Ifi27l2a is highly upregulated after stroke in MG and aging significantly enhances this upregulation. To define the transcriptional signature across multiple cell types in the post-stroke brain, we performed scRNA-seq of young (3-month-old) and aged (20-month-old) male and female C57BL/6J mice subjected to permanent distal middle cerebral artery occlusion (pdMCAO) or sham surgeries (Table 1). The pdMCAO stroke model was used since it produces both primary injury (cortical infarct) and secondary injury in the thalamus 2-weeks post-stroke 11. Since the cortex and thalamus also feature clear increases in microgliosis and astrogliosis following stroke, we included a brain region containing both peri-infarct cortex and thalamus for scRNA-seq study 11.
We evaluated differential gene expression (DGE) patterns, as an initial approach for determining how the molecular signature of cells within the young and aged brain change post stroke. To measure the effect of aging in stroked brains, we first integrated young stroke (AGGR2) and aged stroke (AGGR4) data into a single analysis (Seurat package 12) for analysis. Then, a single integrated analysis (data integration, PCA, UMAP and clustering, and DGE) was performed. Visualization of this merged dataset of 21,092 cells from the aged and young stroke mouse brain through dimension reduction by uniform manifold approximation and projection (UMAP) identified eight clusters of unique cell types based on gene expression differences. Identities were assigned to each of the eight clusters using the expression of conserved cell type markers, including microglia (MG, n = 7180) (Trem2), oligodendrocytes (Oligo, n = 5149) (Plp1), endothelial cells (EC, n = 3943) (Cldn5), astrocytes (Astro, n = 1698) (Aldoc), lymphocytes (Lym, n = 1495) (Plac8), epithelial cells (Epi, n = 1192) (1500015O10Rik), vascular leptomeningeal cells (VLMC, n = 127) (Dcn) and vascular endothelial cells, venous (VECV, n = 89) (Pglyrp1) (Fig. 1a-b). Notably, the proportion of MG and Lym clusters were highly increased in the aged stroke brain, whereas the oligodendrocytes were reduced (Fig. 1c). These findings are consistent with more extensive white matter injury in the aged stroke brain and correlate with increased MG-mediated neuroinflammation and lymphocyte infiltration. As we and others have demonstrated that MG are highly sensitive to inflammation and ischemic stress and act to regulate innate immunity in brains 3,13, we focused our subsequent analysis on transcriptional changes within MG.
Interestingly, our unbiased analyses of 21,092 cells (combined from young and aged stroke brains) showed that Ifi27l2a was the most highly upregulated gene in MG clusters in aged stroke, compared to young stroke (Table 2). The top five genes that were significantly upregulated in MG in the aged stroke brain included MG related genes (Lgals3, Lyz2, Lgals3bp) and another interferon-stimulated gene (ISG), Ifitm3. Dot plots compared the expression level and percent of cells expressing the top five genes upregulated in aged stroke versus young stroke (Fig. 1d). Within the MG cluster, there was a notable increase in the percentage of cells expressing Ifi27l2a between aged stroke and young stroke (62.6% vs 29.3%) (Fig. 1d). Total normalized expression of Ifi27l2a from all cells showed increased Ifi27l2a expression in cells of aged stroke, compared to young stroke (Fig. 1e). Ifi27l2a was more highly upregulated in MG of aged stroke brain, suggesting aging may act synergistically with ischemic stroke to promote Ifi27l2a expression in MG (Fig. 1f). While most of the Ifi27l2a-expressing cells belonged to the MG cluster, Ifi27l2a expression was also detected in Lym and VLMC populations (Fig. 1g). The VLMC showed increased Ifi27l2a expression with aged stroke, whereas stroke-induced Ifi27l2a expression in Lym was not markedly altered by aging. Other MG markers, such as Lgals3, Ifitm3 and Lgals3bp, were also upregulated in MG and other cells following stroke (Fig. 1h-j). In contrast, there was no synergistic effect of aging with stroke on C1qa expression, a MG marker gene (data not shown). As expected, a known marker of activated MG, Cst7, was increased in aged stroke compared to young stroke brain (Extended data Fig. 1a), confirming that MG were more highly activated in aged stroke brains than in young stroke brains. In addition, we found significant upregulation of Apoe and Lyz2, while Aif1 level appeared only slightly increased in MG in aged stroke compared to young stroke (Extended Data Fig. 1b-d). Taken together, the scRNA-seq data suggest an age-dependent upregulation of Ifi27l2a, which occurs predominantly in MG after stroke.
scRNA-seq revealed that aging itself is sufficient to increase Ifi27l2a transcripts in MG. Following our finding that Ifi27l2a is upregulation following stroke in an age-dependent manner, we next sought to determine if aging alone impacts Ifi27l2a expression. Thus, we compared young and aged sham brains, integrating the young and aged sham operated samples (young sham - AGGR1, aged sham - AGGR3). Eight clusters were identified (Extended data Fig. 2a and 2b), including oligodendrocytes (Oligo) (Plp1), MG (C1qa), EC (Cldn5), astrocytes (Astro) (Gpr37l1), epithelial cells (Epi) (Ttr), lymphocytes (Lym) (Nkg7), vascular smooth muscle cells, arterial (VSMCA) (Des), and B cells (CD79a). To determine how aging affects the transcriptional landscape in MGs, we compared the expression and percent of cells expressing the previously identified top five MG genes (Ifi27l2a, Lgals3, Ifitm3, Lyz2, and Lgals3bp) in young and aged sham brains. All 5 genes that were upregulated in MG from aged stroke brain were also increased by aging alone (Extended Fig. 2c). Notably, Ifi27l2a transcript levels significantly increased with aging, as did Rps27rt. C1qa, on the other hand, was not dramatically altered between young and aged sham animals (Extended data Fig. 2d). We also confirmed the increased expression of Ifi27l2a in MG, Lym and B cell clusters in aged brains (Extended data Fig. 3a). The violin plots revealed modest upregulation of Ifi27l2a in MG in aged brain compared to young brains. Interestingly, we found significant age-dependent upregulation of ribosomal protein genes such as Rpl35, Rps27rt, and Rps28. This age-dependent upregulation of Rps27rt in all clusters, including MG and Lym (Extended data Fig. 3b), suggested aging-mediated changes in ribosomal complex composition in MG. Expression of two other genes associated with activated MG (Aif1 and Il-1b) were also modestly increased with aging (Extended data Fig. 3c-d). We repeated the same analyses comparing sham to stroke for aged (Extended data Fig. 4, 5, 6) and young cohorts (Extended data Fig. 7, 8). Together, these data suggest a synergistic effect of aging and stroke on Ifi27l2a expression.
Disease-associated microglia (DAM) are present in the aged brain, and significantly increased following stroke. DAM are a recently discovered sub-population of MG found in the brains of various neurodegenerative diseases, such as AD, PD and ALS (REFs). We asked if DAMs are increased in the aged stroke brain. A recent study reported that homeostatic genes, such as C1qa, Ctss, Hexb, and Csf1r are not upregulated during the transition of MG into DAM, whereas other MG related genes (Spp1, Cst7, Lpl, and Itgax) are highly upregulated in DAM 14. To determine whether a DAM-like MG subpopulation is increased in the aged or stroke brain, we compared the number and relative percentage of DAM in sham and stroke. The DAM subtype was defined by the elevated expression of Aif1, Spp1, Cst7, and Lpl among all MG (filter applied: Aif1 high and Spp1 high and Cst7 high and Lpl high, threshold by count). As expected, we did not detect DAM (0.0%) in young sham brains. However, we a small number of DAM in aged sham samples (0.9%) (Extended data Fig. 9a) shows that stroke increased the percent of DAM in both the young and aged brain (10.2% in young stroke vs 17.9% in aged stroke). These data show that MGs are converted to DAMs during aging, but are DAMs are significantly increased following stroke.
Ifi27l2a is inversely correlated with DAM cell phenotype. We further analyzed our scRNA-seq data (aged sham and aged stroke) to determine whether Ifi27l2a plays a role in microglial activity, particularly in DAM. First, we examined whether Ifi27l2a expression correlated with expression of DAM-related markers, such as Lpl, Spp1, Cst7 and Itgax. We subset MG into 4 different sub-clusters based on their levels of Ifi27l2a expression (normalized Ifi27l2a expression: 0.3–0.99, 1-1.99, 2-2.99, 3+). We consistently observed a negative correlation between Ifi27l2a and DAM related gene expression (Lpl, Spp1, Cst7) (Extended data Fig. 9b).
Furthermore, segregating MG into either an Ifi27l2a “high” or “low” expressing cells followed by correlation analysis with known DAM genes and MG homoeostatic genes revealed a negative correlation between Ifi27l2a expression and phagocytosis/DAM related genes (Lpl, Spp1, Itgax, Cst7, and Tyrobp), but not homeostatic genes or other MG genes (Extended data Fig. 9c). Comparing the expression of DAM genes and homeostatic genes between “low” and “high” Ifi27l2a MG subpopulations revealed that DAM-related transcripts were significantly reduced in Ifi27l2a “high” MG. However, homeostatic genes (Aif1, C1qc, Hexb, and Gapdh) were either not changed or slightly increased (Extended data Fig. 9c). Furthermore, MG genes which are down-regulated in DAMs (Csfr1, Olfml3, Trem119, and P2ry13) were increased in Ifi27l2a “high” MG (Extended data Fig. 9c). Together, these data show a strong negative correlation between Ifi27l2a expression and a mature DAM transcriptional signature in MG.
Regional Ifi27l2a expression with natural aging. While scRNA-seq showed that Ifi27l2a transcripts are enriched in MG and other cell types (e.g. Lym, VLMC) in both young and aged stroke brains, we lacked any data on whether there was a regional basis for these changes within the brain (e.g. within the primary injury in the cortex or within the secondary injury region occurring within the thalamus). The cortex and thalamus were extracted from the brains of naïve young (3 months, n = 4) and aged male mice (18–20 months, n = 4) to determine if there were regional differences in Ifi27l2a expression. Notably, Ifi27l2a mRNA was significantly upregulated in the aged thalamus (Fig. 2a, p < 0.05). Ifi27l2a expression also approached upregulation in the cortex in aged brains (p = 0.23). These findings agree with our earlier scRNA-seq finding and suggest normal aging increases Ifi27l2a expression in the brain. In addition, MG related genes Il-1b, Cst7, and Tyrobp were markedly increased in either the thalamus or cortex of aged brains, further supporting an age-dependent increase in MG activation (Fig. 2b-d). Transcripts for C1qb and Lpl, two genes which are known to be associated with microglia phagocytosis, were indistinguishable between the two stages (Fig. 2e-f). These data indicate that Ifi27l2a is induced along with other genes associated with proinflammatory MG phenotype in the aged brain.
Regional and temporal expression of Ifi27l2a in aged stroke brain. To provide regional and temporal expression of Ifi27l2a and other MG-related genes following stroke, we analyzed young and aged thalamus and cortex by qRT-PCR at 3 and 14 days post-stroke. As expected, Ifi27l2a was significantly elevated at three days (cortex) and two weeks (cortex and thalamus) after stroke, compared to sham (Fig. 2i). We evaluated two other genes associated with MG activation (Cst7) and reparative phagocytosis (Tyrobp), and which were found to be elevated in our scRNA-seq analysis. Both Cst7 and Tyrobp were increased in cortex and thalamus by two weeks post-stroke, but not by 3 days (Fig. 2g-h). These findings suggest that Ifi27l2a expression is associated with the earlier phase of MG activation following stroke. The delayed expression in thalamus reflects the slower progression of the secondary injury mechanism.
To provide spatial context to Ifi27l2a expression at the single-cell level, we profiled Ifi27l2a mRNA transcripts on mouse brain sections using single-molecule in situ hybridization (RNAscope). Probing for Ifi27l2a in aged sham and stroke brains revealed elevated transcripts in the peri-infarct area at 2 weeks post-stroke compared with sham-operated controls (Fig. 2j-l). Combining RNAscope for Ifi27l2a with immunostaining for Iba1 confirmed that the majority of Ifi27l2a transcript is present in activated MG in the peri-infarct region of the aged brain (Fig. 2m).
MG represent the predominant source of Ifi27l2a expression after stroke. Our analyses of stroked brains at post stroke day (PSD) 3 and PSD 14 revealed a significant increase in Ifi27l2a mRNA. To determine whether MG represented the predominant source for the increased Ifi27l2a expression, we used PLX5622 treatment to deplete MG in mice prior to inducing stroke. PLX5622 is a CSF1R antagonist that eliminates CNS-resident MG 15. CSF1R mediated signaling is required for MG survival and proliferation 16,17. Mice were treated with PLX5622 for seven days. On day 7 of administration of PLX5622, pdMCAO was performed. The PLX5622 diet was continued for 3 days after stroke surgery to prevent repopulation by MG. At PSD 3, brains were isolated and analyzed by qRT-PCR (ipsilateral hemisphere) and immunostaining (contralateral hemisphere). As a control, mice were fed normal diet (ND) for the same period (Fig. 3a). Notably, Ifi27l2a mRNA level was significantly reduced by 86% in PLX-stroked brains (ipsilateral hemisphere), compared to ND-stroked brains (Fig. 3b, p < 0.05). The effectiveness of PLX5622 to eliminate MG in brains was confirmed by Iba1 immunofluorescence (Fig. 3c-d) in the contralateral hemisphere. PLX5622 treatment resulted in a profound decrease in the number of MG in brains (Fig. 3d). Moreover, PLX5622 treatment significantly reduced Tmem119 expression in brains after stroke, compared to naïve or normal diet administered brains (Fig. 3e, p < 0.05, compared to naïve and ND-stroke). These data indicate that the induction of Ifi27l2a after stroke is primarily dependent on the MG population in the brain.
MG induce Ifi27l2a/IFI27L2 expression with inflammatory stimuli. We next used cultured MG to evaluate the potential for inflammatory mediators to promote Ifi27l2a expression. First, we used mouse primary MG collected from the mixed glial cell culture obtained from P2 pups. Primary MG were treated with TNF-α (20 ng/mL) and IFN-γ (20 ng/mL) for 24 hours (to measure mRNA level of Ifi27l2a) and 48 hours (to measure protein level of Ifi27l2a by ELISA with cell lysate). Both mRNA (Fig. 3f) and protein levels (Fig. 3g) of Ifi27l2a were significantly increased with treatment.
To determine whether these findings extended to a human in vitro MG model, we challenged human microglial cells (HMC3) by addition of pro-inflammatory cytokines (TNF-α [20 ng/mL] and IFN-γ [20 ng/mL]) in combination with oxygen/glucose deprivation (inflammation/OGD). This inflammatory challenge induced a significant upregulation of IFI27L2 mRNA in HMC3s (Fig. 3h, n = 5–6, p < 0.05). We found that human IFI27L2 protein level was dramatically induced at 20 hours post inflammation/OGD (Stim), compared to control treatment (Control) (Fig. 3i, representative of n = 4).
Given these results, we next tested if IFI27L2 protein was increased in the brains of patients that featured neuroinflammation. Sections from the brains of deceased patients without neurological disease (n = 2, female) and from stroke patients (n = 3, female) who also demonstrated cerebral amyloid angiopathy (CAA) pathology and tauopathy, in which neuroinflammation (microgliosis) is prevalent. Immunohistochemistry showed significant IFI27L2 expression in the stroke brain samples but low expression in age-matched control samples (Fig. 3j, representative of n = 2–3). Together, these data show the responsiveness of Ifi27l2a (murine) and IFI27L2 (human) to inflammatory stimulation and the presence of elevated IFI27L2 in brain of patients with multiple forms of neuroinflammatory disease.
Differential expression of Ifi27l2a in subtypes of microglia (MG) and macrophage (MΦ) populations in the aged brains following stroke. Given the extensive heterogeneity evident within MG and MΦ, we subjected the aged scRNA-seq datasets to more granular analysis to determine if Ifi27l2a expression profiles correlated with different functional roles. We ultimately identified a total of 28 clusters from brain cells of aged sham and aged stroke mouse brains, eight clusters of which were assigned an MG or monocyte/MΦ identity based on the expression of conserved cell markers (Extended data Fig. 10). Two MG homeostatic clusters were identified based on the expression of MG genes such as Siglech, Tmem119, Gpr34, P2ry12, and Selplg. These MG were annotated as Siglech homeostatic MG and P2ry12 homeostatic MG. We also identified two different MG that appeared to be in an activated status (Rag + activated MG and Tyrobp + activated MG). Two Monocyte-Macrophage populations were also identified. We also found the disease-associated MG (DAM) like cluster showing high expression of Lpl, Itgax, Cst7, and Spp1. Note that these genes also correlate with the microglial genes and lipid metabolism genes upregulated in DAMs in other neurodegenerative diseases, such as AD 14,18. Since we found that stroke and aging increase the expression of Ifi27l2a in MG, and that Ifi27l2a expression is negatively correlated with DAM genes, we asked whether expression levels and degrees of Ifi27l2a gene induction from sham to stroke in DAM would be different from MG in other sub-clusters. We therefore compared the degree of Ifi27l2a gene induction among MG sub-clusters in aged sham versus stroke brains (Table 3). Among the non-homeostatic MG clusters, Ifi27l2a induction in DAM (1.7 fold) is lower than any of the other activated MG.
Ifi27l2a expression is sufficient to promote MG activation. Given the induction of Ifi27l2a in MG in aged brains and following stroke, we sought to elucidate the functional role of Ifi27l2a in MG-mediated neuroinflammation. Changes in microglial morphology is an early, quantifiable sign of inflammation in MG and MG functionality. Thus, we asked if Ifi27l2a expression alone (without additional inflammatory mediators) could induce a pro-inflammatory morphology in MG. We infected a murine microglial cell line (Sim-A9 cells) with a lentivirus where the Cx3cx1 promoter drove the expression of Ifi27l2a and an eGFP reporter, or a lenti-eGFP control. At 5 days post-infection, quantification of cell morphology showed that induction of Ifi27l2a expression caused an increase in the percentage of cells with a small, rounded shape (to a more amoeboid morphology or de-ramification) compared to lenti-eGFP control (Fig. 4a-b). Interestingly, MG with higher Ifi27l2a expression (using eGFP intensity as a surrogate maker) showed more dramatic morphological changes compared to cells that had low Ifi27l2a/eGFP expression (Fig. 4c). These results provide direct evidence that Ifi27l2a alone can initiate MG activation, even in basal conditions (i.e. inflammatory stimuli are not required).
Ifi27l2a induces ROS production. Earlier reports showed evidence for Ifi27l2a localization in mitochondria within non-CNS cells 19. We also detected increased IFI27L2 in the peri-nuclear membrane and in mitochondria in HMC3 cells (not shown), leading us to question whether Ifi27l2a could mediate mitochondrial dysfunction in MG. Thus, we asked if Ifi27l2a expression alone could initiate the reactive oxygen species (ROS) generation in activated MG. We used CellROX Red and MitoSox. First, we utilized CellROX Red, a detector of most ROS species, to determine if Ifi27l2a expression induces ROS production in Sim-A9 cells in unstimulated conditions. Quantification by flow cytometry revealed that Ifi27l2a overexpression alone promotes a significant increase in ROS production (Fig. 4d, as expressed in median fluorescence intensity, MFI, Ctrl: lenti-eGFP control, Ifi27l2a: lenti-Ifi27l2a-eGFP, n = 4, * p < 0.05). Next, we only analyzed GFP positive cells, representing those with successful transduction. The ROS level was greater in Ifi27l2a expressing cells compared to eGFP only control cells (Fig. 4e, Ctrl: lenti-eGFP control, Ifi27l2a: lenti-Ifi27l2a-eGFP, n = 4, * p < 0.05).
We checked more specifically if mitochondria contribute as an Ifi27l2a-induced ROS source using Mitosox dye (specific indicator of mitochondria-derived ROS). Ifi27l2a overexpression resulted in a significant increase in mitochondria generated ROS level (Fig. 4f) and the percentage of Mitosox + cells (Fig. 4g). The “no-virus” cells (No) showed negligible effect on ROS levels. These data indicate that Ifi27l2a expression alone can cause ROS generation in mitochondria in activated MG, implying a causative role of Ifi27l2a in mitochondrial dysfunction in MG.
Ifi27l2a hemizygous deletion is protective from ischemic brain injury in mice. Given our finding that increased Ifi27l2a expression alone is sufficient to promote microglial activation, we asked if limiting Ifi27l2a expression could reduce microglial activation and brain injury following stroke. We used a permanent distal middle cerebral artery occlusion (pdMCAO) stroke model in WT and Ifi27l2a +/- (Het) mice (2–3 month old, male). At post-stroke day (PSD) 3, the infarct volume was significantly reduced in Het, compared to WT brain (Fig. 5a-b, n = 5 or 6, * p < 0.05). The area of activated MG (Iba1) was also reduced in the primary injury region at PSD 14 (Fig. 5c-d, n = 5 or 6, * p < 0.05). The pdMCAO model 20 is also a well-established model for evaluating secondary injury in stroke; significant gliosis develops in the ipsilateral thalamus several days after the primary injury. We and others have shown significant gliosis in the ipsilateral thalamus 1 or 2 weeks following stroke 11,21,22. Therefore, to evaluate the role of Ifi27l2a in secondary thalamic injury, we examined thalamic gliosis in WT and Het mice (2–3 months old, male) at PSD 14. Evaluation of the ipsilateral thalamus revealed significant reduction in both microgliosis (Fig. 5e-f, n = 6, * p < 0.05) and astrogliosis (Fig. 5g-h, n = 6 ** p < 0.01) in Het mice compared with WT. Note that the reduced injury in Het mice is not due to developmental differences in MCA territory. Analysis of vascular territory between WT and full Ifi27l2a KO revealed no difference (Extended Data Fig. 11, n = 6, p = 0.19). Together, these findings indicate that reducing Ifi27l2a expression can reduce primary and secondary injury associated with ischemic stroke, likely through attenuation of the microglial-mediated inflammatory response.