Abstract
The purpose of this study was to investigate how Sphingosine-1-phosphate (S1P) signaling regulates glial phenotype, neuroprotection, and reprogramming of Müller glia (MG) into neurogenic MG-derived progenitor cells (MGPCs) in the adult male and female mouse retina. We found that S1P-related genes were dynamically regulated following retinal damage. S1pr1 (S1P receptor 1) and Sphk1 (sphingosine kinase 1) are expressed at low levels by resting MG and are rapidly upregulated following acute damage. Overexpression of the neurogenic bHLH transcription factor Ascl1 in MG downregulates S1pr1, and inhibition of Sphk1 and S1pr1/3 enhances Ascl1-driven differentiation of bipolar-like cells. Treatments that activate S1pr1 or increase retinal levels of S1P initiate proinflammatory NFκB signaling in MG, whereas treatments that inhibit S1pr1 or decreased levels of S1P suppress NFκB signaling in MG. Conditional knock-out (cKO) of S1pr1 in MG, but not Sphk1, enhances the accumulation of immune cells in damaged retinas. cKO of S1pr1 promotes the survival of ganglion cells, whereas cKO of Sphk1 promotes the survival amacrine cells in damaged retinas. Consistent with these findings, pharmacological treatments that inhibit S1P receptors or inhibit Sphk1 have protective effects upon inner retinal neurons. We conclude that the S1P signaling pathway is activated in MG after damage and this pathway restricts the accumulation of immune cells, impairs neuron survival, and suppresses the reprogramming of MG into neurogenic progenitors in the adult mouse retina.
- immune cell recruitment
- MG-derived progenitor cells (MGPCs)
- Müller glia
- NFκB signaling
- retinal injury
- sphingosine-1-phosphate (S1P)
Significance Statement
Understanding the mechanisms of retinal neuron survival and regeneration is fundamental for the development of therapeutic strategies of retinal repair. Here, we show that Sphingosine-1-phosphate signaling kick-starts the glial proinflammatory response, restricts immune cell recruitment, and exacerbates neuron cell death after acute retinal injury. Importantly, blocking sphingosine-1-phosphate activity enhances Ascl1-driven neurogenesis in the mouse retina, highlighting a potential therapeutic target for retinal regeneration.
Introduction
Neuronal damage and inflammation impact the ability of Müller glia (MG) to become proliferating, neurogenic progenitor-like cells in different vertebrate models of retinal regeneration. For example, Nuclear Factor Kappa B (NFκB) cell signaling pathway is a key mediator of inflammation that induces the reactivity of MG, acts to restore glial quiescence, and suppress the neurogenic potential of MG-derived progenitors in the mouse retina (Hoang et al., 2020; Palazzo et al., 2022a). NFκB is known to coordinate with Sphingosine-1-phosphate (S1P) signaling to regulate different cellular processes including inflammation (Blom et al., 2010; Zheng et al., 2019; Pérez-Jeldres et al., 2021). S1P is an aliphatic amino alcohol that is synthesized by sphingosine kinases (SPHK1 and SPHK2) and degraded by a lyase (SGPL1; Fig. 3a). S1P is exported from cells by transporters (MFSD2A and SPNS2) or hydrolyzed back to sphingosine by a phosphatase (SGPP1; Fig. 3a). Secreted S1P is chaperoned by HDL-anchored ApoM or albumin, and binds to and activates G-protein-coupled receptors (S1PR1–S1PR5) to elicit a wide array of functions across different cell types (Fig. 3a; Murata et al., 2000; Obinata and Hla, 2019). S1P signaling is known to mediate inflammatory responses, cellular proliferation, cell survival, and angiogenesis in lymphocytes and endothelial cells through activation of different second messenger systems including MAPK, PI3K/mTor, and Jak/Stat, and cross-talk with inflammatory pathways such as NFκB (Fig. 3a; Obinata and Hla, 2019). In the developing retina, S1P signaling is required for vascular maturation, progenitor cell cycle exit, and axon guidance (Simón et al., 2019). S1P can also have negative effects on the adult retina which include migration of MG, neovascularization, and inflammation associated with proliferative retinopathies and aging (Shiwani et al., 2021). Many of these studies applied clinically relevant S1P analogs, which target S1P receptors and ceramide pathway components nonspecifically, thereby complicating the interpretation of findings. Additionally, most of these studies on retinal cells were conducted in vitro and need to be followed up by in vivo analyses to identify the coordinated impact of S1P signaling on the many different types of retinal cells.
We recently identified the effects of S1P signaling in the avian retina, wherein treatments that inhibited S1pr1 or decreased levels of S1P enhanced MGPC formation and neurogenesis in damaged retinas (Taylor et al., 2024a). S1P-related genes are highly expressed by neurons and glia, and levels of expression are dynamically regulated following damage to the chick retina. S1PR1 was highly expressed by resting MG and is rapidly downregulated following damage. This pattern of expression is similar to that seen in zebrafish and human retinas. Further, ablation of microglia from damaged retinas, wherein the formation of MGPCs is blocked (Fischer et al., 2014), has a significant impact upon expression patterns of S1P-related genes in MG; this may, in part, be mediated by TGFβ/Smad3 signaling which maintains high levels of S1PR1 expression in resting MG (Taylor et al., 2024a). Thus, in the chick retina, S1P signaling is dynamically regulated in MG to suppress MGPC formation, and activation of S1P signaling depends, in part, on signals produced by activated microglia.
The neuroprotective effects of S1P receptor-targeting drugs have been reported previously (Nakamura et al., 2021; Basavarajappa et al., 2023), but the mechanisms underlying neuroprotection are not well understood. Further, S1P signaling has not been targeted in a tissue-specific manner to assess neuroprotection and inflammation. Accordingly, the purpose of this study was to better understand how S1P signaling impacts inflammation, glial reactivity, neuronal survival, and reprogramming of MG into neurogenic MGPCs in the mouse retina.
Materials and Methods
Animals
The animal use approved in these experiments was in accordance with the guidelines established by the National Institutes of Health and the Institutional Animal Care and Use Committee at Ohio State University. Both male and female mice were used. Mice were housed under a 12 hour (h) light/dark cycle and received water and chow ad libitum. NFκB-eGFP reporter mice, which have eGFP expression driven by a chimeric promoter containing three HIV- NFκB-cis elements (Magness et al., 2004), and Ikkbfl/fl mice, with insertion of Cre recombinase binding sites (LoxP) into the intronic regions flanking exon 3 of the wild-type Ikkb gene (Li et al., 2003), were kindly provided by Dr. Denis Guttridge's laboratory at The Ohio State University. We crossed Sphk1fl/fl and S1pr1fl/fl mice (provided by Dr. Timothy Hla; Harvard Medical School) onto Rlbp1-CreERT;R26-stop-flox-CAG-tdTomato mice (provided by Dr. Ed Levine; Vanderbilt University), wherein Cre-mediated recombination occurs in a tamoxifen-dependent manner specifically in MG under the control of the retinaldehyde binding protein 1 (Rlbp1) promoter, herein referred to as Rlbp1-CreERT:S1pr1fl/fl and Rlbp1-CreERT:Sphk1fl/fl. The use of Ascl1-overexpressing mice (Glast-CreER:LNL-tTA:tetO-mAscl1-ires-GFP; provided by Dr. Thomas Reh; University of Washington) was as previously described (Ueki et al., 2015; Palazzo et al., 2022a).
Ablation of microglia
Mice were provided control or PLX5622 (1200 ppm; Research Diets #D1100404i) diet ad libitum for 2 weeks to deplete microglia prior to experimental paradigms. We and other groups have validated the efficacy of this treatment paradigm to deplete microglia in the mouse retina (Todd et al., 2020; Palazzo et al., 2022a).
Intraocular injections
Mice were anesthetized via inhalation of 2.5% isoflurane in oxygen and intraocular injections performed as described previously (Hamilton #7803-05 10 MM 12DEG; Palazzo et al., 2022a). For all experiments, the vitreous chamber of the right eyes of mice were injected with the experimental compound, and the contralateral left eyes were injected with a vehicle control. Injections of 100 mM N-methyl-D-aspartate (NMDA) in sterile saline were administered at a volume of 1 ul. Compounds included in this study are described in Table 1. For adult mice, the average eye weight is 0.02 g. Doses were estimated from common mg/kg doses reported for oral administration. Injection paradigms are included in each figure. Intraperitoneal injections of tamoxifen (Sigma T5648; 1.5 mg/100 µl corn oil per dose) were performed for 4 consecutive days to induce ER-Cre activity.
Pharmacological compounds
Fixation, sectioning, and immunocytochemistry
Retinas were fixed, sectioned, and immunolabeled as described previously (Fischer et al., 1998). To identify MG, we labeled sections for Sox2, which is known to label the nuclei or MG in the INL and the nuclei of astrocytes at the vitread surface of the retina (Fischer et al., 2010). To identify CNS microglia/macrophages and peripheral monocyte-derived macrophages, we labeled sections for Iba1 and CD45, as described previously (Palazzo et al., 2022a). None of the observed labeling was due to nonspecific labeling of secondary antibodies or autofluorescence because sections labeled with secondary antibodies alone were devoid of fluorescence. Primary antibodies used in this study are described in Table 2. Secondary antibodies included donkey-anti-goat-Alexa488/594/647 (Life Technologies A11055; A11058; A21447), donkey-anti-rabbit-Alexa488/594 (Life Technologies A21206; A21207), and goat-anti-mouse-Alexa488/568 (Life Technologies A11001; A-11004) diluted to 1:1,000 in PBS plus 0.2% Triton X-100. Nuclear staining was accomplished using DRAQ5 (Thermo 62251) or DAPI (Sigma D9542).
Antibodies
Labeling for EdU
Regular drinking water was removed 24 h prior to NMDA injections and replaced with sterile water containing 5-ethynyl-2′-deoxyuridine (EdU; Sigma; 50 mg/100 ml dH2O) which was replaced every third day. Mice were maintained on EdU water until the fourth day after TSA treatment. For the detection of nuclei that incorporated EdU, immunolabeled sections were fixed in 4% formaldehyde in 0.1 M PBS pH 7.4 for 5 min at room temperature. Samples were washed for 5 min with PBS, permeabilized with 0.5% Triton X-100 in PBS for 1 min at room temperature, and washed twice for 5 min in PBS. Sections were incubated for 30 min at room temperature in a buffer consisting of 100 mM Tris, 8 mM CuSO4, and 100 mM ascorbic acid in dH2O. Alexa Fluor 647 Azide (Thermo Fisher Scientific #A10277) was added to the buffer at a 11,000 dilution.
Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)
The TUNEL method was used to identify dying cells with fragmented DNA. An in situ Cell Death Detection kit from Roche (Fluorescein, #11684795910) was used according to manufacturer instructions.
Fluorescent in situ hybridization
Standard procedures were used for fluorescent in situ hybridization (FISH), as described previously (Campbell et al., 2021b; Taylor et al., 2024b). In short, retinas from P9 eyes were fixed for 4 h at room temperature in 4% paraformaldehyde buffered in 0.1 M dibasic sodium phosphate, washed in PTW (PBS + 0.2% Tween), and incubated in 30% sucrose at 4°C overnight. The retinas were embedded in OCT-compound and cryo-sectioned at 12 microns. Tissue sections were processed for in situ hybridization with a split-initiator probe pair (Molecular Instruments) according to the manufacturer protocol for fresh/fixed frozen tissues. For slides in which immunocytochemistry was conducted with FISH, primary antibodies were incubated overnight with the hairpin amplification buffer solution, and secondary antibodies were incubated for 1 h the next day. Slides were mounted with glycerol and glass coverslips.
Photography, immunofluorescence measurements, and statistics
Wide-field photomicroscopy was performed using a Leica DM5000B microscope equipped with epifluorescence and Leica DC500 digital camera. Confocal images were obtained using a Leica SP8 imaging system at the Department of Neuroscience Imaging Facility at the Ohio State University. Images were optimized for color, brightness and contrast, multiple channels overlaid, and figures constructed using Adobe Photoshop. Images in Figures 4⇓⇓–7 were assigned different colors or set to grayscale using Photoshop or ImageJ to distinguish and enhance the visualization of EdU+, Sox2+, or DAPI+ cells. Cell counts were performed on representative images. To avoid the possibility of region-specific differences within the retina, cell counts were consistently made from the same region of retina for each data set.
Counts of FISH puncta per cell were made as follows. The number of FISH puncta per cell was determined for fixed regions of interest (ROIs) of the INL by counting the total number of FISH puncta and dividing by the total number of Sox2+ MG per ROI. The number of FISH puncta per transgenic cell was determined by counting the number of FISH puncta within GFP+ cells divided by the number of Sox2+ GFP+ MG within the ROI. The number of FISH puncta per wild-type (WT) cell was determined by counting the number of FISH puncta outside of GFP+ cells and dividing by the number of Sox2+ GFP− MG within the ROI. ImageJ was used to establish threshold fluorescence intensity levels to capture GFP+ cells and FISH puncta, and for water-shedding and counting numbers of FISH puncta.
Single-cell RNA-seq
We analyzed scRNA-seq libraries that were generated and characterized previously (Hoang et al., 2020; Palazzo et al., 2022a; Li et al., 2024). For WT retinas, dissociated retinal cells were loaded onto the 10× Chromium Cell Controller with Chromium 3′ V2 reagents. Using Seurat toolkits (Powers and Satija, 2015; Satija et al., 2015), Uniform Manifold Approximation and Projection (UMAP) for dimensional reduction plots were generated from nine separate cDNA libraries, including two replicates of control undamaged retinas, and retinas at different times after NMDA treatment (Hoang et al., 2020). In the IKK cKO whole retina library, dissociated cells were loaded onto the 10× Chromium Cell Controller with Chromium 3′ V3 reagents. Using Seurat toolkits, UMAP for dimensional reduction plots were generated from aggregated cDNA libraries, including two replicates of undamaged × WT, damaged × WT, undamaged × IKK cKO, and damaged × IKK cKO retinas (Palazzo et al., 2022a). In both groups of libraries, Seurat was used to construct gene lists for differentially expressed genes (DEGs), violin/scatter plots, and dot plots. Genes that were used to identify different types of retinal cells included the following: (1) Müller glia: Glul, Vim, Scl1a3, Rlbp1; (2) MGPCs: PCNA, CDK1, TOP2A, ASCL1; (3) microglia: C1qa, C1qb, Ccl4, Csf1r; (4) ganglion cells: Thy1, Pou4f2, Rbpms2, Nefl; (5) amacrine cells: Gad67, Calb2, Tfap2a; (6) horizontal cells: Prox1, Calb2, Ntrk1; (7) bipolar cells: Vsx1, Otx2, Grik1, Gabra1; (7) cone photoreceptors: Gnat2, Gnb3, Opn1lw; (8) rod photoreceptors: Rho, Nr2e3, Arr3; (9) astrocytes: Pax2, S100b, Gja1; and (10) endothelial cells: Pecam1, Cdh5, Tie1.
Statistical analyses
Data are shown as mean ± standard deviation, and numbers of biological replicates are shown in dot plots; each dot represents one biological replicate, and blue lines connect control and treated eyes from the same individual. Where significance of difference was determined between two treatment groups accounting for interindividual variability (means of treated-control values), we performed a two-tailed, paired t test. Where significance of difference was determined between two treatment groups, we performed a two-tailed, unpaired t test. Significance of difference between multiple treatment groups was determined using one-way ANOVA followed by Sidak’s correction. GraphPad Prism 10 was used for statistical analyses and generation of histograms and bar graphs. For single-cell RNA-sequencing data, significance of difference in violin/scatter plots was determined using a Wilcoxon rank sum or Poisson tests with Bonferroni’s correction.
Results
Expression of S1P-related genes in the retina
We began by analyzing patterns of expression of S1P-related genes in scRNA-seq libraries that were established from control retinas and damaged retinas at different times after NMDA treatment; these libraries were generated and analyzed previously (Todd et al., 2019; Hoang et al., 2020; Campbell et al., 2021a, b). UMAP plots revealed discrete clusters of all major retinal cell types (Fig. 1a,b). Clusters of cells were labeled based on distinct patterns of gene expression as described in the Materials and Methods. Neuronal cells from control and damaged retinas were clustered together regardless of time after NMDA treatment (Fig. 1a,b). In contrast, resting MG, including cells from 48 and 72 h after NMDA, and activated MG from 3, 6, 12, and 24 h after NMDA were spatially separated by UMAP embedding (Fig. 1a–c). Activated MG were identified, in part, based on downregulation of genes such as Slc1a3 and upregulation of genes such as Vim or Nes (Fig. 1c). We examined patterns of expression and changes in expression levels of S1P-related genes (Fig. 1d–i). S1pr4, S1pr5, Sgpl1, and Sphk2 were either not detected at significant levels or detected in very few cells in the retina (data not shown). S1pr2 was expressed by relatively few retinal cells, but S1pr1, S1pr3, and Sphk1 were highly expressed by activated MG (Fig. 1d,e). In addition, S1pr1 was highly expressed by endothelial cells (Fig. 1d). We did not perform detailed analyses of S1P-related genes in endothelial cells because there were <350 cells captured in these libraries (Fig. 1b). S1pr1 and Sphk1 appeared to be robustly upregulated in activated MG at 3 and 6 h after NMDA treatment, but downregulated in MG thereafter (Fig. 1d,e). To perform a detailed analysis of S1P-related genes in MG, we bioinformatically isolated and re-embedded nearly 6,200 MG into a UMAP dimensional reduction (Fig. 1f,g). These MG formed a distinct trajectory of cells with resting MG from control retinas clustered to one side, ++activated MG from 3 h after NMDA-treatment clustered to the other side of the plot, and MG from later times after NMDA bridging the middle of the trajectory (Fig. 1f,g). This analysis revealed that levels of S1pr1 were relatively low in resting MG and levels were significantly upregulated in ++activated MG at 3 h after NMDA and further upregulated in +activated MG at 6 h after NMDA (Fig. 1h,i). Levels of S1pr1 were significantly downregulated in activated MG and returning to resting MG from retinas at 12–72 h after NMDA (Fig. 1i). Levels of S1pr2 were very low in MG, and levels of S1pr3 were not significantly changed, but the percentage of MG that expressed S1pr3 was greatest in MG that were returning to a resting phenotype from retinas 24–72 h after NMDA (Fig. 1h,i). By comparison, Sphk1 was low in resting MG and highly upregulated in ++activated (3 h after NMDA) and +activated MG (6 h after NMDA), but significantly downregulated thereafter (Fig. 1h,i). Collectively, these findings suggest that after acute injury, levels of Sphk1 (S1P synthesis) are very rapidly and transiently upregulated and the upregulation in S1P synthesis is followed by a rapid and transient upregulation in autocrine signaling through S1pr1 in MG.
Patterns of expression of S1P-related genes in damaged mouse retinas. scRNA-seq was used to identify patterns of expression of S1P-related factors among retinal cells with the data presented in UMAP a–h or violin plots i. Aggregate scRNA-seq libraries were generated for cells from control retinas and retinas 3, 6, 12, 24, 36, 48, and 72 h after NMDA treatment. MG were bioinformatically isolated and analyzed from the large aggregate library (f–i). UMAP-ordered cells formed distinct clusters of neuronal cells, resting MG, activated MG, and returning to resting MG based on distinct patterns of gene expression (c). UMAP heatmap plots illustrate patterns and levels of expression of S1P receptors S1pr1, S1pr2, S1pr3, and S1P metabolism gene Sphk1 (h). Violin plots illustrate relative levels of expression in clusters of MG, activated MG, and returning to resting MG (i). Significance of difference was determined by using a Wilcox rank sum with Bonferroni’s correction (supplemental Table 1). Abbreviations: MG, Müller glia; NMDA, N-methyl-D-aspartate; UMAP, Uniform Manifold Approximation and Projection.
We next probed patterns of expression of S1P-related genes in a large integrated scRNA-seq database of mouse retinal cells from different postnatal ages ranging from 2 to 25 weeks; this database has been generated and described previously (Li et al., 2024). This aggregated database contained more than 330,000 cells which form numerous distinct clusters of retinal cells in UMAP plots (Fig. S1a). Glial cells formed distinct clusters based on patterns of expression of cell-distinguishing markers (see Materials and Methods; Fig. S1b). Sphk1 was detected in astrocytes, MG and a few clusters of amacrine cells (Fig. 1c). By comparison, S1pr1 and S1pr3 were detected at high levels in MG (Fig. S1d,e). Additionally, S1pr1 was detected in endothelial cells, and S1pr3 was detected at high levels in pericytes and at lower levels in endothelial cells and astrocytes (Fig. S1d,e). Sgpl1 was expressed at high levels in most microglia and in cells scattered across all different major cell types (Fig. S1f).
We next bioinformatically isolated the MG to perform more detailed analyses of S1P-related genes. We renormalized and re-established principal components for UMAP embedding, filtered clusters with abnormally high features/UMI per cell, filtered contamination from astrocytes (high Pax2 and S100b) and bipolar cells (high Lhx4, Otx2, Scgn). These processes produced distinct clusters of MG; however, these cells were segregated by platform, not developmental stage (Fig. S1g,h). Relative levels of S1pr1 and S1pr3 were significantly increased in older mice (16 and 25 weeks of age), whereas levels of Sphk1 were significantly higher in young mice (2 weeks of age; Fig. S1i,j; Supplemental Table 1). The MG from animals at 8 weeks of postnatal development had elevated levels of S1pr3; however, these cells may have been reactive with significantly elevated levels of Tgfb2 and Klf6 (Fig. S1i,j; Table S1), which is characteristic of activated MG that are returning toward a resting phenotype (Hoang et al., 2020; Palazzo et al., 2022a).
Validation of patterns of expression of S1pr1 and Sphk1
To validate findings from scRNA-seq libraries, we applied different immunolabeling strategies using different antibodies to S1pr1 and Sphk1 (Table 1). However, none of these antibodies produced plausible patterns of labeling regardless of different antigen retrieval strategies including weak fixation, methanol washes, sodium citrate washes, exclusion of detergent, or applying different types of detergents (data not shown). Thus, we applied fluorescent in situ hybridization (FISH) strategies with probes for S1pr1 and Sphk1. We did not probe for S1pr2 or S1pr3 because the scRNA-seq data indicated very low levels of expression in the retina. In undamaged retinas, S1pr1 puncta were associated with Sox2-labeled MG nuclei (Fig. 2a). S1pr1 FISH signal was also detected in putative endothelial cells associated with autofluorescent red blood cells that were scattered across retina (Fig. 2a). Consistent with findings from scRNA-seq, we found an increase in S1pr1 puncta associated with Sox2-labeled MG nuclei at 4 h after NMDA treatment (Fig. 2a).
Fluorescence in situ hybridization (FISH) for S1PR1 and SPHK1. Glast-CreER:LNL-tTA:tetO-mAscl1-ires-GFP mice received intraperitoneal injections of tamoxifen or corn oil once daily for 4 consecutive days. Eyes received intravitreal injections with NMDA or saline, and retinas were collected 4 h after NMDA treatment. Retinal sections were labeled with antibodies to Sox2 (cyan), GFP (green), or FISH probes to S1pr1 (red puncta; a, c) or Sphk1 (red puncta; b). Regions of interest (yellow boxes) are enlarged 1.8-fold and displayed in adjacent channels. Solid arrows indicate Sox2+/GFPlow MG nuclei, and hollow arrows indicate Sox2+/GFPhigh MG nuclei. The area occupied by Sox2+ (b) or GFP+ cells (e), was selected to distinguish FISH puncta labeling within these regions. Small double arrows indicate endothelial cells. Scale bars: a, b, c, and e represent 50 µm. Histograms represent the mean (bar ± SD) number of FISH puncta per MG and each dot represents one biological replicate (d; n = 6 or 7). Significance of difference (p values) was determined by using a paired t test. Abbreviations: ONL, outer nuclear layer; INL, inner nuclear layer; IPL, inner plexiform layer; GCL, ganglion cell layer; NMDA, N-methyl-D-aspartate; ns, not significant.
In undamaged retinas, there were very few Sphk1 FISH puncta scattered randomly across the retina (Fig. 2b). At 4 h after NMDA treatment, there were numerous Sphk1 puncta scattered across the retina, and some of these puncta were associated with Sox2+ MG nuclei (Fig. 2b). According to scRNA-seq data, levels of Sphk1 are increased in MG at 3–6 h after NMDA, whereas the scattered distribution of Sphk1 FISH puncta did not reveal MG-specific upregulation of transcripts.
To investigate whether S1P signaling through S1pr1 is altered during the reprogramming of MG into progenitor-like cells, we applied FISH probes to retinas where the MG have been forced to express the neurogenic basic Helix-Loop-Helix (bHLH) transcription factor Ascl1 (Glast-CreER:LNL-tTA:tetO-mAscl1-ires-GFP; Jorstad et al., 2017). In adult mouse retinas, inducible forced expression of Ascl1 in MG, combined with NMDA-induced damage and HDAC inhibitor (trichostatin A), stimulates the reprogramming of MG into bipolar-like cells that integrate into circuitry and respond to visual stimuli (Jorstad et al., 2017). Ascl1-overexpressing (Ascl1-OE) MG were identified by GFP expression, which is linked to the Ascl1 transgene via an internal ribosomal entry sequence (IRES; Fig. 2). Counts of S1pr1 FISH puncta associated with Ascl1-OE MG (Sox2+/GFP+ cells) and of puncta associated with neighboring WT MG (Sox2+/GFP− cells) within the same section were measured. In undamaged retinas, numbers of S1pr1 puncta in Ascl1-OE MG were not significantly different from WT MG (Fig. 2c–e). In damaged retinas, there were significantly more S1pr1 FISH puncta per MG in NMDA-damaged retinas compared with numbers seen in saline-treated retinas (Fig. 2c–e), consistent with scRNA-seq data. In damaged retinas, with Ascl1-OE MG, numbers of S1pr1 FISH puncta were significantly reduced in Ascl1-OE MG compared with numbers seen in WT MG (Fig. 2c–e). Collectively, these findings indicate that S1pr1 and Sphk1 transcripts are upregulated in response to NMDA damage but downregulated in response to forced expression of Ascl1 in damaged retinas.
S1P signaling and reprogramming of Ascl1-over expressing MG
To investigate how S1P signaling influences Ascl1-mediated neurogenesis, we applied small molecule inhibitors to Sphk1 (PF543) and S1pr1/3 (VPC23019) to the retinas of Ascl1-OE mice. All pharmacological compounds used in this study are described in Table 3. Retinas were treated with NMDA and inhibitors, inhibitors at 1 day post-injury (DPI), TSA at 2 DPI, and harvested at 16 DPI (Fig. 3). Application of PF543 or VPC23019 alone suppressed glial differentiation but did not significantly influence numbers of differentiating neurons (data not shown). Combined application of Sphk1 + S1pr1/3 inhibitors significantly decreased the percent of GFP+/Sox2+ cells (Fig. 3a–c), indicating that the S1P pathway promotes glial dedifferentiation in Ascl1-OE MG. By comparison, application of Sphk1 + S1pr1/3 inhibitors significantly increased the percent of GFP+/Otx2+ and GFP+/Lhx4+ cells (Fig. 3d–f,g,j,l), indicating that S1P pathway suppresses neuronal differentiation in Ascl1-OE MGPCs. Sphk1 + S1pr1/3 inhibitors had no significant effect upon total numbers of proliferating EdU-positive cells or numbers of EdU+/GFP+ cells (Fig. 3g,h). By comparison, there was no significant effects of S1P pathway inhibition on numbers of GFP+ cells colabeled with the cone bipolar cell marker Scgn (Fig. 3i,k), suggesting that the S1P pathway does not promote differentiation toward this type of bipolar cell. Collectively, these findings indicate that inhibition of S1pr1/3 and Sphk1 in Ascl1-OE MG enhances the differentiation of bipolar-like cells, while glial phenotype was suppressed.
Inhibition of S1P signaling enhances Ascl1-mediated reprogramming of MG into neurogenic progenitor cells. Schematic summary of S1P signaling and sites of action for different small molecule agonists and antagonists a, Glast-CreER:LNL-tTA:tetO-mAscl1-ires-GFP mice were intraperitoneally injected with tamoxifen or corn oil once daily for 4 d. Eyes were intravitreally injected NMDA and either a vehicle control or inhibitors to Sphk1 (PF543) and S1pr1/3 (VPC23019). At 24HPI, eyes were treated with vehicle or inhibitors. Eyes were treated with TSA at 48HPI. Retinas were collected 24 d from the first tamoxifen injection. Retinal sections were labeled with DAPI (blue, b, d) or EdU (blue, e, h) and antibodies to GFP (green) and Sox2 (red, b, d), Otx2 (red, e, h), Scgn (red, i), or Lhx4 (red, j). Regions of interest (yellow boxes) are enlarged 1.5 (b, d) or 2-fold (e, h, i, j) and displayed in separate panels. Solid arrows indicate GFP+/Sox2high cells in b and d, and hollow arrowheads indicate GFP+/Sox2low cells. In e and h, solid arrows indicate GFP+/Otx2+/EdU+ cells. Scale bars: b and e represent 50 µm. Histograms represent the mean (bar ± SD) where each dot represents one biological replicate (n = 8), and blue lines connect control and treated eyes from the same individual (c, g, f). Significance of difference (p values) was determined by using a paired t test. Abbreviations: ONL, outer nuclear layer; INL, inner nuclear layer; IPL, inner plexiform layer; GCL, ganglion cell layer; NMDA, N-methyl-D-aspartate; ns, not significant.
Pharmacological compounds, cKO, and effects
Cross-regulation of S1P/S1pr1 and NFκB signaling in MG
S1P receptors differ by their affinity to S1P and by their coupling to different G-proteins, thereby resulting in distinct cellular responses and physiological outcomes. S1pr1 has been shown to predominantly couple to Gi, activating Phospholipase C (PLC), MAPK/Erk, and PI3K/AKT pathways (Pyne and Pyne, 2017), and secondarily impact NFκB signaling (Sun et al., 2024). We have recently reported that NFκB signaling is rapidly, transiently, and selectively activated in MG in damaged mouse retinas, and this activation is mediated by proinflammatory cytokines produced by reactive microglia (Palazzo et al., 2022a, 2023). Additionally, NFκB signaling promotes glial fate in MGPCs of chicks and Ascl1-OE mice (Palazzo et al., 2020, 2022a). S1P signaling is known to activate inflammatory responses via NFκB in many cell types, including endothelial cells, cancer cells, and macrophages (Liang et al., 2013; Campos et al., 2016; Xiao et al., 2018; Zheng et al., 2019; Del Gaudio et al., 2020). Thus, we investigated whether pharmacological activation or inhibition of S1P-related receptors and enzymes influenced NFκB activation and expression of other cell signaling biomarkers in normal and damaged retinas.
Eyes were treated with a single intraocular injection of S1P or S1pr1 agonists (CYM5422 or SEW2871) and retinas were harvested 24 h later. Alternatively, eyes were injected with an S1pr1 agonist (SEW2871 or CYM5442), S1pr1 antagonist (MT1303 or NIBR0213), S1pr3 antagonist (TY52156), S1pr modulator (FTY720), Sphk1 antagonist (PF543), or S1P lyase antagonist (S1PL-in-31) before and with NMDA, and retinas were harvested 24 h after the last injection. FTY720, or Fingolimod, is a clinically approved S1pr1 modulator which initially acts as an agonist to S1pr1 but later induces receptor internalization and degradation (Chun et al., 2021a). We used NFκB reporter mice wherein eGFP expression is driven by a chimeric promoter containing three HIV-NFκB cis-regulatory elements (Magness et al., 2004). In the NFκB-reporter mice, there is MG-specific NFκB activation in retinas treated with NMDA, cytokines, and different growth factors (Palazzo et al., 2023; Fig. 4a). In undamaged retinas, we found that S1pr1 activation, via S1P or different agonists, significantly increased numbers of Sox2-positive MG that express NFκB reporter in undamaged retinas (Fig. 4b,c). Additionally, S1pr1 agonist treatments increased numbers of NFκB-expressing MG in damaged retinas (Fig. 4b). Consistent with findings from studies using S1pr1 agonists, numbers of NFκB-GFP-expressing MG were increased in retinas treated with S1P lyase inhibitor, decreased in retinas treated with S1pr1 antagonists, and decreased in retinas treated with Sphk1 inhibitor (Fig. 4d,e). S1pr3 antagonist (Fig. 4d) and S1pr1 modulator (data not shown) had no significant effects on numbers of MG that expressed NFκB reporter in damaged retinas. Collectively, these findings suggest that autocrine S1P signaling via Sphk1-mediated synthesis of S1P and activation of S1pr1 activates NFκB signaling in MG in normal and damaged retinas.
Cross-regulation of S1P/S1pr1 and NFκB signaling in MG. Retinas were obtained from undamaged or NMDA-injured eyes treated with a vehicle control or a small molecule agonist/antagonist a–g, Retinal sections were labeled with DAPI (e) and antibodies to Sox2 (red, a, c, e; pseudocolored cyan, f) and NFκB-GFP (green, a, c, e) or FISH probes to S1pr1 (red puncta; f). Arrows indicate the nuclei of MG. Scale bars: a, c, and f represent 50 µm. Regions of interest (yellow boxes) are enlarged 1.8-fold and displayed in separate panels (f). Histograms illustrate the mean (bar ± SD) number of NFκB-eGFP in MG (b, d) or number of FISH puncta per MG (g), where each dot represents one biological replicate (n = 5–8), and blue lines connect replicates from control and treated retinas from one individual. Significance of difference (p values) was determined by using a paired t test (b, d, g). Abbreviations: ONL, outer nuclear layer; INL, inner nuclear layer; IPL, inner plexiform layer; EdU, 5-ethynyl-2′-deoxyuridine; NMDA, N-methyl-D-aspartate; ns, not significant; UMAP, Uniform Manifold Approximation and Projection.
S1P signaling is known to activate NFκB (Alvarez et al., 2010a), and NFκB is known to reciprocally regulate S1P signaling (O’Sullivan et al., 2014; Hutami et al., 2017). Accordingly, we sought to identify changes in patterns of S1pr1 expression in retinas where NFκB signaling was inhibited. We applied a small molecule NFκB inhibitor, PGJ2, which has been shown to potently block NFκB reporter activation in NMDA-damaged mouse retinas (Palazzo et al., 2022a), followed by FISH for S1pr1. We found that the number of S1pr1 FISH puncta per MG were significantly increased in NMDA-damaged retinas treated with PGJ2 (Fig. 4f,g). We next probed for changes in levels of S1P-related genes in scRNA-seq of retinas where NFκB signaling has been selectively deleted from MG. We analyzed libraries that we generated and described in a prior study (Palazzo et al., 2022a). Rlbp1-CreERT mice were crossed to mice carrying floxed alleles of Ikkb to prevent NFκB signaling in MG. Activation of canonical NFκB signaling involves the IKK complex (Ikka and Ikkb) which is activated to phosphorylate IkBα/IkBβ, thereby releasing NFκB transcription factors (p65 and p50) and allowing them to translocate into the nucleus to regulate transcription of target genes (Zhang et al., 2017). Deletion of Ikkb blocks signaling through the NFκB pathway by preventing IKK-mediated phosphorylation and degradation of IkBa/IkBb, thereby causing sequestration of NFκB transcription factors in the cytoplasm (Li et al., 2003; Zhang et al., 2017, p. 30). Consistent with patterns of expression seen in scRNA-seq libraries in WT retinas (Fig. 1), at 8 h after NMDA treatment, S1pr1, S1pr3, and Sphk1 were highly expressed by MG (Fig. S2a–f). In addition, Sgpl1 was detected in immune cells and MG (Fig.S2g), consistent with patterns of expression seen in WT retinas. We bioinformatically isolated the nearly 1,400 MG to permit a more detailed analysis. Consistent with the effects of the small molecule inhibitor, in MG where Ikkb was deleted, levels of S1pr1 were significantly upregulated, whereas levels of S1pr3 and Sphk1 were significantly reduced, and levels of Sgpl1 were unchanged (Fig. S2h). Collectively, these findings suggest that NFκB regulation of S1pr1/3 expression and downstream NFκB signaling through S1pr1/3 reciprocally influence expression levels in MG in damaged retinas.
Next, we investigated whether pharmacological activation or inhibition of S1P-related receptors and enzymes influenced ERK phosphorylation, mTor activation (pS6), and cFos expression in normal and damaged retinas. In undamaged retinas treated with exogenous S1P or S1PR1 activator, we applied antibodies to different second messengers for cell signaling pathways in the retina (Fig. S3a–d). In retinas treated with S1pr1 agonist (SEW2871), pERK1/2 levels were significantly elevated in Sox2-expressing MG (Fig. S3a,b). However, intravitreal delivery of S1P did not result in the accumulation of pERK1/2 in MG (Fig. S3b), possibly due to rapid degradation by Sgpl1. Levels of immediate early gene cFos or mTor target pS6 were not significantly different between treatment groups (Fig. S3c,d), indicating that these pathways are not affected by S1P signaling.
The accumulation of immune cells in damaged retinas is influenced by S1pr1, but not Sphk1
To further investigate how signaling through S1pr1 and synthesis of S1P via Sphk1 in MG influences the retina, we generated conditional knock-out (cKO) mice. We crossed Rlbp1-CreERT mice with mice carrying floxed alleles of S1pr1 or Sphk1. Mice were treated with vehicle or tamoxifen for 4 consecutive days, followed by 3 d to allow recombination and tamoxifen to clear. Relative to wild-type animals, S1pr1 activator had no significant effect on pERK1/2 levels in Sox2-expressing MG in mice with S1pr1 cKO (Fig. S3e,f). To evaluate gene expression, we applied FISH probes for S1pr1 and Sphk1 to retinas harvested 4 h after NMDA (Fig. S4a,b). As expected, FISH signals for S1pr1 and Sphk1 were prominent in the INL and were nearly absent in damaged retinas with MG-specific cKO of S1pr1 or Sphk1 (Fig. S4a,b).
Next, we probed for the accumulation of resident microglia and peripheral immune cells that are known to migrate into the retina after injury (White et al., 2017; Yu et al., 2020). In response to NMDA damage, peripheral monocytes infiltrate the neural retina to amplify inflammatory resident microglia signaling and inhibit Ascl1-mediated retinal regeneration (Todd et al., 2020; Blasdel et al., 2024). Eyes were injected with saline or NMDA, and retinas were harvested 2 d after NMDA (Fig. 5). Resident retinal microglia and macrophages typically express low levels of CD45 and high levels of Iba1, whereas peripheral monocyte-derived macrophages maintain steady high levels of CD45 and relatively lower levels of Iba1 in the homeostatic and injured retina (Sedgwick et al., 1991; O’Koren et al., 2016). Accordingly, we identified CD45high/Iba1low cells as putative infiltrating monocyte-derived macrophages (CD45+/Iba1−) and CD45high/Ibahigh or CD45low/Iba1high cells as microglia (CD45+/Iba1+ or CD45−/Iba1+, respectively). We found that cKO of S1pr1 or Sphk1 had no effect on numbers of immune cells in undamaged retinas (Fig. 5a–d). In WT retinas, 2 d after NMDA treatment, we found significant increases in total numbers of Iba1+ cells, but not CD45+/Iba1− cells (Fig. 5a–d). Numbers of Iba1+ and CD45+/Iba1− immune cells in NMDA-damaged retinas with S1pr1−/− MG were significantly increased when compared with numbers seen in undamaged retinas with S1pr1−/− MG and NMDA-damaged retinas with WT MG (Fig. 5a,b). By comparison, there was no significant difference in the number of CD45+/Iba1− immune cells in NMDA-damaged retinas with Sphk1−/− MG compared with numbers seen in NMDA-damaged retinas with WT MG (Fig. 5b–c). However, numbers of microglia in damaged retinas with Sphk1−/− MG were significantly increased compared with numbers seen in undamaged retinas with Sphk1−/− MG (Fig. 5b,d). In summary, the deletion of S1pr1, but not Sphk1, from MG causes the increased accumulation of Iba1-expressing microglia and CD45-expressing immune cells in acutely damaged retinas.
Immune cell accumulation in S1pr1 KO and Sphk1 KO retinas. Rlbp1-CreERT:S1pr1fl/fl and Rlbp1-CreERT:Sphk1fl/fl mice were intraperitoneally injected with tamoxifen or corn oil once daily for 4 d. Eyes were intravitreally injected with NMDA or saline control, and retinas were collected 48HPI. Retinal sections were labeled with antibodies to Iba1 (blue) and CD45 (green; a, b). Solid arrows indicate Iba1+/CD45+ cells, and hollow arrowheads indicate Iba1+/CD45− cells. Scale bars: a and b, represent 50 µm. Histograms represent the mean (bar ± SD) where each dot represents one biological replicate (c, d; n = 5–18). Significance of difference (p values) was determined by using ANOVA with Sidak’s correction. Abbreviations: ONL, outer nuclear layer; INL, inner nuclear layer; IPL, inner plexiform layer; GCL, ganglion cell layer; NMDA, N-methyl-D-aspartate; ns, not significant.
S1P signaling in MG regulates neuronal survival in damaged retinas
NMDA treatment is known to selectively destroy bipolar cells, amacrine cells, and retinal ganglion cells, but not MG in the mouse retina (Todd et al., 2019). Since neuroinflammation is known to impact both neuronal survival and regeneration in NMDA-damaged retinas (Todd et al., 2019), and S1P signaling influences the NFκB pathway and the accumulation of microglia/macrophage (current study), we investigated whether S1P influences cell death and neuronal survival. In undamaged retinas with WT or S1pr1 cKO MG, we found no evidence for dying TUNEL-positive cells (Fig. 6a–c). We found significant increases in numbers of TUNEL-positive cells in the INL/ONL or GCL in NMDA retinas at 2 d after treatment, and these numbers were significantly reduced in the GCL with MG-specific cKO of S1pr1 (Fig. 6a–c). Consistent with these findings, we found significantly more surviving retinal ganglion cells (RGCs) labeled for Brn3a or HuC/D in the GCL at 2 weeks after NMDA treatment in S1pr1 cKO retinas compared with numbers seen in WT retinas (Fig. 6d–g). However, we did not find a significant difference in numbers of HuC/D+ or Pax6+ amacrine cells in the INL at 2 weeks after NMDA treatment in retinas with WT MG and retinas with S1pr1−/− MG (Fig. 6f,h–j).
cKO of S1pr1 from MG is neuroprotective to retinal ganglion cells. Sections of the retina were labeled for fragmented DNA (TUNEL; green; a) and DAPI (blue), or Brn3a (green; d), tdTomato (red) and DAPI (blue), HuC/D (green; f), tdTomato (red) and DAPI (blue), or Pax6 (green, i), tdTomato (red), and DAPI (blue). Arrows indicate TUNEL+ nuclei (a) or Brn3a+ nuclei (d) or HuC/D+ cells (f) and small double arrows indicate GFP+ endothelial cells. Scale bars: a, d, and f, represent 50 µm. Histograms represent the mean (bar ± SD) where each dot represents one biological replicate (b, c, e, g, h, j; n = 5–10). Significance of difference (p values) was determined by using ANOVA with Sidak’s correction. Abbreviations: ONL, outer nuclear layer; INL, inner nuclear layer; IPL, inner plexiform layer; GCL, ganglion cell layer; NMDA, N-methyl-D-aspartate; ns, not significant; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling.
In retinas with cKO of Sphk1 in MG, we found significantly fewer TUNEL+ cells in the INL and ONL compared with WT retinas (Fig. 7a–c). We did not find a significant difference in numbers of RGCs labeled for Brn3a or HuC/D at 2 weeks after NMDA treatment in retinas with Sphk1−/− MG compared with numbers seen in retinas with WT MG (Fig. 7d,e). However, we found a significant increase in numbers of HuC/D+ or Pax6+ amacrine cells in the INL at 2 weeks after NMDA treatment in retinas with Sphk1−/− MG (Fig. 7f–i). Collectively, these findings indicate that cKO or S1pr1 or Sphk1 in MG is neuroprotective to inner retinal neurons that are damaged by NMDA.
cKO of Sphk1 from MG is neuroprotective to inner retinal neurons. Sections of the retina were labeled for fragmented DNA (TUNEL; green; a) and DAPI (blue), or Brn3a (green; d), tdTomato (red) and DAPI (blue), or HuC/D (green; f), tdTomato (red) and DAPI (blue), or Pax6 (green; h), tdTomato (red) and DAPI (blue). Single arrows indicate TUNEL+ nuclei (a) or Brn3a+ nuclei (d) or HuC/D+ cells (f), and small double arrows indicate GFP+ endothelial cells. Scale bars: a, d, f, and h represent 50 µm. Histograms represent the mean (bar ± SD) where each dot represents one biological replicate (b, c, e, g, i; n = 6–8). Significance of difference (p values) was determined by using ANOVA with Sidak’s correction. ONL, outer nuclear layer; INL, inner nuclear layer; IPL, inner plexiform layer; GCL, ganglion cell layer; NMDA, N-methyl-D-aspartate; ns, not significant; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling.
Todd and colleagues reported that microglia suppress Ascl1-mediated neurogenesis without influencing cell survival in mouse retinas (Todd et al., 2020). Additionally, we have previously reported that NFκB signaling in MG is induced by Il1α, Il1β, and TNF from reactive microglia and regulates genes responsible for neuron survival (Palazzo et al., 2022a, 2023). Accordingly, we investigated whether the neuroprotective effects of S1pr1 inhibition are dependent on immune cell proliferation and recruitment. We ablated microglia in S1pr1 KO mice using the Colony Stimulating Factor 1 Receptor (CSF1R) inhibitor PLX5622 prior to NMDA treatment and collected retinas at 2 DPI (Fig. S4). Cell death was not detected in saline-injected retinas of PLX-treated animals, but we observed significant increases in numbers of TUNEL-positive cells in the INL and GCL in NMDA-treated retinas at 2 d after treatment (Fig. S5a–c). In PLX-treated animals, numbers of TUNEL+ cells were significantly reduced in both the GCL and the INL with MG-specific cKO of S1pr1 (Fig. S5a–c). Collectively, these findings indicate that S1pr1 signaling regulates cell survival following NMDA damage, and this process operates independently of signals from immune cells.
Finally, we investigated whether pharmacological inhibition of S1pr1, Sphk1, or Sgpl1 influences cell death. Eyes were injected with vehicle, MT1303 (S1pr1 inhibitor), NIBR0213 (S1pr1 inhibitor), PF543 (Sphk1 inhibitor), FTY720 (S1pr1 modulator), or S1PLin31 (SGPL1 inhibitor) before and with NMDA, and retinas harvested 1 d after NMDA treatment. Although MT1303 had no significant effect, the NIBR0213 significantly decreased numbers of dying cells across all layers of the retina, particularly the INL (Fig. S6a–d). The solubility and pharmacodynamics of the MT1303 may have limited the efficacy of this drug (Shimano et al., 2019). In damaged retinas treated with S1pr1 modulator FTY720, cell death was reduced overall but reached significance only in the GCL (Fig. S6a–d). This was similar to S1pr1 inhibitor NIBR0213 and consistent with the notion that FTY720 functions mostly to decrease signaling through S1pr1. In retinas treated with Sphk1 inhibitor, numbers of dying cells were reduced in the INL/ONL and GCL (Fig. S6a–d). Complementary to these findings, treatment of retinas with S1PLin31 resulted in significant increases in numbers of TUNEL + cells in the INL (Fig. S6a–d). Collectively, these findings suggest inhibition of S1P synthesis or S1pr1 signaling suppresses and inhibition of S1P degradation increases cell death in acutely damaged retinas (Table 3).
Discussion
Patterns of expression of S1pr1 and Sphk1 in the retinas of different vertebrates
We recently described conserved patterns of expression of S1P-related genes in the retinas of chicks, zebrafish, and humans (Taylor et al., 2024a). In the current study, we find that patterns of expression of S1P-related genes in the mouse retina are distinctly different from those seen in other vertebrates. For example, in chick, human, and zebrafish retinas, S1pr1 is highly expressed by resting MG and is downregulated in activated MG (Taylor et al., 2024a). By comparison, S1pr1 is relatively low in resting MG and highly upregulated in activated MG in the damaged mouse retinas. In the mouse and human retinas Sphk1 is upregulated by activated glia, whereas in the chick retina SPHK1 is downregulated in activated MG and MGPCs (Taylor et al., 2024a). It is possible that activation of S1pr1 “primes” or “kick-starts” proinflammatory responses in MG and these responses are needed to begin the process of dedifferentiation and reprogramming in chick and perhaps zebrafish. In contrast, in mouse MG this proinflammatory process involves the rapid activation of S1pr1 to drive reactivity and returning to resting programs.
This study is the first to genetically target Sphk1 or S1pr1 specifically in MG. Some studies have reported S1P receptor antibody labeling in neural cells and RPE (Joly et al., 2017; Porter et al., 2018; Nakamura et al., 2021; Ahmed et al., 2024). In the current study, we did not observe plausible patterns of labeling for Sphk1 or S1pr1 antibodies in the mouse retina. However, we do find that S1pr1 mRNA appears predominantly in the MG in normal and damaged retinas. A recent study using an S1pr1 reporter did not report expression in mature CNS neurons but did find expression in oligodendrocytes, oligodendrocyte progenitors, a subset of neural stem cells, and myeloid cells in the brain (Hashemi et al., 2023). It is possible that neuronal expression of S1pr1 can be induced by some models of retinal damage and not others, similar to the dynamic expression S1P receptors in the CNS in response to different models of multiple sclerosis (Van Doorn et al., 2010; Liu et al., 2016; Hashemi et al., 2023).
S1P signaling and neuroprotection
In the current study we provide evidence that the activity of S1pr1 and Sphk1 in MG regulates neural survival (Fig. 8; Table 3), and this process is not dependent on the presence of microglia (Fig. S5). However, the mechanisms underlying MG-mediated neuronal survival are not well understood. MG span the entire width of the retina and serve many homeostatic functions, including neurotransmitter metabolism, potassium ion buffering, and inner blood–brain barrier maintenance (Karwoski et al., 1989; Izumi et al., 1999; Kugler et al., 2021); it is likely that these functions change following retinal damage and following activation of different cell signaling pathways. Additionally, MG are known to produce different neurotrophic factors that influence the survival of neurons in different models of retinal disease (Fu et al., 2015; Gao et al., 2025). It seems likely that S1pr1 activation in MG leads to changes in the production of secreted factors that impact neuronal survival (Fig. 8; Table 3).
Schematic summary of findings. Sphk1, S1pr1, and S1pr3 are expressed in MG. In the current study, we provide evidence that S1pr1 activity in MG suppresses Ascl1-mediated reprogramming, inhibits neurotrophic activity, and stimulates NFκB activation in the damaged retina. Astrocytes, endothelial cells, and immune cells express components of the S1P pathway, which may interact with S1P:S1pr1 signaling in MG to influence reprogramming, neuroprotection, and inflammation.
Specificity of S1P targeting drugs
In mice, there is substantial evidence that elevated retinal S1P contributes to the degeneration of multiple retinal neuron types (Shiwani et al., 2021). In the current study, we report neuroprotective effects of blocking Sphk1 and S1pr1, wherein knocking out these genes or applying inhibitors promoted cell survival following NMDA-induced neuronal damage. However, there were some inconsistencies in our observations between small molecule inhibitors and genetic knockouts. Differences in cell death might be attributed to the different timepoints at which retinas were harvested (24 HPI vs 48 HPI), or due to the limited inhibition and pharmacodynamics of the different drugs. It is also possible that changes in cell death in retinas treated with drugs could be mediated, in part, by their action on retinal endothelial cells and/or astrocytes, which express Sphk1 and S1P receptors. Finally, a combination of factors could have been responsible for these discrepancies.
It is important to acknowledge that while pharmacological manipulations provide advantages of reversibility and temporal control, they may also have off-target effects. We chose drugs based on specificity of interactions and solubility. We have previously validated the effects of Sphk1 inhibitor PF543 and S1P lyase 1 inhibitor S1PLin31 using LC-MS to measure retinal levels of S1P (Taylor et al., 2025). In the current study, we applied different types of S1pr1 inhibitors and activators, which had consistent and complimentary effects on NFκB signaling. Additionally, we tested the specificity of S1pr1 agonist SEW2871 in MG with S1pr1 conditionally deleted, and this drug failed to stimulate pERK1/2 accumulation. Small-molecule drugs are convenient tools to use alongside genetic manipulations; however, we cannot exclude the possibility of off-target or indirect effects.
S1P signaling and the accumulation of immune cells in damaged retinas
The activation of microglia and MG in the retina are intimately linked. For example, we recently reported that hundreds of genes are up- or downregulated in MG in normal and damaged retinas when microglia are ablated in the chick retina (El-Hodiri et al., 2023). We report here that cKO of S1pr1, but not Sphk1, from MG results in the accumulation of significantly more immune cells in damaged retinas; this accumulation results, in part, from enhanced recruitment of CD45+/Iba1 cells, a profile which distinguishes resident microglia from infiltrating monocyte-derived macrophages (Sedgwick et al., 1991; O’Koren et al., 2016; Palazzo et al., 2022a). Different patterns of expression of S1pr1 and Sphk1 in support cells in the retina may underlie the subtly different outcomes when these genes are specifically deleted from MG. S1pr1 is highly expressed by MG and endothelial cells, whereas Sphk1 is highly expressed by MG and astrocytes. cKO of S1pr1 or Sphk1 in MG should not directly diminish endothelial S1pr1 expression or astrocytic S1P production; however, it is likely that MG-specific KO of S1P-related genes indirectly influences S1P-related gene expression in other retinal cells. cKO of S1pr1 from MG in damaged retinas increased the accumulation of immune cells, suggesting that S1pr1 signaling in MG normally suppresses the production of cytokines that recruit immune cells into the acutely damaged retina (Fig. 8; Table 3). However, cKO of Sphk1 from MG (which should directly reduce retinal levels of S1P and indirectly reduce S1pr1 signaling) in damaged retinas had no effect upon the accumulation of immune cells, suggesting that S1P production by astrocytes may compensate for the loss of S1P secretion from MG. The differences in increased survival of amacrine cells (but not RGCs) with cKO of Sphk1 and increased survival of RGCs (but not amacrine cells) with cKO of S1pr1 likely also resulted from changes to persistent expression of Sphk1 (S1P production) and S1pr1 (S1P signaling) in astrocytes and endothelial cells, respectively. Consistent with this notion, pharmacological inhibition of Sphk1, which should have suppressed S1P production in MG and astrocytes, resulted in reduced cell death in the INL and the GCL. Alternatively, MG-specific cKO of S1pr1 or Sphk1 may have resulted in alternate compensatory mechanisms, such as upregulation of S1pr3 in MG to differentially influence neuroprotective effects on amacrine and ganglion cells. Further studies are required to identify the exact molecular mechanisms that underlie the cell type-specific changes in neuronal survival with MG-specific cKO of S1pr1 and Sphk1.
We have previously reported that NFκB signaling in MG promotes the accumulation of reactive immune cells, decreases neuronal survival, and suppresses the reprogramming of Ascl1-overexpressing MG (Palazzo et al., 2022a). Many secreted factors stimulate NFκB, including proinflammatory cytokines, TNF, CNTF, and osteopontin (Ji et al., 2022; Palazzo et al., 2023). In the current study, we propose that the S1pr1:NFκB signaling in MG is a significant driver of ganglion cell death in NMDA-damaged retinas. This signaling relationship appears to be bidirectional, wherein MG-specific cKO of Ikkb (blocking NFkB signaling) increases S1pr1 expression and decreases S1pr3 and Sphk1 expression. It seems inconsistent that Ikkb cKO and S1pr1 cKO (diminished NFκB signaling) promote neuron survival, but Ikkb cKO suppresses and S1pr1 cKO promotes the accumulation of immune cells in damaged retinas. Thus, NFκB signaling in MG significantly contributes to the MG:microglia coordination, and S1pr1 may impact this coordination via pathways other than NFκB.
S1pr1 inflammatory signaling and neurogenesis
Cytokine-mediated proinflammatory signaling is required to stimulate MG to become activated and proliferate as MGPCs in the retinas of chicks and zebrafish (Fischer et al., 2014; Silva et al., 2020). However, sustained microglial reactivity, cytokine signaling, and NFκB activation suppress MG reprogramming (White et al., 2017; Palazzo et al., 2020). In the current study, we provide evidence that S1P signaling suppresses Ascl1-mediated reprogramming of MG into progenitor cells that produce bipolar-like neurons (Fig. 8, Table 3). This suppression may be mediated, in part, by S1pr1/3 signaling through the NFκB pathway. Unlike direct NFκB inhibition, S1P inhibition did not significantly increase numbers of EdU+ Ascl1-OE MG (Palazzo et al., 2022a). This outcome is similar to findings from Jak/Stat inhibition (Jorstad et al., 2020), supporting the notion that NFκB may be a proinflammatory “hub” that blocks the neurogenic potential of MG. Alternatively, it is possible that while Ascl1-OE and NFκB inhibition stimulate MG proliferation, S1P and Stat3 inhibition guide Ascl1-OE MG toward direct neural trans-differentiation.
In the chick, S1PR1 inhibitors and NFκB inhibitors stimulate the formation of proliferating MGPCs following neuronal damage (Palazzo et al., 2020; Taylor et al., 2024a). In the absence of proinflammatory signals from microglia, S1PR1 inhibitor promotes damage-dependent MGPC formation (Taylor et al., 2024a), whereas NFκB activator increases MGPC formation (Palazzo et al., 2020). These findings suggest that the NFκB pathway must be transiently activated to initiate reactivity and dedifferentiation but thereafter must be suppressed to permit upregulation of progenitor genes and proliferation. Collectively, these findings suggest that signaling through S1pr1/3 potentiates or sustains NFκB signaling to suppress reprogramming of MG into neuron-like cells.
Conclusions
Our findings indicate that S1pr1 and Sphk1 are rapidly and transiently upregulated by MG following acute injury in the mouse retina. Treatments that activate/inhibit S1P receptors result in increased/decreased NFκB signaling in MG in normal and damaged retinas (Fig. 8, Table 3). The expression of S1pr1 in MG is upregulated by NFκB signaling and downregulated by forced expression of Ascl1. Selective deletion of S1pr1 and/or Sphk1 from MG has no apparent effects on undamaged retinas, whereas the deletion of these genes from MG in acutely damaged retinas increases the accumulation of immune cells, decreases cell death, and increases the survival of inner retinal neurons (Fig. 8, Table 3). Consistent with these findings, pharmacological treatments that increase S1P signaling increased cell death, whereas treatments that decreased S1P signaling decreased cell death in damaged retinas (Fig. 8, Table 3). We conclude that inflammation mediated by autocrine signaling via S1pr1 in MG in damaged retinas suppresses the neuroprotective functions of MG and Ascl1-mediated reprogramming of MG into neuron-like cells. Accordingly, the S1P pathway is an attractive druggable target to regulate inflammation and neuronal survival, and possibly promote neuronal regeneration to treat sight-threatening diseases of the retina.
Data Availability
Cell Ranger output files for Gene-Cell matrices for scRNA-seq data for libraries from saline and NMDA-treated retinas are available through SharePoint links for embryonic chick retina databases https://osumc.sharepoint.com/:f:/s/Links/Eoto-Qg2uuxDn1bHWMM6gdkBTft4S_YSBjQJResxY-qehA?e=mdaPlg and post-hatch normal and treated chick retina databases https://osumc.sharepoint.com/:f:/s/Links/Eoto-Qg2uuxDn1bHWMM6gdkBTft4S_YSBjQJResxY-qehA?e=mdaPlg. scRNA-seq datasets are deposited in GEO (GSE135406, GSE242796) and Gene-Cell matrices for scRNA-seq data for libraries chick retinas treated with saline or NMDA retinas are available through NCBI (GSM7770646, GSM7770647, GSM7770648, GSM7770649).
Gene-cell matrices for scRNA-seq data are deposited at GitHub: scRNA-seq data from NMDA-damaged mouse retinas (Hoang et al., 2020) can be queried here: https://proteinpaint.stjude.org/F/2019.retina.scRNA.html.
ScRNA-seq data for mouse retina at different stage of postnatal development (Li et al., 2024) https://cellxgene.cziscience.com/collections/a0c84e3f-a5ca-4481-b3a5-ccfda0a81ecc.
Footnotes
We thank Timothy Hla for providing the Sphk1f/f and S1pr1f/f mice. We also thank Dr. Ed Levine for providing the Rlbp1-CreERT mice and Dr. Dennis Guttridge for providing the NFκB-eGFP reporter mice.
This work was supported by the National Eye Institute R01 EY032141-05 (A.J.F.).
The authors declare no competing financial interests.
This paper contains supplemental material available at: https://doi.org/10.1523/JNEUROSCI.0150-25.2025
- Correspondence should be addressed to Andy J. Fischer at andrew.fischer{at}osumc.edu.














