SorLA expression levels in glioma-associated microglia/macrophages are linked to their activation mode
The activity of SorLA has been especially well documented in neurons, yet recent data suggest that it might also be expressed in other brain cell types in a specific pathological context. In particular, Abdelfattah et al reported high expression levels of the
SORL1 gene in GAMs in human samples (Abdelfattah et al,
2022). In line with this notion, we could indeed detect SorLA in Iba1+ cells in human glioma sections, although not in all studied patient samples (Fig.
1A; Appendix Table
S1). Intrigued by the fact that not all Iba1+ cells in patients specimens stained positive for SorLA, we analyzed the proportions of
AIF1 (encoding for Iba1) expressing cells that also express
SORL1 in several scRNA-seq datasets from GBM samples (Data Ref.: Sankowski et al,
2019; Abdelfattah et al,
2022; Chen et al,
2021; Neftel et al,
2019; Pombo Antunes et al,
2021; Wang et al,
2022b). In all 6 datasets,
SORL1 was expressed in
AIF1+ cells. The percentage of
SORL1+ cells among
AIF1+ cells was variable and ranged from 17.2% to 97.0% depending on the dataset (Appendix Table
S2). Induction of
Sorl1 expression in GAMs was recapitulated in a murine model of glioma (Szulzewsky et al,
2015). Twenty days after GL261 glioma cells implantation,
Sorl1 expression in GAMs (CD11b+ cells) isolated from brain tumors increased more than three times compared to the control cells. At the same time, SorCS2, another VPS10P receptor detected in this dataset, did not show such an induction (Appendix Table
S3). Taken together, these data point to the hypothesis that SorLA levels in microglia/macrophages might be upregulated during activation of these cells toward a tumor-supporting phenotype.
To further investigate
SORL1 expression patterns in GAMs, we re-analyzed scRNA-seq data from human glioma samples (Data Ref.: Abdelfattah et al,
2022), focusing on newly diagnosed GBM samples (ndGBM). From this dataset, we selected 7 clusters that we classified as GAMs based on the expression of marker genes (
AIF1, CD68, ITGAM,
P2RY12, TMEM119, CX3CR1; Fig.
EV1A–C; Appendix Table
S4; Dataset
EV1). As reported by the authors of the study before,
SORL1 expression was limited to this GAMs population (Fig.
EV1B; Appendix Fig.
S1A). The only other cluster with noticeable expression of
SORL1 was characterized by expression of interferon-inducible genes (
IFI6, IFI27, IFITM1, IFITM2, IFITM3) as well as endothelial markers (
EDN1, ABCG2, FLT1, PLVAP, PECAM1) (Appendix Fig.
S1A; Dataset
EV1).
In all subsequent steps, we focused on the population of selected glioma-associated microglia/macrophages (GAMs). These cells were pooled for further analysis to yield 23 GAMs clusters in which
SORL1 expression levels were evaluated (Fig.
1B). To shed light on the potential links between SorLA levels and the functional properties of GAMs, we next investigated the marker genes of five clusters with the highest and five clusters with the lowest
SORL1 levels (Fig.
1C; Dataset
EV2). Among “high-
SORL1” clusters, cluster 7 was characterized by the expression of immediate early genes,
SPP1, a gene associated with tumor-promoting GAMs (Szulzewsky et al,
2015), and
ID2 involved in pro-tumorigenic polarization of myeloid cells (Huang et al,
2017). Cluster 22 showed high expression levels of several microglia/macrophages genes including
TREM2, which was linked before to the pro-tumorigenic properties of tumor-associated macrophages in various cancers (Khantakova et al,
2022). At the same time, among “low-
SORL1” clusters, cluster 9 was characterized by expression of
TLR genes as well as pro-inflammatory cytokines
IL1A and
TNF. Clusters 11 and 17 showed relatively high expression of a pro-inflammatory factor
MIF and several glycolysis-related genes (
PGK1,
ENO1, GAPDH,
LDHA,
PKM). Importantly, a metabolic switch towards glycolysis is a hallmark of pro-inflammatory activation of microglia/macrophages (Lauro and Limatola,
2020).
Furthermore, to provide a more global characteristics of GAMs, we implemented Natural Language Processing (NLP) approach, which allows to identify keywords describing functionally related groups of genes. In doing so, we analyzed marker genes of all GAMs clusters (Datasets
EV2 and
EV3). NLP revealed that genes enriched in “high-
SORL1” clusters were associated with terms as “cancer” (clusters 2, 7, 8, 22) and “angiogenesis” (cluster 12). Concurrently, genes expressed in “low-
SORL1” clusters were described with words like “glycolysis” (clusters 11 and 17), “toll-like receptor” (clusters 9 and 18) or “phagocytosis” (cluster 3). Taken together, based on the results obtained thus far we hypothesized that high and low SorLA levels might be associated with pro-tumorigenic and pro-inflammatory phenotypes of microglia/macrophages, respectively.
To gain deeper insights into the functional relevance of SorLA’s presence in GAMs, we performed Monte-Carlo Feature Selection (MCFS-ID) analysis (Dramiński et al,
2008; Dramiński and Koronacki,
2018) on a single-cell level in the same dataset. MCFS-ID allows to determine features (here expression levels of a given gene) predicting the behavior of another feature (here,
SORL1 expression levels categorized to low, medium, or high). Among 25 top genes from the MCFS-ID, differential levels of gene expression in the context of discretized values of
SORL1 were assessed (Fig.
1D; Dataset
EV4). This analysis highlighted microglia signature genes (
CX3CR1, A2M, C3) and
TREM2 among the best predictors of high-
SORL1 expression levels in GAMs. Moreover, high expression of a gene coding for transcription factor MEF2C, known for its role in restraining microglial inflammatory response (Deczkowska et al,
2017), was also linked to high-
SORL1 transcript levels. Interestingly, we also noted a similar positive association between
SORL1 and
GPR34, coding for a microglial receptor required to maintain microglia in the homeostatic phenotype (Schöneberg et al,
2018). Finally, we identified
LGALS1, S100A10 and
MIF as predictors of low
SORL1 expression in GAMs. While
MIF is a well-established marker of pro-inflammatory activation of microglia/macrophages, the roles of
LGALS1 and
S100A10 in these cells are not entirely clear.
LGALS1 expression in tumor cells has been linked to immunosuppression (Chen et al,
2019), but its induction in microglia might be relevant for inflammatory responses. Thus, increased
LGALS1 expression was observed in microglia after stimulation with LPS (Kiss et al,
2023) and in a subpopulation of disease-associated microglia in multiple sclerosis (Masuda et al,
2019). S100A10 has been mostly studied in other cell types; in macrophages, it seems to be involved in their recruitment upon inflammation (O’Connell et al,
2010).
To further characterize the potential functional interactions between various cell types present in the tumor microenvironment in the context of
SORL1 levels in GAMs, we implemented the CellChat analysis. First, we defined several cell types in the ndGBMs (Data Ref.: Abdelfattah et al,
2022) based on the expression of marker genes (Dataset
EV1 and Appendix Tables
S4–
7). As a result, in addition to GAMs population subdivided into low-, medium- and high-
SORL1 expressing cells, we defined the populations of tumor cells (clusters 0, 7, 8, 9, 15, expressing
CDK4,
MT1X,
ATRX,
CCND2,
MDM2,
SOX4,
CD9,
CDK6,
S100B), lymphocytes (clusters 5, 17, 19;
GZMK,
CD3E,
PTPRC,
CCL5,
IL32,
CD69,
CD52), smooth muscle cells (cluster 11;
ACTA2,
TAGLN), endothelial cells (cluster 14;
EDN1 PECAM1 ANGPT2), oligodendrocytes (cluster 12,
MBP,
CNP) and other cells whose classification was dubious (clusters 4, 18, 20). Cell–cell interaction analysis revealed distinct communication patterns specific for each of these populations (Figs.
2A,B and
EV2A). Tumor cells appeared to be the major contributors to outgoing communication, which at the same time was almost missing for the lymphocytes that mostly exhibited incoming patterns. Of note, GAMs tended to show different communication patterns depending on their
SORL1 expression levels (Figs.
2A,B and
EV2A).
More detailed analysis of the ligand-receptor pairs contributing to these communication patterns revealed several interactions specific for the GAMs with high-
SORL1 expression. This population received CCL5-CCR1 signals from the lymphocytes and ICAM2-(ITGAM + ITGB2) from the endothelial cells, as well as HLA-F–LILRB1 from the lymphocytes (Fig.
2C). These interactions may be associated with enhanced tissue infiltration (Schenkel et al,
2004; Pham et al,
2012) and induction of immunosuppressive GAMs phenotype (Zeller et al,
2023), respectively. In turn, high-
SORL1 GAMs sent pro-tumorigenic signals to the tumor cells (Fig.
2C). In particular, noncanonical Wnt signaling (WNT5A-FZD3) might promote glioma invasion (Pukrop et al,
2010,
2006) and glutamatergic signaling ((SLC1A3 + GLS)–GRIA2) is known to stimulate tumor cells growth, proliferation, and survival (Prickett and Samuels,
2012). Other ligand-receptor pairs revealed by this analysis included TNF-TNFRSF1A sent by medium-
SORL1 expressing GAMs, TNFSF12-TNFRSF12A (high-
SORL1 GAMs to tumor cells), PDGFB-PDGFRA (medium- and high-
SORL1 GAMs to several populations), and THY1-(ITGAM + ITGB2) received by high-
SORL1 GAMs (Fig.
EV2B). In conclusion, CellChat analysis further substantiated our notion that high expression of
SORL1 is one of the features characterizing a distinct GAMs population whose transcriptional profiles point to their pro-tumorigenic potential.
Finally, we repeated key analyzes on an independent dataset published by Neftel et al, (Data Ref.: Neftel et al,
2019). Here,
SORL1 expression was also seen predominantly in GAMs that we selected based on the expression of the marker genes as before for the Abdelfattah et al data (Dataset
EV5; Fig.
EV1D,F; Appendix Fig.
S1B; Appendix Table
S8), but it appeared more uniform and shifted towards higher levels compared to the dataset from Abdelfattah et al (Appendix Fig.
S1C,D). In the next step, GAMs were further grouped into 5 clusters, for which
SORL1 expression levels and the marker genes were analyzed (Appendix Fig.
S1E–H; Dataset
EV6). Marker genes of the two GAMs clusters with the highest
SORL1 expression included
CX3CR1,
P2RY12,
AIF1,
ITGAM,
TMEM119,
TREM2,
CCL3, and
CCL4, while GAMs cluster with the lowest
SORL1 levels was characterized by
LGALS1,
GAPDH,
PGK1,
ENO1,
LDHA,
MIF,
FTH1,
HMOX1, and
TLR4 marker genes (Appendix Fig.
S1H). Moreover, the genes that we previously linked to
SORL1 expression on a single-cell level (Abdelfattah et al data, Fig.
1D) showed similar expression patterns associated with
SORL1 in this additional scRNA-seq dataset (Appendix Fig.
S1I). In conclusion, our key findings on
SORL1 expression patterns in GAMs were recapitulated in this independent analysis.
Taken together, we propose a functional link between the activation status of microglia/macrophages and SORL1 expression levels. In particular, our results point to the scenario where high SORL1 expression occurs in tumor-supportive GAMs, while low SORL1 expression is associated with pro-inflammatory phenotypes of microglia/macrophages.
Loss of SorLA promotes TNFα release from cultured microglia
Since
SORL1 expression appeared related to the functional properties of GAMs, we tested whether SorLA levels might be specifically regulated by the cues triggering diverse microglial phenotypes. To study this phenomenon, we used primary murine microglia treated with LPS or co-cultured with GL261 glioma cells, an in vitro model to mimic pro-inflammatory stimulation and the impact of glioma-secreted factors, respectively. In line with our hypothesis,
Sorl1 expression increased in the presence of glioma cells, while it dramatically decreased upon LPS treatment (Fig.
3A; Appendix Fig.
S2A,B).
Distinct changes in microglial
Sorl1 expression seen upon activation by LPS and in the presence of glioma cells suggested that SorLA might be an active player in shaping functional properties of microglia. SorLA controls the intracellular sorting of target proteins defining plasma membrane transport and secretion properties (Schmidt et al,
2017). As cytokines release from activated microglia is crucial for their activity and response to disease (Colonna and Butovsky,
2017), we profiled cytokines released by WT and SorLA-deficient (SorLA-KO, SLKO) murine microglia upon PMA stimulation to uncover potential factors secreted in a SorLA-dependent manner. We did not observe any global alterations in cytokines secretion from SorLA-KO microglia as compared to WT cells. Several cytokines were released in similar amounts in both genotypes, including IL-9, MCP1, MIP1α, MIP2, and MIP3 (Fig.
EV3). Secretion of MIP1β tended to be decreased in SorLA-KO cells. However, the most remarkable difference was seen in the secretion of the pro-inflammatory cytokine TNFα, which was released in higher amounts from SorLA-deficient microglia (Fig.
3B). This enhanced secretion of TNFα was not due to its increased expression, as mRNA levels were not changed in SorLA-KO cells (Fig.
3C; Appendix Fig.
S2C). These results indicated that the alterations in TNFα release occur post-transcriptionally and might result directly from SorLA-dependent sorting mechanisms present in WTs, but absent in SorLA-KO microglia.
To further establish the relevance of our findings for human cells, we used microglia derived from induced pluripotent stem (iPS) cells (Fig.
3D,E) either wild-type or genetically deficient for
SORL1 (SLKO). As expected, expression of pluripotency markers (
SOX2,
NANOG,
OCT4) dropped during differentiation, while microglia markers (
P2RY12,
TREM2,
AIF1,
CX3CR1,
ITGAM) levels increased (Fig.
3F; Appendix Figs.
S2D and
S3A). Immunodetection of Iba1 and P2RY12 additionally confirmed the microglial identity of generated cells (Fig.
3G). Differentiation was unaltered by SorLA deficiency, as expression levels of marker genes
AIF1,
ITGAM and
P2RY12 were comparable for WT and SorLA-deficient human-induced microglia (iMG, Appendix Figs.
S2E and
S3B). As anticipated, SorLA protein was completely lost from SLKO iMG (Appendix Fig.
S3C). Using this model, we confirmed that LPS stimulation drives a remarkable decrease in
SORL1 expression in WT iMG (Fig.
3H; Appendix Fig.
2F). Moreover, loss of SorLA activity led to an increased TNFα release from iMG (Fig.
3I), indicating that SorLA-dependent control of TNFα secretion is conserved between the species and highlighting its potential relevance for disease pathogenesis.
SorLA binds TNFα to control its intracellular trafficking
Typically, SorLA exerts its functions by binding target proteins and directing their intracellular trafficking. For example, this sorting activity of SorLA was documented for protein sorting between the TGN, endosomes and lysosomes, as well as in the recycling route via the Rab11+ compartment (Schmidt et al,
2016,
2007; Caglayan et al,
2014). To corroborate the potential role of SorLA in TNFα sorting, we first tested the colocalization of the two proteins in microglial cells. Indeed, immunostaining of PMA-stimulated BV2 cells revealed partial overlap of SorLA and TNFα signals (Fig.
4A,B). Next, we examined the interaction of SorLA with TNFα in co-immunoprecipitation (co-IP) assays. Towards this end, we overexpressed SorLA and GFP-tagged TNFα (or GFP alone) in HEK293 cells and pulled down the GFP tag. SorLA was present in the immunoprecipitate containing TNFα-GFP, while it was not visible in the control GFP-IP (Fig.
4C), supporting our notion that TNFα is a SorLA ligand.
As the structure of SorLA entails several domains capable of cargo binding (Fig.
4D), we sought to identify the domain responsible for the interaction with TNFα. Using deletion mutants lacking particular SorLA domains (Fig.
EV4A) in our co-IP experiments, we observed that removing EGF-type repeat and the β-propeller (ΔEGF/βP mutant) tended to weaken SorLA binding to TNFα, although these results did not reach statistical significance (Fig.
EV4B,C). Of note, these experiments did not rule out additional binding sites outside the EGF-type repeat and the β-propeller for TNFα in SorLA. Using more stringent co-IP conditions (300 mM NaCl) in order to increase the specificity of our results led to loss of binding of TNFα by full-length SorLA (Appendix Fig.
S4). As an alternative approach, we used myc-tagged mini-receptors composed exclusively of particular SorLA domains (Fig.
EV4A; Appendix Table
S9). In line with our prior observations, the most efficient co-IP with TNFα was noted for the mini-receptor encompassing the EGF-type repeat and β-propeller (EGF/βP, Fig.
4E,F). In summary, it is plausible that SorLA binds TNFα predominantly via an extracellular motif containing EGF-type repeat and β-propeller.
Finally, we elucidated the impact of SorLA on the intracellular trafficking of TNFα. We analyzed its colocalization with the markers of subcellular compartments in WT and SorLA-deficient primary microglia stimulated with PMA. We did not observe remarkable TNFα presence in lysosomes (stained with anti-Lamp1) in any of the genotypes (Appendix Fig.
S5). Rather, TNFα was colocalizing with the Golgi (GM130-positive structures), Rab7+ late-endosomes, Rab11+ recycling endosomes, as well as with Vti1b, known for its important role in the TNFα secretory route (Murray et al,
2005). SorLA deficiency did not affect the presence of TNFα in the GM130 + , Lamp1+ and Rab7+ compartments (Appendix Fig.
S5), but it increased the colocalization of TNFα with Vti1b and caused a concurrent loss of TNFα from Rab11+ endosomes (Fig.
4G). These results indicated that loss of SorLA shifts the trafficking of TNFα toward the secretory pathway, which could explain the increased TNFα release from SorLA-KO microglia.
Loss of SorLA limits glioma growth, promotes inflammation and necroptosis
SorLA emerged as a critical factor controlling the functional properties of microglia, restricting their pro-inflammatory activities. Moreover, in glioma patients, SORL1 expression levels in GAMs were related to their transcription profiles. We further speculated that the presence of SorLA might have an impact on the functional properties of GAMs and, consequently, on tumor microenvironment and glioma progression.
Thus, we asked whether SorLA presence in the host cells has an impact on glioma progression in a murine model. Glioma GL261 cells carrying luciferase and tdTomato transgenes were implanted to the striata of WT and SorLA-KO mice, and the tumor growth was followed for 21 days. Using this model, we showed that glioma growth is limited in SorLA-deficient mice (Fig.
5A,B), supporting our notion that the presence of this sorting receptor in host cells is critical for establishing a tumor-promoting microenvironment.
To evaluate the combined impact of the developing glioma and SorLA loss from host cells on the tumor microenvironment, we first assayed the levels of selected cytokines (TNFα, MIP2, CCL5, CXCL1, IL1β, IL2, IL6, and IL10) in the tissue lysates derived from tumor-bearing and tumor-free hemispheres. At 14 days post-implantation, we did not document any major induction of the cytokines (Appendix Fig.
S6A). Twenty-one days after implantation, induction of all cytokines in glioma-bearing hemispheres was visible and comparable between genotypes, except for MIP2, which was decreased in the SLKO tumor samples as compared to WTs (Appendix Fig.
S6B).
Next, we evaluated the properties of microglia in the glioma model in WT and SLKO mice. The microglia activation status is reflected by the changes in their morphology (Morrison et al,
2017; Franco-Bocanegra et al,
2021; Kvisten et al,
2019). In essence, the tumor-supportive phenotype is characterized by ramified morphology with longer and more branched processes. By contrast, pro-inflammatory microglia present compact morphology and shorter extensions. These features can be evaluated by Sholl analysis, which quantifies the number of processes crossing the spheres of increasing radius, centered at the cell soma. This analysis performed on the tumor-surrounding Tmem119+ cells revealed remarkable differences between WT and SorLA-KO microglia (Fig.
5C,D). In detail, SorLA-KO microglia showed a compact morphology with shorter processes, which can be attributed to its more pro-inflammatory phenotype, while a branched morphology of microglia observed in WTs can correspond to a homeostatic or tumor-supporting state. These differences between the genotypes were also reflected by an apparent global decrease in Tmem119 signal in the tumor-surrounding tissue in SorLA-KO mice (Appendix Fig.
S7A,B). Of note, in the contralateral glioma-free hemisphere, we did not observe any genotype-dependent alterations in microglia morphology (Appendix Fig.
S7C,D).
To further verify the hypothesis that the pro-tumorigenic activities are blunted in the SorLA-KO animals, we focused on the phosphorylation status of STAT3 in the glioma-bearing brains. STAT3 is a transcription factor, which, when phosphorylated, activates the expression of multiple genes related to pro-tumorigenic properties of GAMs (De Boeck et al,
2020; Dumas et al,
2020). It was demonstrated that inhibition of STAT3 enhances pro-inflammatory potential of these cells, which results in suppression of tumor growth in a murine model of glioma (Zhang et al,
2009). In line with our hypothesis, the levels of p-STAT3 were remarkably reduced in glioma-bearing hemispheres of SorLA-deficient mice as compared to WTs (Fig.
5E,F).
An important consequence of the inflammatory response is the influx of peripheral immune cells into the affected tissue. Locally released factors attract circulating leukocytes and promote their migration, eventually driving their infiltration into the inflamed area. In GL261 gliomas, the infiltration of galectin-3+ macrophages and CD8 + T lymphocytes into the tumor mass was similar for both WT and SorLA-KO mice (Fig.
6A–C). Also the staining for the common GAMs marker Iba1 did not reveal any genotype-dependent differences in terms of glioma infiltration (Appendix Fig.
S8). However, we noted a striking genotype-dependent difference in the neutrophil influx into the glioma. Thus, infiltration of the MPO+ neutrophils was evident in the gliomas in SorLA-KO brains, while it did not occur in the WTs and in the contralateral hemispheres (Fig.
6D,E). This was not due to the overall increase in neutrophil amounts in the circulating blood of SorLA-KO mice, as the numbers of circulating neutrophils, as well as of erythrocytes, monocytes, lymphocytes, basophils, and eosinophils, were comparable in glioma-bearing WT and SorLA-KO mice (Appendix Fig.
S9). We propose that this massive neutrophil infiltration is a direct consequence of a pro-inflammatory milieu promoting their migration into the brain parenchyma in SorLA-deficient mice. One of the key mechanisms facilitating neutrophils influx into the tissue involves TNFα-driven induction of ICAM-1, an adhesion molecule critical for the transendothelial migration of these cells (Peterson et al,
2006; Yang et al,
2005). Increased soluble ICAM-1 (sICAM1) levels are also a well-established hallmark of inflammation (Bui et al,
2020). As anticipated from our data, sICAM1 was elevated in the tumor-containing hemispheres derived from SorLA-KO mice as compared to the WTs (Fig.
6F). These results strongly supported our hypothesis that loss of SorLA shifts the properties of the glioma microenvironment toward pro-inflammatory.
Finally, to further elucidate the mechanisms limiting tumor growth in SorLA-KO mice, we focused on cell death mechanisms that might be activated by TNFα itself, or by the infiltrating neutrophils. TNFα can trigger apoptosis or necroptosis via its receptor TNFR1 (Webster and Vucic,
2020), while neutrophils elicit ferroptosis (Yee et al,
2020). We thus checked which of these pathway(s) are activated in glioma specifically in SorLA-KO mice. We did not observe induction of apoptosis, as the cleavage of PARP and caspase-3 was negligible and similar for both WT and SorLA-deficient mice (Fig.
7A,B). Induction of ferroptosis was not visible either (Fig.
7C). At the same time, we noted increased levels of necroptosis markers p-RIP1 and p-RIP3 in the glioma-bearing hemispheres from SorLA-KO mice, as compared to the WTs (Fig.
6D,E). In line with these observations, we further documented a time-dependent increase of p-RIP1 in cultured glioma cells treated with TNFα (Fig.
7F). These results suggested that necroptosis might contribute to the elimination of glioma cells and, in consequence, to limiting tumor growth in SorLA-KO brains.