in neural cell plasma membrane initiates pro-inflammatory signaling [
66,
78,
79,
80]. A recent evidence that a highly mechanosensitive Piezo1 is an upstream interaction partner of Panx1 [
39,
81,
82] makes it feasible to suggest that a serial activation of the Piezo1–Panx1 by repetitive IOP spikes produces progressively higher levels of ATP release and inflammasome induction[
34]. In support of the key role of mechanosensory Panx1-inflammasome signaling in the retina, recent reports showed its pivotal role in both acute ischemia-induced and glaucomatous degeneration, where its activity strongly correlated with the induction of inflammasome, production of IL-1β [
33,
54]. The activity of TRP channels were linked to neuroinflammation via release of glial TNFα and infiltration of immune cells [
66,
78,
79,
80]. Consistently, our results showed that blockade of Panx1 provided suppression of inflammation and protection of RGC similar to the blockade of inflammasome activity by ablation of caspase-1 and GsdmD.
The neurotoxic pathways downstream of the inflammasome include pyroptotic and non-pyroptotic extracellular paracrine pathways, such as the GsdmD-NT pore-mediated release of IL-1β, several types of alarmins, miRNAs and ASC speck complexes [
83,
84,
85], as well as cleavage-activation of Casp1 and Casp8 substrates. Most of these pathways facilitate strong pro-inflammatory responses
via local glial activation, blood neutrophil attraction, tissue migration, [
86,
87,
88,
89,
90,
91], and spreading inflammation [
92]. In particular, activation of GsdmD-NT pores has previously been shown to mediates cytokine release that sustains and propagates proinflammatory signaling in the retina in a way similar to a “cytokine storm” during sepsis or viral infection[
93,
94].
Furthermore, similar to other gasdermins, GsdmD activation by the inflammasome was reported to facilitate mitochondrial damage [
95,
96] and pro-apoptotic signaling
via the release of mitochondrial DNA and cytochrome C [
61,
95,
97]. Since mitochondrial dysfunction and subsequent loss of RGC functionality and viability are the key pathological events in cerebral ischemia [
98] and glaucoma[
99,
100,
101], we obtained direct evidence of GsdmD involvement in dysfunction and death of RGCs using GsdmD
-/- retinas. Overall, since functional and structural damage to RGCs was prevented by the ablation of the
nlrp1, panx1 or gsdmD genes, we concluded that the NLRP1 and NLRP3 inflammasomes, expressed by RGCs and macrophages, are key mediators of neurotoxicity in the spOHT injury model.
Finally, the high level of neuroinflammation and cytokine release strongly correlated with infiltration of blood monocytes. The concurrent release of IL-1β, TNFα, and particularly MCP-1/CCL2 cytokines, observed in the vitreous and retina of the spOHT-challenged eyes, is also known to weaken the blood-retina-barrier and promote infiltration of blood-borne monocytes and leukocytes elsewhere [
45,
102,
103]. Such infiltration was evident at 48h post-OHT induction, as confirmed by abundant CD45
+ CD11b
+ cells in the retina and optic nerve (
Figure 9), and strongly correlated with unexpectedly high for such a mild injury, levels of cytokines. Importantly, monocyte and leukocyte infiltration is implicated in various retinal degenerations, including OHT-induced ischemia-reperfusion[
104] , AMD[
105] and glaucoma[
86,
106].
Pyroptosis vs. apoptosis.The activation of the inflammasome end-products caspase-1 and GsdmD in response to spOHT challenge suggested cell death via pyroptosis ([
107,
108]; however, we used Casp1/3/8-FLICA labeling and DEVD blockade of Casp3 activation to obtain evidence that the spOHT challenge triggered RGCs loss
via apoptosis. This conclusion was made despite our data indicating the key roles of Casp1 and GsdmD in RGC death, which are typically associated with pyroptotic death, as we reported earlier in the retinal ischemia model [
34]. The molecular mechanism for Casp1- and Casp8-dependent activation of Casp3 is well-characterized [
61,
62,
109]. Moreover, the most recent insights in non-canonical functions of GsdmD and Casp8 showed a non-pyroptotic GSDMD-ASC-Casp8 interaction as well as alternative cleavage of GsdmD by activated Casp3 into a non-pore forming fragment [
59]. Both of these events prevent pyroptosis and facilitate apoptosis in a heightened inflammatory environment. In support of this mechanism, we observed the co-activation of Casp1 and Casp3/7 specifically in RGCs in spOHT-challenged retinas (
Figure 6), which was blocked in either Casp1
-/- retinas or in WT retinas treated with the Casp1 inhibitor VX-765 both of which protected RGCs functionally and structurally (
Figure 5). It is reasonable to conclude that the neurotoxic activity of Casp1 is directly essential for endogenous OHT-induced RGC death since it is present in RGCs directly following injury, and its ablation caused a significant inhibition of IL-1β release into the vitreous, resulting in RGC protection [
34,
54]. At the same time, the activity of retinal Casp1 may also affect the viability of RGCs indirectly, since the bulk of Casp1-containing inflammasomes are not RGC-specific and are activated by astrocytes, microglia, and retina-infiltrating immune cells.
Author Contributions
For research articles with several authors, a short paragraph specifying their individual contributions must be provided. The following statements should be used “Conceptualization, V.S., and V.P.; methodology, V.S., M.S.; V.P., T-SC, and J.P.deRV data collection and validation, WA, GR, DPh, RW, HW, MB, G.S, SK, OJG, formal analysis, V.S.,, T-SC, PJdRV, V.P., S.K.; data curation, S.K., resources, PJdRV, S.K., writing—original draft preparation, V.S., M.S., and VP; writing—review and editing, V.S., M.S., PdeRV., RW, V.P., S.K.; funding acquisition, V.S., V.P. All authors have read and agreed to the published version of the manuscript.”
Figure 1.
Repetitive IOP spikes induce activation of inflammatory biomarkers, RGC dysfunction and loss. A. Experimental paradigm of the spiking OHT (spOHS) model and steady OHT (stOHT) control; B. qRT-PCR data on relative changes in transcript abundances of inflammasome pathway genes in retinas. Mean fold change vs. naïve ±SE, * P, 0.05, n=3-5. C. intravitreal IL1-β measured at 6, 12 and 24 hours after spOHT (red) and stOHT eyes (OD, blue). Mean ± SE, *P <0.05, n=5. Negative control: Casp1-/- eyes (Cs1KO). D. Quantification for HIF1a fluorescent labeling in retina wholemounts from eyes exposed to sham, spOHT expression sha or spOHT. Mean percentile change vs. sham SE, ** P< 0.05, n=5.
Figure 1.
Repetitive IOP spikes induce activation of inflammatory biomarkers, RGC dysfunction and loss. A. Experimental paradigm of the spiking OHT (spOHS) model and steady OHT (stOHT) control; B. qRT-PCR data on relative changes in transcript abundances of inflammasome pathway genes in retinas. Mean fold change vs. naïve ±SE, * P, 0.05, n=3-5. C. intravitreal IL1-β measured at 6, 12 and 24 hours after spOHT (red) and stOHT eyes (OD, blue). Mean ± SE, *P <0.05, n=5. Negative control: Casp1-/- eyes (Cs1KO). D. Quantification for HIF1a fluorescent labeling in retina wholemounts from eyes exposed to sham, spOHT expression sha or spOHT. Mean percentile change vs. sham SE, ** P< 0.05, n=5.
Figure 2.
PERG changes after single spOHT challenge are stable over time. Electro-physiological recordings of changes in PERG amplitude in the retinas exposed to either spOHT stress or stOHT (green bars) elevation for 7, 14 and 21 dpi. *P<0.05 . A. Representative PERG grand-average waveforms recorded in WT eyes prior to (baseline, blue) or 7d (red) after the challenge. B. Representative PERG grand-average waveforms recorded in WT eyes at 7d after either spOHT (red) or stOHT (yellow) challenges. C. Dynamics of PERG amplitude at 7, 14 and 21 dpi in C57Bl6/J eyes after spiking vs steady OHT challenges. Mean ± SE, *P <0.05 D. SpOHT-induced RGC loss at 1-3 weeks after spike or steady OHT. Mean ± SE, *P <0.05.
Figure 2.
PERG changes after single spOHT challenge are stable over time. Electro-physiological recordings of changes in PERG amplitude in the retinas exposed to either spOHT stress or stOHT (green bars) elevation for 7, 14 and 21 dpi. *P<0.05 . A. Representative PERG grand-average waveforms recorded in WT eyes prior to (baseline, blue) or 7d (red) after the challenge. B. Representative PERG grand-average waveforms recorded in WT eyes at 7d after either spOHT (red) or stOHT (yellow) challenges. C. Dynamics of PERG amplitude at 7, 14 and 21 dpi in C57Bl6/J eyes after spiking vs steady OHT challenges. Mean ± SE, *P <0.05 D. SpOHT-induced RGC loss at 1-3 weeks after spike or steady OHT. Mean ± SE, *P <0.05.
Figure 3.
Cytokine release into the vitreous strongly correlate with inflammasome activity. Cytokine levels in the vitreous of WT, NLRP1-/- , Casp1-/-, GsdmD-/- and Panx1-/- mice were measured simultaneously by multiplex ELISA at 24h after spOHT. Mean intravitreal concentration was measured as pg/ml (± SE) in a given transgenic mouse line and normalized by internal standards for every plate to allow between plate comparison. P<0.05, n=3-5.
Figure 3.
Cytokine release into the vitreous strongly correlate with inflammasome activity. Cytokine levels in the vitreous of WT, NLRP1-/- , Casp1-/-, GsdmD-/- and Panx1-/- mice were measured simultaneously by multiplex ELISA at 24h after spOHT. Mean intravitreal concentration was measured as pg/ml (± SE) in a given transgenic mouse line and normalized by internal standards for every plate to allow between plate comparison. P<0.05, n=3-5.
Figure 4.
Panx1 is essential for transcriptional activation of cytokines and inflammasome pathways in response to spOHT. Gene expression analysis was performed by RT-PCR in retinal samples collected 24 hrs after spOHT. Significant increases in il11b, tnfα, ccl2, casp1, nlrp3 and gsdmD transcripts found in WT mice (A, red) were blocked by PANX1 channel inactivation in Panx1-/- retinas (A, purple). Panx1 inactivation did not affect the upregulation of the gsdmD gene (B). *p<0.05, n=3-5.
Figure 4.
Panx1 is essential for transcriptional activation of cytokines and inflammasome pathways in response to spOHT. Gene expression analysis was performed by RT-PCR in retinal samples collected 24 hrs after spOHT. Significant increases in il11b, tnfα, ccl2, casp1, nlrp3 and gsdmD transcripts found in WT mice (A, red) were blocked by PANX1 channel inactivation in Panx1-/- retinas (A, purple). Panx1 inactivation did not affect the upregulation of the gsdmD gene (B). *p<0.05, n=3-5.
Figure 5.
PERG and RGC after spOHT injury with inflammasome knockout. A. significant PERGamp deficit was detected in WT and GsdmD-/- eyes at 1w after spOHT. No significant change in PERGamp was detected in spOHT-challenged eyes of the inflammasome-deficient strains NLRP1-/-, Casp1-/-, and Panx1-/- , in WT eyes pre-treated by Casp3 inhibitor DEVD and in control WT eye with stOHT challenge. B. RGC loss is significantly suppressed by inactivation of all key inflammasome proteins, the IL-1 release pore protein GsdmD, or caspase 3. The density of RBPMS+ RGCs was assayed by direct counting in wholemounts 7d after OHT challenge.
Figure 5.
PERG and RGC after spOHT injury with inflammasome knockout. A. significant PERGamp deficit was detected in WT and GsdmD-/- eyes at 1w after spOHT. No significant change in PERGamp was detected in spOHT-challenged eyes of the inflammasome-deficient strains NLRP1-/-, Casp1-/-, and Panx1-/- , in WT eyes pre-treated by Casp3 inhibitor DEVD and in control WT eye with stOHT challenge. B. RGC loss is significantly suppressed by inactivation of all key inflammasome proteins, the IL-1 release pore protein GsdmD, or caspase 3. The density of RBPMS+ RGCs was assayed by direct counting in wholemounts 7d after OHT challenge.
Figure 6.
Acute caspase activation in the GCL of spOHT-challenged retinas. A. Caspase-1 (red) and caspase-3/7 (green) activity in wholemount retinas was detected by an in vivo injection of FLICA substrate. Retinas were co-stained with GFAP (light grey, top panels) and RBPMS (light grey, bottom panels) white/merge to identify astrocytes and RGCs, respectively. White arrows indicate cells that co-activate caspases 1 and 3/7; yellow arrows-cells point at Casp3/7 activity only. D. Pre-treatment with VX765 casp1 blocked both Casp1 and Casp3/7 activity after spOHT. Size bar, 20 µm.
Figure 6.
Acute caspase activation in the GCL of spOHT-challenged retinas. A. Caspase-1 (red) and caspase-3/7 (green) activity in wholemount retinas was detected by an in vivo injection of FLICA substrate. Retinas were co-stained with GFAP (light grey, top panels) and RBPMS (light grey, bottom panels) white/merge to identify astrocytes and RGCs, respectively. White arrows indicate cells that co-activate caspases 1 and 3/7; yellow arrows-cells point at Casp3/7 activity only. D. Pre-treatment with VX765 casp1 blocked both Casp1 and Casp3/7 activity after spOHT. Size bar, 20 µm.
Figure 7.
Pro-inflammatory cytokines in the spOHT-challenged retina. Immunolabeling for IL-1β (blue) co-localized with neurons in the GCL and INL; TNFα-specific labelling (green) confined to GFAP+ astroglia (arrows), infiltrating monocytes (arrowheads) in the retina 48h after the rspOHT challenge. ASC-citrine specks (magenta) co-localized with the GFAP+ TNFα+ astrocytes and Muller glia (yellow arrows) in the inner retina, and with astrocytes in the optic nerve. In control stOHT-treated retinas (bottom panels) both TNFα and IL1β -specific labeling were reduced, no infiltrating monocytes were detected. Size bars in µm.
Figure 7.
Pro-inflammatory cytokines in the spOHT-challenged retina. Immunolabeling for IL-1β (blue) co-localized with neurons in the GCL and INL; TNFα-specific labelling (green) confined to GFAP+ astroglia (arrows), infiltrating monocytes (arrowheads) in the retina 48h after the rspOHT challenge. ASC-citrine specks (magenta) co-localized with the GFAP+ TNFα+ astrocytes and Muller glia (yellow arrows) in the inner retina, and with astrocytes in the optic nerve. In control stOHT-treated retinas (bottom panels) both TNFα and IL1β -specific labeling were reduced, no infiltrating monocytes were detected. Size bars in µm.
Figure 8.
Inflammasome sensor proteins in the spOHT-challenged inner retina. A. Immunolabeling specific for NLRP1 co-localized with TUJ1+ neurons, their axons, GFAP+ astrocytes (yellow arrowheads) in the GCL of both spOHT and sham-treated control retinas; in spOHT samples NLRP1 was also detected in infiltrating monocytes (yellow arrowheads). B. NLRP3 labeling co-localized with TUJ1+ neurons and infiltrating monocytes (yellow arrowheads) in the GCL of spOHT-challenged retinas; in sham controls it also labeled glial cells (white arrows). C. AIM2 sensor co-localized with GlutSyn+ Muller glia (white arrows) in both sham-treated controls and spOHT retinas, and showed an increased accumulation in the GCL region after spOHT challenge. Size bars, 20µm.
Figure 8.
Inflammasome sensor proteins in the spOHT-challenged inner retina. A. Immunolabeling specific for NLRP1 co-localized with TUJ1+ neurons, their axons, GFAP+ astrocytes (yellow arrowheads) in the GCL of both spOHT and sham-treated control retinas; in spOHT samples NLRP1 was also detected in infiltrating monocytes (yellow arrowheads). B. NLRP3 labeling co-localized with TUJ1+ neurons and infiltrating monocytes (yellow arrowheads) in the GCL of spOHT-challenged retinas; in sham controls it also labeled glial cells (white arrows). C. AIM2 sensor co-localized with GlutSyn+ Muller glia (white arrows) in both sham-treated controls and spOHT retinas, and showed an increased accumulation in the GCL region after spOHT challenge. Size bars, 20µm.
Figure 9.
Monocyte/macrophages infiltration into the inner retina and optic nerves after spOHT challenge. A,B. Representative micrographs for cells expressing CD45 (red) and CD11b (green) markers in rounded monocyte/macrophage cells that were abundant at the inner retinal surface, in the GCL, INL and in the optic nerves of the spOHT-challenged eyes. Co-staining with GFAP (light grey, top panels) identified astrocytes. C,D. Control immunostaining in stdyOHT-challenged optic nerves showed no CD45+ monocytes in both retina and optic nerves. Small punctate CD45+CD11b+ cells are ramified microglia. Bright green puncta represent citrin-labelled ASC-specks of mature inflammasomes that are abundant in the optic nerve astrocytes and in infiltrating cells (yellow arrows).
Figure 9.
Monocyte/macrophages infiltration into the inner retina and optic nerves after spOHT challenge. A,B. Representative micrographs for cells expressing CD45 (red) and CD11b (green) markers in rounded monocyte/macrophage cells that were abundant at the inner retinal surface, in the GCL, INL and in the optic nerves of the spOHT-challenged eyes. Co-staining with GFAP (light grey, top panels) identified astrocytes. C,D. Control immunostaining in stdyOHT-challenged optic nerves showed no CD45+ monocytes in both retina and optic nerves. Small punctate CD45+CD11b+ cells are ramified microglia. Bright green puncta represent citrin-labelled ASC-specks of mature inflammasomes that are abundant in the optic nerve astrocytes and in infiltrating cells (yellow arrows).
Figure 10.
Inflammasome activation in the Ad5-MYOC-indiced chronic OHT/glaucoma model. Dynamics of initial IOP changes (A) was similar in WT, Casp1- and NLRP1- strains with relatively higher levels of IOP elevation in knockouts at 2-4 wks. An increase in IOP in experimental eyes temporally correlated with RGC dysfunction detected as with a ~50% decline in PERGamp (B)in WT but not in Casp1- and NLRP1- mice at 2 wks post-induction. C. IL-1β release was detectable 3-8 wk post-OHT induction, peaking at 5wks at ~60% of that in positive control eyes with injection of ATP or ischemia-reperfusion (IR) injury. D. RGC density changes in experimental eyes vs eyes from naïve controls showed 32% and 19.1% loss in WT center (/C) and periphery (/P). The loss was significantly less in Casp1- and Nlrp1- retinas at 7dpi after spOHT challenge, *P<0.05 (E-F). ASC-speck labeling (green specks/filaments) revealed mature inflammasome complexes co-localizing with RBPMS+(red) RGCs (yellow arrows), GFAP+ (grey) astrocytes (white arrows) and blood vessels (unstained dark grey structures, blue “BV”) G. Quantification of ASC-citrin+ cell types in retinal wholemounts at 6 and 8wks post-induction indicated predominant localization to RGCs and astrocytes, *P<0.05.
Figure 10.
Inflammasome activation in the Ad5-MYOC-indiced chronic OHT/glaucoma model. Dynamics of initial IOP changes (A) was similar in WT, Casp1- and NLRP1- strains with relatively higher levels of IOP elevation in knockouts at 2-4 wks. An increase in IOP in experimental eyes temporally correlated with RGC dysfunction detected as with a ~50% decline in PERGamp (B)in WT but not in Casp1- and NLRP1- mice at 2 wks post-induction. C. IL-1β release was detectable 3-8 wk post-OHT induction, peaking at 5wks at ~60% of that in positive control eyes with injection of ATP or ischemia-reperfusion (IR) injury. D. RGC density changes in experimental eyes vs eyes from naïve controls showed 32% and 19.1% loss in WT center (/C) and periphery (/P). The loss was significantly less in Casp1- and Nlrp1- retinas at 7dpi after spOHT challenge, *P<0.05 (E-F). ASC-speck labeling (green specks/filaments) revealed mature inflammasome complexes co-localizing with RBPMS+(red) RGCs (yellow arrows), GFAP+ (grey) astrocytes (white arrows) and blood vessels (unstained dark grey structures, blue “BV”) G. Quantification of ASC-citrin+ cell types in retinal wholemounts at 6 and 8wks post-induction indicated predominant localization to RGCs and astrocytes, *P<0.05.