Cell cultures
Murine glioma GL261 cells were obtained from Prof. Helmut Kettenman (MDC, Berlin, Germany) and modified to GL261 tdTomato+luc+ glioma cells as described previously [11]. The murine and human glioma cell lines: U251-MG, U87-MG (ATCC, Manassas, VA) and U87-MG RFP+ (AntiCancer Inc., San Diego, CA, USA) were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS) (Gibco, MD, USA). After thawing, the GL261 tdTomato+luc+ and U87-MG RFP+ cells were supplemented with 400 μg/ml G418 (Roche, Manheim, Germany) for two passages. Human SV40 immortalized microglia (HMSV40) were cultured in PriCoat T25 flasks in Prigrow III medium (Applied Biological Materials, Richmond, Canada) supplemented with 10% FBS (Gibco, MD, USA). Murine immortalized microglial BV2 cells (received from Prof. Klaus Reymann from the Leibniz Institute for Neurobiology) were cultured in DMEM GlutaMAX™ supplemented with 2% FBS (Gibco, MD, USA). All the cells were cultured with antibiotics (100 U/ml penicillin and 100 µg/ml streptomycin) in a humidified atmosphere of CO2/air (5%/95%) at 37°C (Heraeus, Hanau, Germany). Mycoplasma contamination-free status was checked regularly.
Cell viability and proliferation assays
Cell viability was evaluated using the MTT metabolism test as previously described [Ciechomska et al. 2023]. Cell proliferation was measured using a BrdU ELISA kit (Roche, Mannheim, Germany) according to the manufacturer’s protocol. Briefly, 5 × 103 cells were seeded onto 96-well plates. The next day, the media was changed to DMEM GlutaMAX™ supplemented with 2% FBS (Gibco, MD, USA), and the 7aaRGD and 7aaRAE peptides were added at a final concentration of 100 μM for 18 h. Peptides with N-terminal acetylation and C-terminal amidation were purchased from GenScript (Rijswijk, Netherlands). The peptides were dissolved in DMSO, and 0.2% solvent was added as a control. MTT solution (0.5 mg/mL; Sigma‒Aldrich, Taufkirchen, Germany) or BrdU was added for 1.5 and 2 h, respectively. Optical densities were measured at 570 nm for the MTT assay and at 450 nm for the BrdU assay using a scanning multiwell spectrophotometer.
Invasion assays
The invasion assay was performed as previously described (26). BV2 and HMSV40 cells (4×104) were plated onto a 24-well plate and co-cultured with GL261 cells (1×105/insert), U87 and U251 cells (4×104/insert) on Matrigel-covered inserts in 2% FBS-containing media. The cells were treated with 100 μM 7aaRGD and 7aaRAE (control) peptides or 0.2% DMSO. GL261 cells were co-cultured with BV2 cells for 24 h, and human glioma cells were cultured with BV2 or HMSV40 cells for 18 h. Invading cells fixed on the membranes were stained with DAPI.
Peptide stability
The 7aaRGD peptide was dissolved in water at a concentration of 2 mg/ml and incubated in osmotic pumps for 1, 7 and 14 days at 37°C. The peptide concentration was measured using Jupiter Proteo C12 2.1 × 250 mm, 4 μm column (Phenomenex, Torrance, USA) and HPLC Prominence coupled LCMS-IT-TOF (Shimadzu, Duisburg, Germany) under the following conditions: phase A: 0.1% HCOOH in Milli-Q water; phase B: 0.1% HCOOH in ACN; and gradient conditions: 0 min 2% B, 20 min 50% B, 25 min 95% B, 30 min 95% B, 35 min 5% B, and 55 min 5% B at a 0.2 ml/min flow rate. One milliliter of sample was injected. A Shimadzu IT-TOF ESI-MS system was used for mass analysis in automatic mode with a scan range of 150–2000 Da and an ion accumulation time of 10 ms. Electrospray ionization was performed in positive ionization mode with a spray capillary voltage of 5.0 kV. The interface temperature was maintained at 250°C, and the heat block temperature was 520°C. Nebulizing gas was introduced at 1.5 L/min, and the drying gas pressure was set to 100 kPa. Mass spectra were recorded in positive ion mode using the LCMSsolution software provided by Shimadzu. The analysis was performed at the Department of Chemistry of Warsaw University, Poland.
Stereotactic implantation of glioma cells and treatments
Male C57BL/6 mice or Athymic Nude-Foxn1nu mice (Charles River Laboratories, USA) (10–12 weeks old) were housed with free access to food and water on a 12 h/12 h day/night cycle. All efforts were made to minimize the number of animals and animal suffering. The mice were anesthetized with isoflurane (4–5% induction, 1–2% maintenance) via an isoflurane vaporizer (Temsega, Tabletop Anesthesia Station). Before starting the surgical procedure and during the surgery, the depth of anesthesia was verified. The choice of anesthetics was recommended by the veterinarian and approved by the local Ethics Committee. GL261 tdTomato+luc+ glioma cells (80,000 in 1 μL of DMEM) in C57BL/6J or U87-MG RFP (50,000 in 1 μL of DMEM) in Athymic Nude-Fox1nu mice were stereotactically injected into the right striatum at the following coordinates: 1 mm anterior and 2 mm lateral from bregma, 3 mm deep from the surface of the brain. At the same time, Alzet osmotic micropumps (DURECT Corporation, Cupertino, CA, USA) were installed in a subcutaneous pocket on the back, slightly posterior to the scapulae. A small incision was made in the shaved skin, and a hemostat was used to create a subcutaneous pocket for the pump, which was then inserted into the pocket, and the wound was closed with tissue glue. Prior to implantation, the metal flow moderators were replaced with PEEK flow moderators to be MRI compatible. To ensure that the pumps were active when implanted, the filled pumps were placed in sterile saline at 37°C for 24 h before implantation. Osmotic pumps were filled with H2O (vehicle), 7aaRGD or 7aaRAE peptides at a concentration of 2 mg/ml in H2O, and by means of a catheter, they continuously delivered the solutions intratumorally at a controlled rate of 0.11 μL/h for 21 or 28 days.
The mice were monitored until they completely recovered from anesthesia. The animals were weighed weekly and observed daily for clinical symptoms and evidence of toxicity by evaluating their eating, mobility, weight loss, hair loss, and hunched posture. Anti-PD-1 antibody (BioLegend, GoInVivo™ Purified anti-mouse CD279) was injected intraperitoneally (i.p.) at a dose of 10 mg/kg on days 8, 10, 12, and 14 post-implantation. The control groups received IgG antibody by i.p. injection. Animals were euthanized when they lost more than 20% of their body weight at day 0.
Tumor size measurement using magnetic resonance imaging
The heads of the animals were scanned with a 7T BioSpec 70/30 MR system (Bruker, Ettlingen, Germany) equipped with Avance III console and an actively shielded gradient system B-GA 20S (amplitude 200 mT/m) with an integrated shim set up to the 2nd order. A combination of a transmit cylindrical radiofrequency volume coil (8.6 cm inner diameter, Bruker) and a head-mounted mouse dedicated receive-only array coil (2x2 elements, Bruker) was used. The animals were anesthetized with 1.5–2% isoflurane (Baxter, Deerfield, IL, USA) in oxygen and positioned prone with the head placed in the MR-compatible bed integrated with an anesthesia mask. Respiration and rectal temperature were monitored throughout the experiment with an MR-compatible small animal monitoring system (SA Instruments, Stony Brook, NY, USA). All the imaging sessions started with a localizer protocol consisting of three orthogonal scout scans to accurately position the animal inside the magnet center. To evaluate the volumes of the brain structures, structural transverse MR images covering the whole brain were acquired with T2-weighted TurboRARE (TR/TEeff = 7000/30 ms, RARE factor = 4, spatial resolution = 86 μm × 86 μm × 350 μm, 42 slices, no gaps, number of averages (NA) = 4, scan time ~ 23 min). MRI scans were evaluated using OsiriX software (Pixmeo, Geneve, Switzerland), and manually delineated tumor regions in the image series were used for volumetric assessments.
Cytokine analysis
Pro- and anti-inflammatory cytokines were measured in serum and brain homogenates from control and treated animals. Blood was collected before perfusion and allowed to clot for 30 min before centrifugation (10,000 × g, 10 min at room temperature). The serum was collected and stored at -80°C. Brain homogenates were prepared by adding an equal volume of Cell Lysis Buffer 2 (R&D Systems, Minneapolis, MN, USA) to dissociated brain tissues. The samples were then incubated at room temperature for 30 min with gentle agitation, and debris were removed via centrifugation. The levels of cytokines were measured using the Luminex Assay Mouse Premixed Multi-Analyte Kit (R&D Systems, Minneapolis, MN, USA) according to the manufacturer’s protocol. Cytokine levels were determined using the MAGPIX Multiplexing Instrument (Luminex, TX, USA) with XPonent software. The results for each cytokine are expressed as pg/ml for serum samples and pg/mg protein for brain homogenates. The protein concentration in the brain homogenates was estimated using Bradford reagent (Sigma Aldrich), and the optical density at 570 nm was measured using a scanning multiwell spectrophotometer.
Immunohistochemistry on brain slices
The animals were sacrificed 21 daysafter GL261 tdTomato+luc+ cell implantation and perfused with 4% paraformaldehyde in phosphate-buffered saline (PBS). The brains were removed, fixed for 48 h in the same fixative solution and placed in 30% sucrose in PBS at 4°C until the tissue sank to the bottom of the flask. The tissue was frozen in Tissue Freezing Medium (Leica Biosystems, Richmond, IL, USA) and cut into 12 µm coronal sections using a cryostat. The slides were dried at room temperature for 2 h after being transferred from -80°C storage. The cryosections were blocked in PBS containing 10% donkey serum and 0.1% Triton X-100 for 2 h and incubated overnight at 4°C with rabbit anti-IBA1 and goat anti-Arg1 antibodies or FITC-conjugated Lycopersicon esculentum (Tomato) lectin (Vector Labs, FL-1171-1). Next, the sections were washed in PBS and incubated with the corresponding secondary antibodies for 2 h at room temperature. All the antibodies were diluted in 0.1% Triton X-100/PBS solution containing 3% donkey serum. Nuclei were counterstained with DAPI (0.001 mg/ml). Images were acquired using an Olympus microscope (Fluoview, FV10i) and a Leica DM4000B fluorescence microscope. For reagent specifications, catalog numbers, and concentrations, see Supplementary Table 1. We quantified the percentage of IBA1+ Arg1+ cells in 3 different areas of the tumor per animal. For lectin staining, three different sections from each mouse brain were analyzed. In each section, 2 regions of interest (ROIs) on the ipsilateral side (near the tumor core) were analyzed.
Tissue dissociation, flow cytometry and FACS sorting
On day 21 or 28 after GL261-tdTomato+luc+ or U87-MG RFP+ cell implantation, the mice were perfused transcardially with cold PBS prior to excision of the brain and spleen. The tumor-bearing hemispheres were dissociated enzymatically with collagenase type IV and DNase I (both from Merck, Darmstadt, Germany) at final concentrations of 2.5 mg/ml and 0.5 mg/ml, respectively, using gentleMACS Octo Dissociator (Miltenyi Biotec) according to the manufacturer’s protocol. Next, the enzymatic reaction was stopped by the addition of Hank’s Balanced Salt Solution with calcium and magnesium (Gibco, Germany). The resulting single-cell suspension was filtered through 70 μm and 40 μm strainers and centrifuged at 300 × g and 4°C for 10 min. Myelin was removed by density gradient centrifugation in 22% Percoll as described previously (27). Next, the cells were collected, washed with PBS and counted using NucleoCounter (Chemometec, Gydevang, Denmark).
The spleen was passed through a 70 μm strainer and gently grounded using the plunger of a syringe with 3 ml of PBS to yield a single-cell suspension. Following centrifugation (300 × g, 5 min), red blood cells were lysed using ACK Lysing Buffer (Life Technologies, Grand Island, NY, USA) for 10 min at room temperature. The splenocytes were collected via centrifugation, washed with PBS and counted using NucleoCounter (Chemometec, Gydevang, Denmark).
For preparation for flow cytometry analysis, samples were handled on ice and protected from light exposure. Prior to staining with antibodies, the samples were incubated with eFluor 506 fixable viability dye (Thermo Fisher) in PBS for 10 min. Next, the samples were incubated for 10 min with rat anti-mouse CD16/CD32 Fc Block™ (BD Pharmingen) in Stain Buffer (BD Pharmingen) to block FcγRIII/II and reduce nonspecific antibody binding. Then, the cell suspensions were incubated for 30 min with an antibody cocktail in Stain Buffer (BD Pharmingen) for the detection of surface antigens. For intracellular staining, the cells were fixed and permeabilized (Foxp3 fixation/permeabilization buffer, eBioscience) prior to incubation with antibodies. For a list of antibodies, see Table S1. For FACS sorting, the cells were stained with an anti-CD11b (M1/70 clone) antibody labeled with FITC (BD Pharmingen) and an anti-CD45 (30-F1 clone) antibody labeled with PE-Cy7 (BD Pharmingen).
For intracellular cytokine staining, freshly isolated cells from the tumor-bearing brain hemispheres were resuspended in stimulating culture media supplemented with 50 ng/ml PMA, 1 µg/ml ionomycin (Sigma Aldrich) and protein transport inhibitor cocktail (brefeldin A and monensin at final concentrations of 10.6 µM and 0.2 mM, respectively; Life Technologies, Carlsbad, CA, USA) for 4 h and then processed for staining of surface and intracellular antigens as described above.
All the antibodies were titrated prior to staining to establish the amount yielding the best stain index. Data were acquired using a BD LSR Fortessa Analyzer cytometer and analyzed with FlowJo software (v. 10.5.3, FlowJo LLC, BD). Gates were set on the basis of FMO (fluorescence minus one) controls and back-gating analysis. The percentages on the cytograms are given as the percentage of a parental gate. For computational analyses, each sample was downsampled to obtain 10000 CD45+CD11b+ or CD45+CD11b- cells. For tSNE, all samples were concatenated and processed using specific plugins in FlowJo v10.
CD11b+ cells were FACS-sorted from naïve or tumor-bearing hemispheres (pooled from 2 animals per sample at day 21 and from individual mice at day 28) using Cell Sorter BD FACSAria II. All flow cytometry experiments were performed at the Laboratory of Cytometry, Nencki Institute of Experimental Biology. For reagent specifications, catalog numbers and dilutions, see Supplementary Table 1. The gating strategies used in the analysis are shown in Fig. S9 and Fig. S10.
RNA isolation, mRNA library preparation and RNA sequencing
Immediately after sorting, CD11b+ cells were centrifuged and lysed for further isolation of RNA using the RNeasy Plus Mini Kit (Qiagen, Germany) according to the manufacturer’s protocol. The integrity and quality of the RNA were assessed on an Agilent 2100 Bioanalyzer with an RNA 6000 Pico Kit (Agilent Technologies, CA, USA). Strand-specific RNA libraries were prepared for sequencing (3–4 biological replicates/treatment) using a KAPA Stranded mRNA-Seq Kit (Kapa Biosystems, MA, USA). Poly-A mRNAs were purified from 100 ng of total RNA using poly-T-oligo-magnetic beads (Kapa Biosystems, MA, USA). mRNAs were fragmented, and first-strand cDNA was synthesized using reverse transcriptase and random hexamers. Second-strand cDNA synthesis was performed by removing RNA templates and synthesizing replacement strands, incorporating dUTP in place of dTTP to generate double-stranded (ds) cDNA. The dsDNA was then subjected to the addition of “A” bases to the 3′ ends and the ligation of adapters from NEB, followed by uracil digestion by USER enzyme (NEB, MA, USA). The amplification of fragments with adapters ligated at both ends was performed by PCR using primers containing TruSeq barcodes (NEB, Ipswich, MA, USA). The final libraries were analyzed using a Bioanalyzer and Agilent DNA High Sensitivity chips (Agilent Technologies, Santa Clara, CA, USA) to confirm the fragment sizes (~300 bp). Quantification was performed using a Quantus fluorometer and the QuantiFluor dsDNA System (Promega, Madison, Wisconsin, US). Libraries were loaded onto a rapid run flow cell at a concentration of 8.5 pM onto a rapid run flow cell and sequenced on an Illumina HiSeq 1500 paired-end platform.
Data processing and analysis
Illumina-specific adapters, short reads, and low-quality 5′ and 3′ bases were filtered out in the FASTQ files using the Trimmomatic [10.1093/bioinformatics/btu170] tool (version 0.36). The resulting RNA sequencing reads were aligned to a reference mouse genome sequence (mm10) with STAR aligner [28] (version 2.6.1b) using the two-pass Mode Basic option. Duplicate reads were then identified and flagged using Picard Tools (version 2.17.1) [broadinstitute.github.io/picard/]. The quantification of mapped reads and summarization by gene was performed using HTSeq-count (version 0.11.1), with paired mode (-p) and reverse stranded mode (-s reverse) enabled, and only reads with MapQ values of 10 or higher were considered. Low-expressed features were filtered out, and an analysis of differentially expressed genes was performed using NOIseq. Only mRNAs encoding protein-coding genes were retained for downstream analysis.
To identify transcriptomic differences between groups, differential expression analysis was performed using NOIseq methods, with the control (Vehicle) as the reference group compared with 7aaRGD, 7aaRAE or 7aaRGD, anti-PD-1 and combination of 7aaRGD+anti-PD-1. The variance stabilizing transformation (vst function) was used for visualization. Pathway enrichment analysis was performed using fgsea [29] on genes ranked by the fold change. Gene Ontology Biological Processes (GO:BP) was used to better understand the mechanistic findings of the enriched gene lists. The clusterProfiler, VennDiagram and ggplot2 R packages were used to visualize the data.
Bulk RNA-seq data deconvolution was performed using the bisque deconvolution tool [30] with default settings. Deconvolution was based on the cell type-specific transcriptomic signatures generated in our laboratory using CITE-seq on isolated brain CD11b+ cells derived from the same glioma model in mice [31].
Statistical analysis
All in vitro data represent at least three independent experiments performed in duplicate or triplicate. The statistical significance of the invasion assay results was calculated using the chi-square test. The numbers of animals per group are specified in the figure captions. Comparisons between two groups were performed with a two-tailed Student’s t test. For multigroup comparisons, one-way analysis of variance (ANOVA) was used, followed by Tukey’s HSD test. All the statistical analyses were performed using GraphPad Prism 7.0 software. P < 0.05 was considered statistically significant.