Abstract Genomic alteration can reshape tumor microenvironment to drive tumor malignancy. However, how PTEN deficiency influences microenvironment-mediated cell-cell interactions in glioblastoma (GBM) remains unclear. Here, we show that PTEN deficiency induces a symbiotic glioma-M2 macrophage interaction to support glioma progression. Mechanistically, PTEN-deficient GBM cells secrete high levels of galectin-9 (Gal-9) via the AKT-GSK3β-IRF1 pathway. The secreted Gal-9 drives macrophage M2 polarization by activating its receptor Tim-3 and downstream pathways in macrophages. These macrophages, in turn, secrete VEGFA to stimulate angiogenesis and support glioma growth. Furthermore, enhanced Gal-9/Tim-3 expression predicts poor outcome in glioma patients. In GBM models, blockade of Gal-9/Tim-3 signaling inhibits macrophage M2 polarization and suppresses tumor growth. Moreover, α-lactose attenuates glioma angiogenesis by down-regulating macrophage-derived VEGFA, providing a novel antivascularization strategy. Therefore, our study suggests that blockade of Gal-9/Tim-3 signaling is effective to impair glioma progression by inhibiting macrophage M2 polarization, specifically for PTEN-null GBM. __________________________________________________________________ PTEN-deficient glioma promotes macrophage M2 polarization via Gal-9/Tim-3 signaling, serving as a viable target in glioblastoma. INTRODUCTION Genomic profiling studies have indicated that glioblastoma (GBM) can be classified into four subtypes, including the mesenchymal, classical, neural, and proneural subtypes ([50]1). Among the four subtypes, mesenchymal GBM exhibits the most abundant stromal components ([51]2), most of which are tumor-associated macrophages (TAMs) and they generally appear to be type 2 polarized macrophages (M2) ([52]3). TAM populations are bone marrow–derived infiltrating myeloid cells ([53]4) and constitute as much as 30 to 50% of all cells in human GBM tumors ([54]5). This cell population plays a key role in glioma progression. Mesenchymal GBM is a high-grade malignant tumor that typically harbors gene mutations in phosphate and tension homology deleted on chromosome ten (PTEN), TP53, and NF1 ([55]1). PTEN deletion or mutation results in enhanced recruitment of macrophages into the tumor microenvironment (TME) ([56]6), suggesting that macrophage-targeted therapy may specifically work for PTEN-null glioma. The interplay between cancer cells and macrophages plays a crucial role in tumor development, with functions in supporting angiogenesis, nurturing cancer stem cells, and promoting an immunosuppressive TME ([57]7, [58]8). The TME is shaped by tumor cell–intrinsic signaling pathways and secreted factors ([59]9, [60]10). Some studies reported that CD163^+ macrophages are essential for promoting angiogenesis by expressing high levels of vascular endothelial growth factor A (VEGFA) ([61]11), and VEGFA derived from tumor-infiltrating myeloid cells is important for initiating vascularization in gliomas ([62]12). Hence, macrophage-targeted strategies might be a novel therapeutic antivascularization approach. Now, immunotherapy has not been widely applied to glioma because of the unique immune microenvironment characterized by a paucity of T cells but abundant M2-like macrophages ([63]13). Many approaches have been attempted to eliminate TAMs or reprogram macrophages to perform antitumor functions ([64]14). One strategy used colony-stimulating factor 1 receptor (CSF1R) inhibition to block glioma progression by reducing macrophage M2 polarization ([65]15), but acquired resistance was observed in animal model experiments ([66]16) and limited efficacy was found in clinical trials ([67]17). Thus, identifying other strategies to improve the effectiveness of macrophage-targeted therapies for glioma patients is critical. Given that PTEN-null glioma can recruit a large number of macrophages, clarifying how PTEN deficiency in glioma cells modulates macrophage M2 polarization might provide new targets for precise immunotherapy in specific GBM subtypes or genotypes. In our study, we explored whether and how PTEN alterations in GBM cells influence macrophage polarization, and how the infiltrating macrophages in turn function to support the growth and angiogenesis of glioma. Our aim is to identify practical therapeutic targets functioning at the maintenance of the symbiotic glioma-macrophage interactions. RESULTS PTEN deficiency facilitates macrophage infiltration and macrophage M2 polarization in GBM To assess the role of PTEN in modulating macrophage chemoattraction and polarization, we conducted a series of functional studies in PTEN–wild-type (PTEN-WT) and PTEN-deleted/mutated (PTEN-Del/Mut) glioma models. First, we evaluated PTEN expression in 23 GBM cell lines by immunoblot analysis and found that PTEN expression was undetectable in U118, U87, U138, U251, SYU354, SF295, U373, and MGR3 cell lines ([68]Fig. 1A). By using Sanger sequencing technology, we confirmed the mutation status of PTEN gene in all glioma cell lines used in our study (table S1). We cultured the 23 GBM cell lines in fetal bovine serum (FBS)–free medium for 24 hours and collected the supernatants for use as glioma cell–derived conditioned medium (GCM) in subsequent experiments. Fig. 1. PTEN deficiency facilitates macrophage infiltration and M2 polarization. [69]Fig. 1. [70]Open in a new tab (A) Immunoblots for PTEN in 23 glioma cell lines. (B) Quantification of relative migration of THP-1 macrophages following stimulation with CM from eight PTEN-null cell lines (U118, U87, U138, U251, SYU354, SF295, U373, and MGR3) and eight PTEN–wild-type (WT) GBM cell lines (SKMG-1, LN229, SYU687, MGR2, LNZ308, GL261, SHG44, and T98). n = 3 biological replicates. (C) Immunoblots showing PTEN expression status in PTEN-deleted (Del) U87/U118 cells after PTEN overexpression (OE) and PTEN-WT SKMG-1/LN229/GL261 cells after PTEN knockout (KO). (D and E) Flow cytometry analysis of CD163 expression in BMDMs treated with CM from PTEN-Del/OE U87 cells and PTEN-WT/KO GL261 cells. (F and G) Immunofluorescence staining of CD86 and CD163 was determined and quantified in intracranial PTEN-WT/KO GL261 and PTEN-Del/OE U87 xenograft tumors. PTEN-WT/KO GL261 xenografts were established in C57BL/6J mice, and PTEN-Del/OE U87 xenografts were established in BALB/c-nu/nu mice. Scale bar, 200 μm. **P < 0.01 and ***P < 0.001. Transwell migration assays showed that more macrophages migrated in GCM from eight PTEN-Del GBM cell lines, compared with GCM from eight PTEN-WT GBM cell lines ([71]Fig. 1B and fig. S1A). We then knocked out the PTEN gene (PTEN-KO) in the PTEN-WT cell lines SKMG-1, LN229, and GL261 using CRISPR technology and overexpressed WT PTEN (PTEN-OE) in the PTEN-Del cell lines U87 and U118 ([72]Fig. 1C). Transwell migration assays using these cell lines confirmed that GCM from PTEN-KO SKMG-1, LN229, and GL261 cells greatly enhanced macrophage migration compared with GCM of the parental PTEN-WT cells, while GCM from U87 and U118 (PTEN-Del) cells reduced macrophage migration after WT PTEN restoration (fig. S1, B and C). To figure out how PTEN affected macrophage polarization, flow cytometry and quantitative real-time polymerase chain reaction (qRT-PCR) were used to evaluate M1 and M2 markers in macrophages treated with GCM. As compared with GCM from PTEN-normal cells (GL261-WT and U87-OE), the expression level of M2 marker CD163 on bone marrow–derived macrophages (BMDMs) was enhanced by GCM from PTEN-deficient cells (GL261-KO and U87-Del) ([73]Fig. 1, D and E, and fig. S1D). In addition, the expression level of the M1 marker CD86 on BMDM and THP-1 macrophages was down-regulated by GCM derived from PTEN-deficient cells (fig. S1E). The qRT-PCR results indicated that GCM obtained from PTEN-normal glioma cells seemed to induce higher expression of M1 markers (IL1, IL12, IL23, and TNF-α) in macrophages, compared with GCM obtained from PTEN-deficient cells (fig. S1F). Consistently, compared to PTEN-normal xenografts (GL261-WT and U87-OE) grown in vivo, PTEN-deficient xenografts (GL261-KO and U87-Del) showed faster growth in subcutaneous mouse models, shortened the survival time of orthotopic mouse models (fig. S1, G and H), and enhanced macrophage M2 polarization with higher CD163 and lower CD86 expression scores ([74]Fig. 1, F and G). Together, these data demonstrate that PTEN-deficient GBM cells facilitate macrophage infiltration and promote macrophage M2 polarization more efficiently than PTEN-WT GBM cells. Gal-9 is preferentially secreted by PTEN-deficient glioma cells and promotes macrophage M2 polarization To identify the secreted proteins that modulated macrophage M2 polarization in PTEN-deficient GBM, we examined putative macrophage-recruiting factors and macrophage polarization–related factors exhibiting a >1-fold change in PTEN-KO LN229 cells according to a secreted protein database ([75]18) (table S2). Transcriptome and qRT-PCR analyses of LN229-WT and LN229-KO cells showed increased expression in PTEN-KO cells for LGALS9, IL6, VEGFC, SPP1, TGFB1, and TGFB2, with LGALS9 [which encodes the galectin-9 (Gal-9) protein] showing a marked increase ([76]Fig. 2, A and B). Similarly, upon PTEN deletion, LGALS9 mRNA levels were increased to the greatest extent among these six genes in other PTEN-WT GL261 and SKMG-1 cells (fig. S2A). We further observed that genes encoding macrophage polarization–related factors (LGALS9, IL6, TGFB1, and TGFB2) were down-regulated upon PTEN rescue in U87 cells, with the LGALS9 gene showing a marked decrease (fig. S2B). Moreover, we used Quantibody Human Cytokine Antibody Array 440 to perform a more comprehensive proteomic analysis of the CM from LN229-KO and LN229-WT cells. We screened out the top 22 factors among the 440 secreted proteins evaluated in the array (LN229-KO CM/LN229-WT CM > 3) (fig. S2C). Next, among the 22 factors, 5 macrophage M2 polarization–related factors {Gal-9, macrophage migration inhibitory factor (MIF), latency-associated peptide (LAP) [transforming growth factor–β1 (TGF-β1)], brain-derived neurotrophic factor (BDNF), and basic fibroblast growth factor (bFGF)} were chosen for further validation. We found that human recombinant Gal-9 protein was much easier than other four cytokines to induce macrophage M2 polarization, within higher production of interleukin-10 (IL-10) and TGF-β1 by THP-1 macrophages (fig. S2D). To further confirm how PTEN deficiency affects the cytokine profiles of GBM cells, we compared the cytokines secreted by PTEN-deficient and PTEN-normal GBM cells (PTEN-WT versus KO SKMG-1 cells; PTEN-Del versus OE U87 cells) using antibody microarrays. Compared with PTEN-normal cells, we noticed that PTEN deficiency did not cause notable changes in the expression of some macrophage polarization–related factors [interferon-γ (IFN-γ), tumor necrosis factor–α (TNF-α), IL-4, IL-10, TGF-β, etc.] by GBM cells (U87 and SKMG-1 KO cells) ([77]Fig. 2C and fig. S2E). Unexpectedly, enzyme-linked immunosorbent assay (ELISA) results confirmed that the GCM of PTEN-KO or PTEN-Del cells showed markedly high Gal-9 protein levels, while the GCM of PTEN-WT or PTEN-OE cells exhibited low levels ([78]Fig. 2D). Consistently, LGALS9 mRNA levels were up-regulated upon CRISPR-mediated deletion of PTEN in PTEN-WT GL261, SKMG-1, and LN229 cells, and it was down-regulated by reexpression of PTEN in PTEN-Del U87 and U118 cells (fig. S2F). Moreover, by analyzing with Cancer Cell Line Encyclopedia datasets, we noticed that the LGALS9 mRNA levels of 25 PTEN-Del/Mut GBM cell lines were significantly higher than those of 26 PTEN-WT GBM cell lines (fig. S2G), which was similar to the Gal-9 secretion pattern in the GBM cell lines cultured in our laboratory (fig. S2H). The relationship between Gal-9 and PTEN was ultimately confirmed by immunoblotting ([79]Fig. 2E) and further reinforced by the positive correlation between phospho-AKT and Gal-9 in the indicated glioma cell lines ([80]Fig. 2F). In addition, eight patient-derived short-term glioma stem cell lines (GSCs) were used to verify the up-regulation of Gal-9 in PTEN-negative GSCs ([81]Fig. 2G). Fig. 2. Gal-9, a modulator of macrophage M2 polarization, is abundantly secreted by PTEN-deficient glioma cells. [82]Fig. 2. [83]Open in a new tab (A) Transcriptome analyses of up-regulated genes encoding secreted proteins in PTEN-KO LN229 cells. Values are expressed as the fold change between PTEN-KO and PTEN-WT LN229 cells. The top 11 genes (with fold changes of >2.4) were chosen for further validation. (B) qRT-PCR validation of the 11 genes and the TNF gene in PTEN-KO/WT LN229 cells. Values are expressed as the fold change between PTEN-KO and PTEN-WT LN229 cells after normalization to the housekeeping gene GAPDH. (C) Macrophage-inducible factors in PTEN-KO/WT SKMG-1 cells and PTEN-Del/OE U87 cells were detected using antibody microarrays. (D) ELISA analysis of GCM showed Gal-9 secretion up-regulated by PTEN-KO in LN229/SKMG-1 cells and down-regulated by PTEN-OE in U87/U118 cells. **P < 0.01 and ***P < 0.001, Student’s t test. (E and F) Immunoblots of Gal-9 and PTEN or p-AKT expression in the indicated glioma cell lines. (G) Immunoblots for PTEN and Gal-9 in cell lysates of eight short-term patient-derived glioma stem cell lines (GSCs): GSC-1, GSC-#363, GSC-#624, GSC-11, GSC-#354, GSC-#687, GSC-#481, and GSC-#530. (H) Flow cytometry analysis of CD163 and CD206 on THP-1 macrophages treated with U87 CM or recombinant Gal-9 protein (50 ng/ml). (I) Immunoblots showing successful knockdown of Gal-9 in U87 cells by shGal-9. (J) Flow cytometry analysis of CD163 and CD206 on THP-1 macrophages treated with U87 CM or U87 shGal-9#1 CM. (K) Flow cytometry analysis of CD163 and CD206 on THP-1 macrophages treated with U87 shGal-9#1 CM or U87 shGal-9#1 CM supplemented with α-lactose (40 μM). To validate the capacity of the Gal-9 protein to function as a modulator of macrophage M2 polarization in vitro, we performed flow cytometry and short hairpin RNA (shRNA)–mediated Gal-9 knockdown experiments and observed that recombinant human Gal-9 protein–supplemented medium induced M2 marker CD163 and CD206 expression in macrophages, demonstrating the same effect as U87 cell-derived CM ([84]Fig. 2H and fig. S3A). Together, CM obtained from PTEN-Del U87 cells expressing Gal-9 shRNA ([85]Fig. 2I) significantly inhibited M2 markers CD163 and CD206 expression levels ([86]Fig. 2J and fig. S3B), and also inhibited macrophage migration rate (fig. S3C), compared with the control GCM. In addition, Gal-9 concentrations in the CM from 16 GBM cell lines exhibited a strong positive correlation with the number of migrating macrophages (fig. S3D), and elevated concentrations of human recombinant Gal-9 protein obviously increased the migration rate of macrophages (fig. S3E). α-Lactose (40 μM), a Gal-9 inactivator, negligibly inhibited CD163 and CD206 expression further in CM derived from U87 cells expressing Gal-9 shRNA ([87]Fig. 2K and fig. S3F). In general, these results suggest that up-regulated Gal-9 expression in PTEN-deficient GBM cells is strongly associated with macrophage M2 polarization and macrophage migration. Gal-9 is up-regulated by PTEN deficiency via the AKT-GSK3β-IRF1 pathway in glioma cells To explore PTEN regulation of Gal-9 protein expression, a Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was performed to catalog signaling pathways modulated by PTEN deletion. We found that the phosphatidylinositol 3-kinase (PI3K)–AKT signaling pathway was the top pathway activated in PTEN-KO LN229 cells (fig. S4A), which was consistent with other studies ([88]6), reporting that AKT, SRC, NOTCH1, and YAP1 pathways were enriched in PTEN-null glioma cells compared with PTEN-WT controls. To investigate the relevance of these pathways in regulating Gal-9, PTEN-KO LN229 cells were treated with various pathway inhibitors, including the AKT inhibitor MK2206, SRC inhibitor KX2-391, NOTCH1 inhibitor LY3039478, and YAP1 inhibitor verteporfin. In PTEN-KO LN229 cells, the AKT inhibitor, rather than other inhibitors, significantly inhibited Gal-9 protein expression and mRNA expression (fig. S4, B and C). On the other hand, the AKT activator SC 79 up-regulated AKT phosphorylation, and then Gal-9 expression increased in PTEN-WT cells (fig. S4D). Moreover, the AKT inhibitor suppressed Gal-9 expression in dose- and time-dependent manners in PTEN-KO cells ([89]Fig. 3, A and B). Glycogen synthase kinase 3β (GSK3β) and mammalian target of rapamycin (mTOR) are two downstream molecules of the AKT pathway, and we noticed that the GSK3β inhibitor tideglusib up-regulated Gal-9 levels in PTEN-KO SKMG-1 cells in a dose-dependent manner, while the mTOR inhibitor rapamycin had no regulatory effect on Gal-9 expression (fig. S4E). AKT activator SC 79 could inhibit GSK3β activity by up-regulating p-GSK3β^Ser9 levels in PTEN-WT cells (fig. S4F). In contrast, the AKT inhibitor MK2206 reduced p-GSK3β^Ser9 levels ([90]Fig. 3, A and B) and up-regulated p-GSK3β^Try216 levels (fig. S4G) in PTEN-KO glioma cells, resulting in the switch of more GSK3β to its active state. We also observed that PTEN-KO cells exhibited higher levels of p-GSK-3β^Ser9 and thus repression of GSK3β activity compared to PTEN-WT cells (fig. S4H). Therefore, the inactivation of GSK3β mediated by the AKT pathway may contribute to Gal-9 up-regulation. Fig. 3. PTEN deficiency–induced Gal-9 production is regulated via the AKT-GSK3β-IRF1 pathway. [91]Fig. 3. [92]Open in a new tab (A and B) Immunoblots of Gal-9, AKT, p-AKT^Ser473, and p-GSK3β^Ser9 in PTEN-KO LN229/SKMG-1 cells treated with the AKTi MK2206 at different concentrations and time points. (C and D) Immunoblots of p-GSK3β^Ser9, GSK3β, p-GSK3α^Tyr279, p-GSK3β^Tyr216, IRF1, and Gal-9 in PTEN-KO LN229 cells treated with tideglusib at different concentrations and for different time lengths. (E) Immunoblots of IRF1 and lamin B1 in the cytoplasmic and nuclear components of PTEN-KO LN229 cells treated with different concentrations of tideglusib. (F) Immunoblots of Gal-9 and IRF1 in PTEN-KO LN229/SKMG-1 cells transfected with siIRF1 or control siRNA (siNC). (G) A dual-luciferase reporter system containing the 2.0/1.5/0.5-kb human LGALS-9 promoter DNA fragments was used to predict their transcription activity in indicated glioma cells. The values are expressed as the ratio of Firefly and Renilla luciferase signals. ***P < 0.001, one-way ANOVA. (H) The transcriptional activity of the 0.5-kb human LGALS-9 promoter DNA fragment was detected in PTEN-KO LN229/SKMG-1 cells transfected with siIRF1 or siNC. ***P < 0.001, one-way ANOVA. Band density of immunoblots was determined by ImageJ software, and values represent the expression levels after normalization to the density of GADPH/β-tubulin. To determine how the AKT pathway transcriptionally regulates Gal-9 expression, we used the USCS genome browser to predict the potential transcription factors (TFs) that bind to the promoter region of the LGALS-9 gene (fig. S5A). The top five TFs with the greatest promoter-binding potential—EHF, ZNF460, Sox11, IRF1, and ZNF684—were chosen to determine their putative binding sites in the promoter region of LGALS-9 using an in silico promoter region screening approach (JASPAR) (fig. S5B). Among the five TFs, only IRF1 was specifically highly expressed in mesenchymal subtype GBM in the Chinese Glioma Genome Atlas (CGGA) database (fig. S5C). qRT-PCR results indicated that only IRF1 mRNA levels were significantly up-regulated by PTEN-KO in LN229 cells, and EHF, ZNF460, IRF1, and ZNF684 were all reduced by PTEN-OE in U87 cells (fig. S5D). LGALS-9 expression positively correlated with IRF1 expression in the CGGA (fig. S5E) and Gene Expression Profiling Interactive Analysis (GEPIA) glioma samples (fig. S5F). Then, we focused on IRF1 as the most possible TF regulating LGALS-9 gene expression and conducted immunoblot analysis of Gal-9 and IRF1 in PTEN-WT or KO LN229 and SKMG-1 cells treated with GSK3β inhibitors. Previous studies have validated that active GSK3β promotes IRF1 degradation, which led to less IRF1 in cytoplasm entering the nucleus ([93]19). In our study, it showed that the expression levels of IRF1 and Gal-9 were increased by tideglusib in both LN229 KO cells ([94]Fig. 3, C and D) and SKMG-1 WT cells (fig. S6A) in a dose- or time-dependent manner upon up-regulation of p-GSK3β^Ser9. We also used another GSK3β inhibitor, LiCL, to examine whether it could phenocopy the effect of AKT activator SC 79 on Gal-9 and IRF1 expression. The results showed that LiCL could up-regulate the levels of p-GSK3β^Ser9, IRF1, and Gal-9 in both PTEN-KO and PTEN-WT cells (fig. S6B). In addition, the AKT activator SC 79 could also up-regulate IRF1 levels, similar with PTEN-KO effect in PTEN-WT cells (fig. S6C). Furthermore, IRF1 protein expression was up-regulated by tideglusib in the nuclei of PTEN-KO LN229 cells in a dose-dependent manner ([95]Fig. 3E). Therefore, high levels of the inactive GSK3β (p-GSK3β^Ser9) contributed to Gal-9 and IRF1 up-regulation. To assess the role of IRF1 in regulating Gal-9 expression directly, we silenced IRF1 using small interfering RNA (siRNA) in PTEN-deficient GBM cells, which showed marked suppression of Gal-9 expression, compared with the control group ([96]Fig. 3F). To identify the promoter region involved in IRF1-mediated regulation of LGALS-9 expression, we used a series of dual-luciferase reporter vectors containing various fragments of the human LGALS-9 promoter (2.0/1.5/0.5 kb). In PTEN-KO LN229/SKMG-1 cells, the Firefly luciferase activity driven by the 0.5-kb fragment was higher than that of the 1.5-kb fragment; the full-length 2.0-kb fragment showed a lower ratio of Firefly/Renilla luciferase signals, perhaps because of low transfection efficiency ([97]Fig. 3G). Moreover, the Firefly/Renilla values of the 0.5-kb fragment were significantly down-regulated by PTEN expression rescue in U87 cells ([98]Fig. 3G). Further, si-IRF1 obviously inhibited the expression of the Firefly luciferase reporter (0.5 kb), suggesting that IRF1 truly enhanced LGALS-9 transcription via binding to its promoter at 0.5-kb promoter region ([99]Fig. 3H), consistent with the predicted binding sites by JASPAR. Together, the above results indicate that Gal-9 production in PTEN-null glioma cells is modulated through enhanced AKT activation that suppresses IRF1 degradation by phosphorylating GSK3β into inactive form, leading to maintained IRF1 activation and thereby Gal-9 transcription. Multilayer signaling network reveals Gal-9/Tim-3–mediated interactions between glioma cells and macrophages To further figure out how glioma-secreted Gal-9 promoted macrophage polarization, we leveraged the single-cell RNA sequencing (scRNA-seq) data of human GBM tissues from three patients (PTEN-null: M47; PTEN-WT: M48 and M58) (fig. S7, A and B) to infer cell-cell interactions. The cells were clustered and annotated into seven types ([100]Fig. 4A)—glioma cells (malignant), macrophages, oligodendrocytes, T cells, fibroblasts, neutrophils, and endothelial cells—according to marker gene expression (fig. S8) and copy number variation scores (fig. S9). The fraction of macrophages in the PTEN-null sample was larger than that in the PTEN-WT samples (fig. S10), consistent with the aforementioned results ([101]Fig. 1B). On the basis of the scRNA-seq data, we developed a multilayer network approach to infer inter- and intracellular signaling networks (see Materials and Methods). Cell-cell communication networks ([102]Fig. 4B) globally depicted the intercellular ligand-receptor (LR) interactions between the above cell types. Abundant LR interactions between malignant glioma cells and macrophages were observed in both PTEN-null and PTEN-WT samples. However, compared to the PTEN-WT samples, Gal-9 was found to interact with T cell immunoglobulin and mucin domain 3 (Tim-3), a receptor of macrophages, specifically in the PTEN-null sample that expressed high levels of Gal-9 and Tim-3 ([103]Fig. 4C and fig. S7B). Moreover, the multilayer network analysis ([104]Fig. 4D) revealed that Gal-9/Tim-3 signaling activated several TFs (e.g., STAT1, ESR1, and ELK1) in macrophages, and the downstream target genes involved many marker genes related to macrophage polarization [e.g., HMOX1, PPARD, CEBPA, and ADCY7 genes ([105]16)]. Fig. 4. scRNA-seq data analysis reveals Gal-9/Tim-3–mediated interactions between glioma cells and macrophages. [106]Fig. 4. [107]Open in a new tab (A) Distribution of single cells’ gene expression colored by cell types or samples. On the basis of the expression of marker genes (fig. S8), seven cell types were identified. (B) Cell-cell communication networks for PTEN-null and PTEN-WT samples. The color of the dot represents the cell type, the edge represents the communication between the two cell types whose thickness represents the number of downstream target genes of upstream activated ligand-receptor (LR) pairs, and color represents the direction of cell communication (e.g., the purple edge represents the signals delivered by malignant glioma cells). (C) Stacked violin plots of gene expressions of Gal-9 and Tim-3 in two groups of samples (PTEN-null and PTEN-WT). (D) Gal-9/Tim-3 interaction between glioma cells and macrophages inferred by the multilayer network revealed that macrophage M2 marker genes were regulated by Gal-9/Tim-3 signaling through the downstream pathways and transcription factors (TFs) (e.g., ESR1, ELK1, and STAT1). (E) GO enrichment analysis for the Gal-9/Tim-3 downstream target genes. (F) GSEA enrichment analysis for the ranked gene lists in macrophages according to fold changes of gene expression in the PTEN-null sample compared with PTEN-WT samples. Furthermore, we performed functional enrichment analysis to investigate the functions of the downstream target genes of Gal-9/Tim-3 in the multilayer network. The Gal-9/Tim-3–regulated target genes were significantly enriched in Gene Ontology (GO) biological processes including macrophage activation (polarization) or vascular growth and development ([108]Fig. 4E). Consistently, gene set enrichment analysis (GSEA), a functional class scoring analysis, showed that highly expressed genes in macrophages in the PTEN-null sample relative to the PTEN-WT samples were positively correlated with the gene sets related to macrophage activation, as well as vascular growth and development ([109]Fig. 4F). These results imply that Gal-9/Tim-3–mediated glioma-macrophage interactions play an important role in macrophage polarization and angiogenesis. Enhanced Gal-9/Tim-3 signaling predicts poor prognosis in glioma patients To ascertain the clinical significance of Gal-9 and its receptor Tim-3 in glioma, we evaluated Gal-9 and Tim-3 expression in human glioma tissues. In the CGGA database, Gal-9 and Tim-3 expressions were analyzed in four GBM subtypes, and we found that Gal-9 and Tim-3 expression in the mesenchymal GBM subtype was the highest among the four subtypes (fig. S11A). Patients with mesenchymal subtype GBM exhibited significantly poorer survival than those with nonmesenchymal subtype GBM (fig. S11B). We also observed positive correlations of Tim-3, Gal-9, and CD68 expression, suggesting the importance of Gal-9/Tim-3 signaling up-regulation in glioma macrophages (fig. S11C). The GEPIA database demonstrated that Gal-9 expression in GBM and low-grade glioma (LGG) was significantly higher than in normal tissue (fig. S11D). Notably, glioma patients with high Gal-9 or (and) Tim-3 expression showed shorter survival in the CGGA dataset ([110]Fig. 5A), TCGA dataset ([111]Fig. 5B), and [112]GSE16011 dataset ([113]Fig. 5C), respectively. Moreover, tyramide signal amplification immunohistochemistry (TSA-IHC) staining verified that Gal-9 and CD163 were expressed at higher levels in human PTEN-Del GBM tissues than in PTEN-WT GBM tissues ([114]Fig. 5D). We also examined 240 human glioma samples stained for Gal-9 and Tim-3 using TSA-IHC ([115]Fig. 5E), and staining intensity was quantified using the inForm System. Kaplan-Meier analysis revealed that high expression scores for Gal-9/Tim-3, individually or together, predicted shorter survival of GBM patients than those with low Gal-9/Tim-3 features (P < 0.0001; [116]Fig. 5F). Taken together, these results indicate that Gal-9/Tim-3 signaling strongly correlates with the mesenchymal GBM subtype and predicts a poor prognosis of glioma patients. Fig. 5. High activation of Gal-9/Tim-3 signaling predicts poor survival of human glioma patients. [117]Fig. 5. [118]Open in a new tab Kaplan-Meier survival curves of glioma patients stratified by Gal-9 expression, Tim-3 expression, or the product of Gal-9 and Tim-3 expression (Gal-9 × Tim-3) in the CGGA dataset (A), TCGA dataset (B), and [119]GSE16011 dataset (C). (D) Immunofluorescence staining and quantification of Gal-9 and CD163 expression in human GBM tissues with WT (n = 9) or deleted (n = 9) PTEN gene. Scale bar, 100 μm. **P < 0.01 and ***P < 0.001, Student’s t test. (E) Representative immunofluorescence staining for Gal-9 and Tim-3 in human glioma tissue microarrays. Scale bar, 200 μm. (F) Kaplan-Meier survival curves indicate that high Gal-9 and/or Tim-3 expression correlates with a poor prognosis for glioma patients. Differences in Kaplan-Meier curves were assessed using the log-rank test. Blockade of Gal-9/Tim-3 signaling inhibits macrophage M2 polarization, suppresses tumor growth, and prolongs survival in GBM models To validate the role of Gal-9/Tim-3 signaling in modulating macrophage M2 polarization, we used both pharmacologic (α-lactose/anti–Tim-3 antibody) and genetic (shRNAs) interference methods in PTEN-deficient glioma cells. Immunofluorescence showed that GCM supplemented with α-lactose markedly reduced Gal-9 protein binding to macrophage surface in vitro (fig. S12A). PTEN-deficient GCM supplemented with different concentrations of α-lactose was used to treat THP-1 macrophages for 72 hours. Further, flow cytometry results showed that α-lactose had a dose-dependent effect on reducing M2 polarization, as indicated by the reduced expression of CD163 and CD206 markers in THP-1 macrophages ([120]Fig. 6A and fig. S12B). The qRT-PCR results showed that GCM supplemented with α-lactose induced significant up-regulation of the M1 markers (TNF-α, IL-12, and IL-23) and down-regulation of M2 markers (IL-10 and TGF-β1) to some extent in THP-1 macrophages and BMDMs (fig. S12C). In contrast to GCM-treated macrophages, GCM mixed with anti–Tim-3 antibody or α-lactose increased the expression level of macrophage M1 marker CD86, as detected by flow cytometry ([121]Fig. 6B and fig. S12D), and the expression level of IL-1β, as detected by cytokine microarray analysis (fig. S12E). Notably, neither Gal-9 knockdown (fig. S12F) nor α-lactose/anti–Tim-3 antibody (fig. S12G) affected the proliferation of PTEN-deficient glioma cells in vitro. Fig. 6. Blockade of Gal-9/Tim-3 signaling inhibits macrophage M2 polarization and GBM growth in vivo. [122]Fig. 6. [123]Open in a new tab (A) Flow cytometry analysis of CD163 and CD206 on THP-1 macrophages treated with different doses of α-lactose–supplemented GCM. (B) Flow cytometry analysis of CD86 on BMDMs or THP-1 macrophages treated with GCM, GCM + anti–Tim-3 antibody (1 μg/ml), or GCM + α-lactose (40 μM). (C to E) Survival curves of BALB/c-nu/nu mice bearing orthotopic U87 xenografts (C), C57BL/6J mice bearing orthotopic GL261 KO xenografts (D), and Wistar rats bearing orthotopic C6 xenografts (E) treated with α-lactose or anti–Tim-3 antibody. (F) Gal-9 knockdown prolonged the survival of BALB/c-nu/nu mice bearing orthotopic U87 xenografts. (G) Representative immunofluorescence staining and quantification of Gal-9/CD163 in orthotopic PTEN-Del U87 and PTEN-KO GL261 tumors treated with α-lactose or anti–Tim-3 antibody. Scale bar, 200 μm. ns, no significance. ***P < 0.001, one-way ANOVA. (H) Representative immunofluorescence staining and quantification of Gal-9/CD163 in orthotopic and subcutaneous U87-shC and U87-shGal-9 tumors. Scale bar, 200 μm. Tumors were harvested on day 22 after implantation. n = 3 biological replicates. ***P < 0.001, Student’s t test. In (C) to (F), *P < 0.05, **P < 0.01, and ***P < 0.001, log-rank test. We next assessed the effect of Gal-9/Tim-3 blockade on tumor growth and macrophage polarization in vivo. We found that α-lactose or anti–Tim-3 antibody significantly delayed tumor growth (fig. S13, A and B) and extended the survival of both orthotopic PTEN-null U87 ([124]Fig. 6C) and PTEN-KO GL261 ([125]Fig. 6D) mouse glioma models, and an orthotopic PTEN-null C6 rat glioma model ([126]Fig. 6E). Unexpectedly, shRNA-mediated depletion of Gal-9 expression markedly slowed tumor growth in subcutaneous and orthotopic PTEN-null U87 GBM models (fig. S13, C to E) and extended the survival of mice bearing orthotopic U87 xenografts ([127]Fig. 6F). Accordingly, CD163^+ macrophage infiltration was markedly reduced in GBM xenografts by α-lactose or anti–Tim-3 antibody treatments ([128]Fig. 6G), or by shRNA-mediated Gal-9 knockdown ([129]Fig. 6H) in GBM mouse models, in accordance with the down-regulated Gal-9 expression induced by α-lactose or shGal-9. In orthotopic C6 xenografts, the expression of the M1 macrophage marker CD86 was accordingly up-regulated in the α-lactose and anti–Tim-3 antibody groups (fig. S13F). α-Lactose inhibits TAM-derived VEGFA expression levels and attenuates angiogenesis in glioma We noticed that inhibition of Gal-9 expression by α-lactose or shRNA effectively impeded glioma growth in vivo but not in vitro, prompting speculation that TAMs may secrete factors supporting glioma growth and angiogenesis. Our computational analysis ([130]Fig. 4, E and F) implied that the downstream target genes of Gal-9/Tim-3 signaling were significantly enriched in VEGF production and vasculature development. Therefore, we examined tumor angiogenesis to understand how α-lactose–regulated TAMs inhibit GBM progression. We found that blood vessels were reduced in response to α-lactose in subcutaneous ([131]Fig. 7A) and orthotopic ([132]Fig. 7B) PTEN-KO GL261 xenografts. In subcutaneous PTEN-KO GL261 tumors treated with or without α-lactose (fig. S14A), IHC analysis (fig. S14B) indicated that CD31 expression (vessel density) positively correlated with tumor size (fig. S14C), Gal-9 expression (fig. S14D), and CD163 expression (fig. S14E). GSEA and RNA-seq analysis revealed that VEGFA signaling was highly activated, and 50 angiogenesis-related genes were up-regulated in GCM-treated monocytes, compared with those in untreated monocytes ([133]Fig. 7, C and D). GSEA for two VEGFA signatures in TCGA samples suggested a highly activated VEGFA pathway in the GBM samples with macrophage M2–positive correlation ([134]Fig. 7E), and VEGFA was positively correlated with the M2 macrophage marker CD163 in the CGGA database ([135]Fig. 7F). Furthermore, qRT-PCR and immunoblots demonstrated that GCM supplemented with α-lactose significantly inhibited VEGFA expression in GCM-treated THP-1 macrophages and BMDMs ([136]Fig. 7, G and H). Thus, these data suggest that α-lactose can decrease VEGFA expression that was initially up-regulated by TAMs, thereby inhibiting glioma angiogenesis. A schematic model that summarizes results of our study is shown in [137]Fig. 7I: (i) PTEN deficiency in GBM cells promotes macrophage M2 polarization via the intercellular Gal-9/Tim-3 interaction, with Gal-9 secretion from glioma cells directly modulated by the AKT-GSK3β-IRF1 pathway; (ii) Gal-9/Tim-3 signaling can be blocked by α-lactose or anti–Tim-3 antibody; and (iii) Gal-9/Tim-3 inhibition impairs PTEN-deficient GBM progression by decreasing TAM-derived VEGFA, thus attenuating angiogenesis. Fig. 7. α-Lactose inhibits glioma angiogenesis by impairing TAM-derived VEGFA expression. [138]Fig. 7. [139]Open in a new tab (A) Representative images of subcutaneous PTEN-KO GL261 tumors treated with or without α-lactose. (B) IHC showing CD31^+ vessel density in intracranial PTEN-KO GL261 tumors treated with or without α-lactose. Scale bar, 200 μm. n = 3 biological replicates. (C and D) GSEA for VEGFA signatures and heatmap representation of 50 up-regulated VEGFA pathway–associated genes in healthy monocytes treated with or without GCM. (E) GSEA for two VEGFA signatures in M2 macrophage–positive versus M2 macrophage–negative correlated TCGA GBM samples. NES, normalized enrichment score. (F) VEGFA positively correlates with CD163 in the CGGA database. (G) qRT-PCR and (H) immunoblot validation of VEGFA expression in THP-1 macrophages and BMDMs treated with GCM or GCM + α-lactose. ***P < 0.001, Student’s t test. (I) Schematic model of the mechanism underlying how glioma cells with PTEN deficiency regulate macrophage polarization through Gal-9/Tim-3 signaling, which can be blocked by α-lactose or anti–Tim-3 antibody. DISCUSSION Surgical resection followed by adjuvant radiochemotherapy is the standard treatment for GBM and produces only modest benefits for survival ([140]20). Immunotherapy with checkpoint inhibitors has revolutionized the treatment of a variety of tumors, but only a small subset of GBM patients (<10%) shows objective responses to immunotherapy ([141]21). In this study, we explored the mechanisms underlying the polarization of TAMs, the most abundant nontumor cells in GBM tumors ([142]4), and assessed their roles in angiogenesis in PTEN-null GBM. Previous studies have shown that high stromal and immune signatures are enriched in mesenchymal GBM and are specifically correlated with genetic alterations of the PTEN-PI3K pathway ([143]6). These findings suggested that genetic driver mutations such as PTEN alteration can create unique TMEs with abundant macrophage infiltration ([144]2, [145]6). Here, we confirmed that PTEN-null glioma cells recruited more macrophages than PTEN-WT glioma cells, consistent with other studies ([146]6). First, we found that these macrophages tended to be polarized into M2 macrophages when cocultured with GCM, and PTEN-deficient glioma cells were more effective in promoting macrophage M2 polarization than PTEN-WT glioma cells. Subtype-specific alterations like PTEN deficiency can provide promising therapeutic targets in GBM. Our results indicate that macrophage-centered approaches may be effective for treating PTEN-null glioma but may not be effective for glioma with the WT PTEN gene. To mimic the effect of PTEN gene alterations on the TME in glioma, we knocked out PTEN in LN229, SKMG-1, and GL261 cells, and overexpressed WT PTEN in U87 and U118 cells. Erasing PTEN protein in PTEN-WT cells using CRISPR technology could simulate one kind of PTEN gene deficiency causing PTEN protein loss, similar to the natural loss of PTEN protein in PTEN-Del cells. We examined the functions of both PTEN-OE and PTEN-WT cell lines in modulating macrophage polarization. In response to PTEN overexpression, the mRNA levels of LGALS9 in LN229-KO cells were restored to levels observed in LN229-WT cells (fig. S15A). There was no significant difference in the modulation of macrophage M2 polarization among LN229-WT cells, PTEN-overexpressing LN229-KO cells (LN229 KO + OE), and PTEN-overexpressing LN229-WT cells (LN229 WT + OE) (fig. S15B). These results suggested that our cell models were suitable to explore how PTEN loss in glioma affected macrophage polarization. We then found that PTEN deficiency–mediated high expression of Gal-9 was involved in macrophage M2 polarization. Furthermore, combined profiling and functional experiments revealed that PTEN deficiency in glioma cells promoted Gal-9 expression and secretion. Gal-9, a β-galactoside–binding protein implicated in modulating cell-cell and cell-matrix interactions, is inhibited by α-lactose ([147]22). Another report showed that Gal-9 promoted M2 polarization by activating Tim-3 receptors on BMDMs and that α-lactose blocked Gal-9/Tim-3 signaling ([148]22). Therefore, in this study, α-lactose and anti–Tim-3 antibody were used to block Gal-9/Tim-3 signaling in vitro and in vivo, and both treatments markedly reduced macrophage M2 polarization in PTEN-deficient GBM models. We found that Gal-9 inhibition or anti–Tim-3 antibody treatment impaired GBM growth by reducing macrophage M2 polarization in vivo, while no direct impact on glioma cell proliferation was observed in vitro. We also established that PTEN-null–dependent Gal-9–driven up-regulation of macrophage M2 polarization supports GBM growth in animal models. To explore the link between PTEN and Gal-9, we used several inhibitors to block multiple downstream signaling pathways activated by PTEN deficiency in glioma cells, including the AKT, SRC, YAP1, and NOTCH1 pathways ([149]6, [150]23). We demonstrated that the AKT pathway played a key role in Gal-9 up-regulation. We then evaluated the molecules downstream of the AKT pathway that up-regulated Gal-9 expression and found that GSK3β, but not mTOR, participated in the up-regulation of Gal-9 in PTEN-KO glioma cells. Using biological databases coupled with an in vitro qRT-PCR–based validation method, we identified IRF1 that regulates LGALS-9 gene expression, likely through binding a specific promoter region. IRF1, a mediator of GBM resistance to bevacizumab ([151]24), was shown to regulate PD-L1 expression in cancer cells ([152]25). We found that IRF1 strongly correlates with the mesenchymal subtype GBM, similar to Gal-9/Tim-3. In addition to these findings, AKT-GSK3β-IRF–1driven mechanisms of Gal-9 up-regulation support the idea that genetic PTEN deficiency in glioma promotes macrophage M2 polarization by regulating specific gene expression profiles. Although many previous studies ([153]6) used immunocompromised mouse models [BALB/c-nu/nu and nonobese diabetic–severe combined immunodeficient (Nod-SCID) mice] to examine the function of macrophages, such models are not yet realistic enough to mimic the macrophage-glioma interactions in humans in vivo. Therefore, we additionally used immunocompetent C57BL/6J mice and Wistar rats to establish orthotopic glioma (GL261-KO and C6) models, and treated the models with Gal-9/Tim-3 blockers. The results in both kinds of animal models verified that Gal-9/Tim-3 blockers were effective in impairing PTEN-null glioma progression. Growing evidence has shown that TAMs can promote glioma cell survival and angiogenesis ([154]8, [155]12). VEGFA secreted from macrophages plays a critical role in triggering angiogenesis ([156]26), and it is significantly up-regulated in macrophages isolated from GBM tissues ([157]27). Recently, a study showed that genetic ablation of macrophages delayed tumor angiogenesis and that VEGFA alone was sufficient to promote angiogenesis and compensate for macrophage deficiency ([158]28). Another study highlighted that knockout of VEGFA expression in myeloid cells attenuated glioma progression in experimental glioma models ([159]12). Here, we demonstrated that M2-polarized macrophages promoted glioma angiogenesis through VEGFA secretion. Unfortunately, antiangiogenic therapy of GBM with bevacizumab may accelerate glioma cell invasion and activate alternative angiogenic pathways ([160]29). In our study, analysis of scRNA-seq data of GBM tissues reveals that Gal-9 ligands activate Tim-3 receptors on macrophages followed by activation of M2 polarization–related TFs and up-regulation of M2 markers and angiogenesis-related genes in macrophages. Our results suggest that Gal-9/Tim-3 signaling–mediated glioma-macrophage interactions may represent as a novel target for glioma antivascularization. Here, the identification of VEGFA as an abundant TAM-secreted factor, along with α-lactose capable of decreasing VEGFA secretion, encourages the potential application of α-lactose in antivascularization treatment for GBM patients. Thus, our work reinforced the importance of TAMs in GBM angiogenesis and identified α-lactose as a potential and specific intervention targeting glioma angiogenesis. Given the known immunosuppressive and angiogenic role of TAMs, it is urgent to investigate whether GBM patients can benefit from treatments that decrease the number of M2 macrophages. Although CSF1R inhibition with BLZ945 alters macrophage polarization and blocks glioma progression ([161]15), the antitumor responses are transient and tumors recur in >50% of GBM mouse models ([162]16). A recent study reported that PI3K pathway activity was elevated in recurrent GBM, and combining PI3K blockade with CSF1R inhibition in these recurrent tumors significantly prolonged overall survival ([163]16). A clinical trial showed that the CSF1R inhibitor PLX3397 was not effective in patients with recurrent GBM ([164]17). Remarkably, PLX3397 extended the progression-free survival of two GBM patients (2/37), who presented with the mesenchymal subtype GBM with common gene alterations, such as PTEN deficiency ([165]17). Additional studies are ongoing, testing potential biomarkers to identify glioma patients with greater likelihood of response after macrophage-targeted therapy. Correspondingly, we found that Gal-9/Tim-3 signaling strongly correlated with mesenchymal subtype GBM, and predicted poorer clinical outcomes for glioma patients. Furthermore, we found that PTEN deficiency leads to up-regulated Gal-9 secretion by GBM cells via activation of the PI3K-AKT pathway, suggesting a novel mechanism of GBM resistance to macrophage-targeted therapy, such as CSF1R inhibitor treatment. We highlighted that α-lactose, a Gal-9 inhibitor, can impair glioma progression by reducing macrophage M2 polarization. The finding that Gal-9/Tim-3 signaling blockade increased survival in PTEN-deficient GBM models supports the importance of clinical testing of α-lactose specifically in PTEN-deficient GBM patients. Our in vivo data showed that blocking Gal-9/Tim-3 could significantly inhibit macrophage M2 polarization and angiogenesis, resulting in retardation of glioma growth. These results have significant therapeutic implications that targeting Gal-9/Tim-3 may be exploited as a precision immunotherapy specifically for PTEN-null gliomas. Furthermore, combining Gal-9/Tim-3 inhibitors with other existing targeted therapies (e.g., anti-VEGF antibody and anti–PD-1/PD-L1) is anticipated to produce a synergistic therapeutic effect for glioma. In future studies, it is necessary to conduct the clinical trials to confirm the efficacy of Gal-9/Tim-3 inhibitors in glioma. In conclusion, compared with PTEN-normal glioma cells, PTEN-deficient glioma cells recruited more macrophages and induced their polarization toward the M2 phenotype by abundantly secreting Gal-9 and activating Tim-3 on macrophages. Gal-9/Tim-3 blockade can thus impair glioma-macrophage interaction and attenuate angiogenesis to inhibit tumor growth. MATERIALS AND METHODS Human glioma samples A tissue array (240 glioma samples) containing 12, 85, 76, and 67 cases of grade I, II, III, and IV glioma was obtained from patients who received surgery at Sun Yat-sen University Cancer Center (SYSUCC) between 2001 and 2016 with written informed consents. All samples were pathologically diagnosed by experienced pathologists following the World Health Organization (WHO) 2016 Classification of Gliomas. The three human GBM tissues were collected for 10× Genomics scRNA-seq, and the PTEN gene deletion status was confirmed by fluorescence in situ hybridization performed in the Department of Molecular Diagnosis of SYSUCC (fig. S7). We also used Sanger sequencing to ascertain the PTEN gene mutation status of other human GBM tissues collected for primary culture. This study was approved by the Ethics Committee of SYSUCC (no. GZR2013-052). Reagents and antibodies Information on all reagents and antibodies is shown in table S3. Mice and xenograft tumor models All animal experiments were performed with the approval of the institutional ethics committee of SYSUCC (no. L102042020120S). Four-week-old Nod-SCID female mice were purchased from SPF Biotechnology Co. Ltd. (Beijing, China). Female BALB/c-nu/nu mice and C57BL/6 mice were purchased from Guangdong Medical Laboratory Animal Center (Guangzhou, China). Female Wistar rats were purchased from Zhejiang Vital River Laboratory Animal Technology Co. Ltd. (Zhejiang, China). The mice were inoculated with 1 × 10^6 cells (100 μl) in the right flank to establish subcutaneous xenografts, and the tumor weight was determined immediately after tumor removal at the end of the observation period. Intracranial tumor models were established as previously described ([166]30). A total of 1 × 10^5 cells were injected intracranially in 5 μl of 1× phosphate-buffered saline (PBS) into the mice, and tumor growth was monitored by bioluminescence imaging. After 1 week of recovery, the glioma animal models were assigned to treatment groups. Mice received tail intravenous injection with 100 μg of purified anti-mouse Tim-3 antibody once a day for seven consecutive days. In addition, α-lactose (10 mg) was intraperitoneally injected into each mouse once a day for 14 consecutive days. Mice with moribund appearance were sacrificed for collecting brain samples. The samples were processed for embedding in paraffin blocks. Cell culture The culture conditions and PTEN gene mutation status of all GBM cell lines used in this study are listed in table S1. Consistent with previous studies ([167]31, [168]32), our immunoblot results showed that C6 rat glioma cells did not express PTEN protein (fig. S16). THP-1 macrophages, BMDMs, and human monocytes were cultured in RPMI 1640 supplemented with 10% FBS. BMDMs from C57BL/6 mice were cultured as previously described ([169]33). Before transwell assays, differentiation was induced in THP-1 cells by phorbol 12-myristate 13-acetate (PMA) (100 nM) for 72 hours, and differentiation was induced in BMDMs by CSF1 (10 ng/ml) for 6 days. Human monocytes were treated with CSF1 (10 ng/ml) for 3 days before use in in vitro experiments. After culturing glioma cells in FBS-free medium for 24 hours, supernatants were collected as the glioma cell–derived CM and then filtered with a polyethersulfone (PES) 0.22-μm membrane for use for in vitro experiments. DNA extraction and DNA sequencing Genomic DNA was extracted from fresh human GBM tissue samples or cell lines using the TIANamp Genomic DNA Kit in accordance with the manufacturer’s instructions. DNA samples were sent to Guangzhou Tianyi Huiyuan Company for amplification and Sanger sequencing of the PTEN gene. PCR amplification of exons 1 to 9 of the PTEN gene was performed in a reaction volume of 30 μl containing specific forward and reverse primers (table S3). The PCR cycling conditions were as follows: initial denaturation at 94°C for 2 min, followed by 35 cycles of denaturation at 94°C for 10 s, annealing at 60°C for 30 s, and extension at 72°C for 30 s, with a final extension at 72°C for 5 min. The PCR sequencing primers are listed in table S3. Regions of introns before and after the exons were also sequenced. Migration assay Cells (1 × 10^5 THP-1 cells and 1 × 10^4 BMDMs) were suspended in serum-free medium and seeded into 24-well inserts (8.0 μm). GCM was added to the receiver wells. Cells were cultured for 24 hours, and the migrated macrophages were fixed with 10% formaldehyde, stained with crystal violet, and counted under a microscope. Immunoblotting Cell lysates were subjected to SDS–polyacrylamide gel electrophoresis and transferred to polyvinylidene difluoride membranes. Blots were incubated with primary antibodies at 4°C overnight. After three washes with 1× Tris-buffered saline containing Tween 20 (TBST), the membrane was incubated with secondary antibody for 1 hour at room temperature. Enhanced chemiluminescence was used to detect protein signals. Band densities were normalized against the loading control β-actin, β-tubulin, lamin B1, or glyceraldehyde-3-phosphate dehydrogenase (GAPDH) using ImageJ software. Flow cytometry Macrophages were pretreated with anti-mouse or human Tim-3 antibody, or α-lactose at the indicated concentrations, diluted with GCM for 72 hours. Cells were collected and resuspended in 100 μl of PBS (1 × 10^7 cells/ml). Fluorescence-conjugated antibodies were added, and the samples were incubated for 30 min at room temperature. After washing with 1× PBS, cells were analyzed by flow cytometry to detect CD68, CD86, CD163, and CD206 expression levels. Enzyme-linked immunosorbent assay ELISA kits (MM-51152HI) were used to detect Gal-9 levels in CM collected from 23 glioma cell lines, including 13 PTEN-WT cell lines (UWR7, MGR2, SHG44, GL261, T98, SKMG-1, LN229, SKMG-4, SYU596, SYU488, SYU214, LN308, and SYU687), 8 PTEN-null cell lines (U118, SYU354, SF295, U373, MGR3, U87, U138, and U251), and 2 PTEN-mutated cell lines (U343 and A172). Glioma cells (1 × 10^7) were cultured in FBS-free medium for 24 hours; cell-free supernatants were collected for protein quantification with a bicinchoninic acid (BCA) protein assay kit, followed by evaluation using the ELISA kit. Cytokine antibody microarrays The Quantibody Human Cytokine Antibody Array 440 was used to perform comprehensive proteomic analysis of the LN229-KO CM and LN229-WT CM. The cytokine profiles of PTEN-deficient and PTEN-normal GBM cells (PTEN-Del or PTEN-OE U87 cells; PTEN-WT or PTEN-KO SKMG-1 cells) were detected using RayBio C-Series Human T Cell Response Array 1. The cytokine profiles of BMDMs treated with GCM, GCM + α-lactose, or GCM + anti-mouse Tim-3 antibody were detected using Quantibody Mouse TH17 Array 1. The detailed experiment protocols are available at [170]www.RayBiotech.com. Single-cell RNA sequencing Sample preparation GBM tissues from three patients were collected and mechanistically minced into approximately 1- to 3-mm^3 pieces. Samples were enzymatically digested in Dulbecco’s modified Eagle medium containing 10% FBS, collagenase II (0.8 mg/ml; Sigma-Aldrich, catalog no. C6885), collagenase IV (1.5 mg/ml; Sigma-Aldrich, catalog no. C5138), 3 mM CaCl[2], deoxyribonuclease I (1 mg/ml; Sigma-Aldrich, catalog no. DN25), and 20% dispase buffer (Corning, catalog no. 354235) on a rotor at 37°C for approximately 30 min. The cell suspension was filtered through 70- and 40-μm cell strainer and then centrifuged for 4 min at 400g. Cell pellets were resuspended in red blood cell lysis buffer (Boster, AR1118) and incubated on ice for 5 min. After centrifugation at 300g for 5 min, cell pellets were washed and resuspended in 1× PBST with 0.04% bovine serum albumin. scRNA-seq library preparation and sequencing The single-cell suspension (15 μl, ~900,000 cells/ml) was loaded into one channel of the Chromium Single-Cell G Chip (10× Genomics, 1000120), aiming to capture 8000 to 9000 cells. The Chromium Single-Cell 3′ Library & Gel Bead Kit v3.1 (10× Genomics, 1000121) was used for single-cell bar coding, complementary DNA (cDNA) synthesis, and library preparation, following the Single-Cell 3′ Reagent Kits User Guide (version 3.1). Libraries were then subjected to 150–base pair (bp) paired-end sequencing on an Illumina NovaSeq 6000 platform. scRNA-seq data analysis We used Seurat v4.0.3 ([171]34) with default parameters to perform quality control, sctransform (SCT) normalization, integration, Uniform Manifold Approximation and Projection (UMAP) dimensionality reduction, and visualization for the scRNA-seq data. The “FindClusters” function was used for cell clustering (resolution parameter set to 0.1). The cell types were annotated on the basis of the expression of known marker genes ([172]35, [173]36). Specifically, the following marker genes (fig. S8) were used for cell type annotation: CD14, CD68, CSF1R, and FCGR3A for macrophages; SOX2, OLIG2, and ASCL1 for malignant cells; CD14, CD68, S100A8, and S100A9 for neutrophils; CD3D and CD3E for T cells; FBLN1 and DCN for fibroblasts; MAG and MOG for oligodendrocytes; and CLDN5 and VWF for endothelial cells. The annotation of malignant cells was verified by comparing the copy number patterns with nonmalignant cells ([174]37) using InferCNV v.1.6.0 with default parameters. Multilayer network approach for cell-cell communication inference We developed an scRNA-seq data-driven multilayer network analysis approach to infer intercellular communications and intracellular gene regulations on the basis of our previous method scMLnet ([175]38, [176]39). The cell type–specific up-regulated genes were selected using the “FindAllMarkers” function in Seurat with default parameters. We collected prior information of ligand-receptor (LRDB) and TF-target gene pairs (TFTGDB) from Omnipath database ([177]40), a state-of-the-art knowledge base of intra- and intercellular signaling information. To construct receptor-TF pairs, we first extracted the protein-protein interaction from Omnipath as an undirected weighted graph G. The receptor-TF pairs were sorted according to the Dijkstra’s distance between receptors and TFs in the graph G, and the top 20% were used as confident receptor-TF pairs (RTFDB). The above LRDB, TFTGDB, and RTFDB were used as prior information for the following multilayer network construction. We leveraged scRNA-seq expression data to construct a cell type–specific multilayer network. For a pair of sender (cell type A) and receiver (cell type B), we ranked LR pairs on the basis of the product of mean expression of the corresponding ligand and receptor, and the top 20% of LR pairs were assumed as potential activated LR pairs. Fisher’s exact test was used to verify the activation of the TFs on the basis of the expression of the target genes (cell type B–specific up-regulated genes), which were then used to verify the activation of receptors. The filtering criteria were set as P value of <0.05. At last, the activated LR pairs, TFs, and the corresponding target genes were coupled together into a multilayer network. We performed the cell-cell communication analysis for each pair of cell types, and the number of the downstream target genes of activated LR pairs was used as the communication strength. To investigate functions of the cell-cell communication, we performed GO term (biological process) enrichment analysis for the Gal-9/Tim-3 downstream target genes using clusterProfiler v3.18.1 ([178]41). In addition, we performed GSEA enrichment analysis to identify biological processes enriched by the ranked gene lists in macrophages on the basis of fold changes of gene expression in PTEN-null sample compared with PTEN-WT samples. Gene sets related to biological process classification in the GO database were downloaded from the MSigDB database for enrichment analysis. Bulk RNA-seq data analysis For analysis of human GBM data, the gene expression and clinical and survival data of patients were downloaded from the CGGA database ([179]www.cgga.org.cn/). We analyzed the correlation between CD163 and VEGFA expression, between CD68 and LGALS9 (encoding Gal-9) or HAVCR2 (encoding Tim-3) expression, and between IRF1 and LGALS9 expression as recorded in the mRNAseq_693 dataset. The expression of genes of interest (IRF1, LGALS9, and HAVCR2) in different GBM subtypes (mesenchymal, classical, neural, and proneural subtypes) was analyzed using the CGGA database. Using the GEPIA database ([180]http://gepia.cancer-pku.cn/), we confirmed the correlation between LGALS9 and IRF1 expression. We also analyzed LGALS9 expression in tumor and normal tissues in GBM and LGG patients. The gene expression profile and immune infiltration levels of GBM samples from The Cancer Genome Atlas (TCGA) database ([181]https://cancergenome.nih.gov/) was downloaded from UCSC Xena ([182]https://xenabrowser.net/datapages/) and TIMER2.0 ([183]timer.cistrome.org). The gene expression data are presented as in log[2](x + 1) transformed RNA-Seq by Expectation-Maximization (RSEM) normalized count. GSEA of genes ranked by correlation with M2 macrophage infiltration levels was conducted on the basis of VEGFA targets and VEGFA signaling gene sets ([184]www.gsea-msigdb.org/gsea/msigdb/genesets.jsp?collection=C6). Survival analysis To assess the prognostic significance of Gal-9 and Tim-3 expression in glioma patients, we collected both gene expression data and the clinical information for glioma patients from three independent databases, including the CGGA database ([185]www.cgga.org.cn/), TCGA database ([186]https://cancergenome.nih.gov/), and Gene Expression Omnibus (GEO) database ([187]www.ncbi.nlm.nih.gov/geo/). The CGGA dataset, TCGA dataset, and [188]GSE16011 dataset included 400, 663, and 264 samples, respectively. The patients in each dataset were stratified into two groups on the basis of Gal-9 expression, Tim-3 expression, or their product (Gal-9 × Tim-3), with an optimal cutoff value determined by using the receiver operating characteristic (ROC) method [i.e., on the basis of the highest value of (sensitivity + specificity)] ([189]42). Kaplan-Meier curves of overall survival for patients in the two groups were analyzed, and the statistical significance of the difference was assessed using the two-sided log-rank test. Multiplex staining, multispectral imaging, and immunofluorescence To identify macrophage polarization in the TME of PTEN-deficient glioma, multiplex immunofluorescence staining of glioma tissue slices was performed using TSA Plus Fluorescence Kits combined with IHC (TSA-IHC) in accordance with the company protocols. The slices were sequentially incubated with primary antibodies (one by one) and horseradish peroxidase–conjugated secondary antibody (a mixture of anti-rabbit and anti-mouse). Slices were then incubated with TSA liquid reagent, which was respectively mixed with dyes (1:200), including PPD520 dye for staining Gal-9, PPD570 dye for staining Tim-3, PPD650 dye for staining CD163, PPD690 dye for staining CD86, and 4′,6-diamidino-2-phenylindole (DAPI) dye for staining nuclei. Before the final staining for nuclei, the slides were microwave heat–treated after each TSA operation. As for other operation details, the instructions of TSA-IHC kit could be followed. Slides were scanned at ×20 magnification for capturing multiplex IHC images or 3,3’-diamino-benzidine (DAB)–stained IHC images using the Vectra Automated Quantitative Pathology Imaging System (Vectra 2.0.8; PerkinElmer). Five regions of interest (20×) were captured in each sample slice. First, the images were manually scored by a pathologist (S.X.), who served as a blinded reviewer. Tumor multispectral images were unmixed using a spectral library built from images of single-stained control tissues for each dye (PANOVUE) using the inForm software (inform 2.1.1; PerkinElmer). A selection of 10 representative multispectral images was used to train the inForm software. Then, all settings applied to the training images were saved as a project. All images were subsequently evaluated by the inForm analysis to confirm the manual scoring and calculate the accurate percentages of label-positive cells. In a sample slice, the positive percentages of five high-powered images (20×) were averaged and determined as the score (%) of the specific markers. A Gal-9–positive percentage of >25% was defined as Gal-9^High, and Tim-3-positive percentage of >5% was defined as Tim-3^High. (Gal-9 & Tim-3)^High was defined if the samples showed both Gal-9–positive percentage of >25% and Tim-3–positive percentage of >5%. Immunofluorescence in cells was performed using standard protocols. To determine the localization of Tim-3 and Gal-9 expression in macrophages, images were captured using a laser confocal microscope (Olympus). qRT-PCR and RNA-seq For qRT-PCR, total RNA was extracted using the RNA Quick Purification Kit (ESscience, Shanghai, China) and reverse-transcribed to cDNA in a total volume of 20 μl, in accordance with the manufacturer’s instructions (catalog no. 11141ES60; Yeasen, Shanghai, China). All primer sequences for qRT-PCR are shown in table S3. qPCR (10 μl per well) was performed at 50°C (2 min) for Uracil-DNA Glycocasylase (UDG) activation and 95°C (2 min) for Dual-Lock DNA polymerase, followed by 40 cycles at 95°C (15 s) for denaturing, 60°C (15 s) for annealing, and 72°C (1 min) for extension using the Bio-Rad CFX96 Real-Time PCR System (Bio-Rad Laboratories Inc., Hercules, CA, USA). PowerUp SYBR Green Master Mix (01000439, Thermo Fisher Scientific) was used for signal detection. The number of cycles required to generate a given threshold signal (Ct) was recorded, and fold changes were calculated using the cycle threshold ΔΔCt method. Each reaction was performed in triplicate. The expression of each gene was normalized to that of mouse or human GAPDH expression. For RNA-seq, RNA was isolated from cells using TRIzol reagent (BCCC8200; Sigma-Aldrich) in accordance with the manufacturer’s instructions. RNA-seq analysis was performed with RNA prepared from PTEN-WT and PTEN-KO LN229 cells (biological triplicates for the control and PTEN-KO LN229 cells) at LC-Bio (a subsidiary of LC Sciences). Differentially expressed genes between the control and PTEN-KO LN229 cells were subjected to GSEA. The up-regulated genes encoding secreted proteins in PTEN-KO LN229 cells were analyzed on the basis of the raw transcriptome data, and the results were validated by qRT-PCR. PTEN knockout using CRISPR and PTEN overexpression A pool of three single-guide RNA (sgRNA) plasmids targeting human PTEN (sc-400103) was purchased from Santa Cruz Biotechnology, and a pool of three sgRNA plasmids targeting mouse PTEN (MCP227985-CG04-3-10) was purchased from GeneCopoeia Inc. (Rockville, MD, USA). The plasmids were transiently transfected into cells (LN229, SKMG-1, and GL261 cells) using Lipofectamine 3000. After 72 hours, the cells were harvested and green fluorescent protein (GFP)–positive cells sorted by fluorescence-activated cell sorting (FACS) were inoculated into wells of 96-well plates within one GFP-positive cell per well. PTEN-deficient single clones were confirmed by immunoblot, and a clone with successful PTEN knockout (PTEN-KO) was chosen for subsequent experiments. We overexpressed WT PTEN in glioma cells using lentivirus purchased from Obio Technology Corp. Ltd. (Shanghai, China). shRNA-mediated knockdown Three shRNA hairpins targeting the human LGALS9 gene and a negative control shRNA were purchased from GeneCopoeia (Guangzhou, China). Recombinant lentiviral particles were produced using 293T cells. Briefly, 5 μg of the shRNA plasmid and 2 μg of VSVG, PLP1, and PLP2 lentivirus packing plasmids were transfected into 293T cells plated in 10-cm dishes using Lipofectamine 3000. Viral supernatant was collected at 48 and 72 hours after transfection and filtered for subsequent experiments. Cells were infected with viral supernatant containing polybrene for 48 hours and selected by incubation with puromycin (2 μg/ml) for 2 weeks. The expression of Gal-9 in glioma cells was determined by immunoblots and qRT-PCR. siRNA interference and dual-luciferase reporter gene assay siRNAs against human IRF1 (si-h-IRF1_02/06/07) and scramble siRNAs were purchased from Ribobio (Guangzhou, China). siRNAs were transfected into cells using Lipofectamine 3000 in accordance with the manufacturer’s instructions. The luciferase plasmids (pEZX-FR01-LGALS9-0.5/1.5/2.0 kb) and the control plasmids were purchased from GeneCopoeia. Three luciferase plasmids contain the 500/1500/2000-bp fragments of the LGALS9 promoter in the pEZX-FR01 vector containing the Firefly and Renilla luciferase genes. PTEN-KO LN229 and PTEN-KO SKMG-1 cells were transfected with pEZX-FR01-LGALS9-0.5/1.5/2.0 kb. PTEN-deficient U87 cells and PTEN-rescued U87 cells were transfected with pEZX-FR01-LGALS9-0.5 kb. In other experiments, pEZX-FR01-LGALS9-0.5 kb was cotransfected with siIRF1 or control siRNAs into PTEN-KO LN229 cells. At 48 hours after transfection, the cells were washed twice with cold 1× PBS, lysed in the lysis buffer provided in the dual-luciferase reporter gene assay kit (Yeasen, 11402ES60), and assayed for luciferase activity using a Tecan luminometer (Tecan Spark TM10M) in accordance with the manufacturer’s protocol. All transfections were performed in triplicate. Data are presented as the ratio of Firefly to Renilla luciferase activity. Statistical analysis All statistical analyses were performed with Student’s t test or one-way ANOVA. Analysis of the survival data was performed using the log-rank test (GraphPad Prism 7). Data are presented as means ± SD, and P < 0.05 was considered significant. Acknowledgments