Synthesis of hydrothermal condensed galactose nanoparticles (CG NPs)
25 mg of ortho-nitrophenyl-β-galactoside (ONPG) was dissolved in 10 mL of deionized (DI) water to yield a stock solution with a concentration of 8.3 mM. The solution was transferred to a Teflon-lined stainless-steel autoclave. The autoclave was cooled to room temperature after a hydrothermal reaction in an oven at 200℃ for 24 hours. Afterward, the resulting CG NP product was collected by centrifugation at 10000 rpm and redispersed with DI water three times. The purified CG NP sample was dissolved in 1 mL DI water for storage.
Synthesis of rhodamine B isothiocyanate (RITC)- and fluorescein isothiocyanate (FITC)-CG NPs
CG NPs (100 ppm) were mixed with 2 mL of RITC (0.02 mM). After a gentle spin at 35 rpm in a 4 ℃ environment, the RITC-CG NP product was obtained by centrifugation at 10000 rpm. The redispersion of CG NPs with DI water was repeated more than three times. The purified RITC-CG NPs were redispersed in 2 mL DI water for storage before use. The FITC-CG NPs were synthesized by the same experimental method used for RITC-CG NP synthesis. Fluorescence was measured by a microplate reader (Synergy H1, BioTek).
Synthesis of IONPs@CG
The synthesis of IONPs was carried out according to our previously published article21. The IONPs (25000 ppm[Fe]) and 25 mg of ONPG were dissolved in 10 mL of DI water. The solution was transferred to a Teflon-lined stainless-steel autoclave. After heating in an oven at 200 ℃ for 24 hours, the autoclave was cooled to room temperature. The IONPs@CG were collected by centrifugation at 10000 rpm and redispersed with DI water three times. The purified IONPs@CG was dispersed in 1 mL DI water for storage. The formula used for comparing the iron ion concentration (mg/mL) and solid content of IONP@CG was X ppm [Fe] = 0.002X mg/mL (IONP@CG)
CG NP stability
The CG NPs (100 ppm) were separated into different buffer solutions (H2O, PBS, medium, and H2O2). The solution was subjected to UV‒Vis spectral measurements over time (0, 4, 7, and 14 days). The size and morphology were observed by TEM and DLS on day 14.
Intracellular localization using fluorescence imaging
Macrophages were treated with RITC-CG and FITC-CG NPs for 24 hours. The macrophages were washed with PBS three times and fixed with 4% paraformaldehyde at 4°C for 10 min and 1% Triton X-100 for 15 min. Afterward, the cells were washed three times with PBS and incubated with DAPI (DNA nucleic acid stain) for 5 min. The stained macrophages were observed under a fluorescence microscope (Olympus IX73).
Cell culture
Raw264.7 (mice), THP-1 (human), bEnd.3 (human brain endothelial), GL261 (murine GBM), U-87 MG, T98G (mammalian GBM), and Pan02-Luc (murine PC) cells were purchased from American Type Culture Collection or Prof. Yan-Shen Shan (Dean, College of Medicine, National Cheng Kung University). These cell lines were subsequently cultured in high glucose Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (PS). SVG-p12 (human microglia) cells were cultured in Eagle's minimum essential medium supplemented with 10% FBS, 1% nonessential amino acids, and 1% PS. All cell lines were maintained in a 37°C incubator with a humidified 5% CO2 atmosphere.
3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay
Raw264.7, THP-1, mouse bone marrow-derived macrophage (BMDM), GL261, and SVG-p12 cells were cultured in 24-well plates at a density of 3×104 cells/well with 100 μl DMEM overnight. Cells were treated with different concentrations of CG or IONPs@CG (0, 5, 10, 20, 40, and 80 ppm) for 48 hours. After treatment, the cells were washed with fresh medium, and 200 μl of medium containing 0.5 mg/mL MTT was added to each well. After 2 hours of incubation at 37°C, 200 μl of DMSO was added to the wells to dissolve the MTT formazan for 5 min, and the supernatant was extracted and centrifuged at 1500 rpm for 5 min. Finally, the absorbance of each well was determined using an ELISA reader (Power Wave X340; Bio-Tek Instrument Inc., Winooski, VT, USA) at an excitation wavelength of 570 nm.
Isolation of bone marrow-derived macrophages
Mice were sacrificed by anesthesia overdose. Both the left and right femurs were isolated and placed on ice. Bone marrow in the femur bone was then added to a centrifuge tube with 5 mL of PBS using a 26 G needle. Red blood cells were lysed by ammonium-chloride-potassium lysing buffer, and a 70 μm strainer was used to filter the remaining cells. Following 1200 rpm centrifugation for 10 min, cells (1×106 cell/well) were seeded in 6-well plates filled with 2 mL DMEM and MCS-F (2 ng/mL) and stored in a 37°C incubator with 5% CO2.
CD11b/CD86/CD206 (M1/M2 macrophage) staining
Raw264.7, THP-1, and naive BMDMs were seeded in 6-well plates at a density of 2×105 cells/well with 2 ml DMEM overnight. LPS (20 ng/ml) and IFN-γ (20 ng/ml) were used to differentiate Raw264.7 into the M1 macrophages. After being induced into the M2 type by IL-13 (20 ng/ml) and IL-4 (20 ng/ml), Raw264.7-differentiated M2 macrophages were treated with CG (10 and 40 ppm), IONPs@CG (20, and 40 ppm) combined with anti-MCT-4 (1 mg) or not, respectively, for 72 hours. Cells were then collected for CD86 and CD206 (BD Pharmingen, San Diego, CA, USA) staining and incubated for 1 hour at 37°C. After staining, the intensity of CD11b, CD86, and CD206 was evaluated by flow cytometry.
Detection of reactive oxygen species (ROS) by immunofluorescence (IF) staining
GL261 cells (2×104/well) were seeded in chamber slides and treated with different concentrations of CG NPs or IONPs@CG (40 ppm) for 48 hours. Chamber slides were mounted with a mitochondria tracer (CellLight™ ER-RFP, BacMam 2.0) overnight. After treatment, cells were fixed with 4% formaldehyde. Then, the slides were permeabilized with 0.1% Triton-X100 for 10 min using the ROS peroxide-sensitive fluorescent probe 2′,7′-dichlorofluorescein diacetate (1:300, DCFH-DA, Molecular Probes) at room temperature in the dark for 1 hour and then covered with DAPI Fluoromount-G dye. Finally, the slides were photographed using a fluorescence microscope (ECHO Revolve).
Erythrocyte hemolysis
Blood was collected from the hearts of mice (C57BL/6) after anesthesia overdose. In brief, 200 ml of the erythrocyte stock dispersion was added to 200 ml of saline containing different concentrations of CG NPs (10 and 40 ppm) and IONPs@CG (20 and 40 ppm) and incubated in an Eppendorf tube at a mixing frequency of 414 rpm at 37°C for 1 hour. Intact erythrocytes were lysed by centrifugation at 10000 rpm for 5 min. Saline solution (0.9% NaCl) was employed as a negative control (0% lysis), and distilled water was employed as a positive control (100% lysis).
The equation of hemolysis rate:
where Dt, Dnc, and Dpc are the absorbance of the tested sample, negative control, and positive control, respectively40.
Enzyme-linked immunosorbent assay (ELISA)
Raw264.7 cells were seeded in 24-well plates at a density of 1×105 cells/well with 1 ml DMEM overnight, and the induction procedure was previously described. After being differentiated into M2 macrophages, cells were then treated with CG NPs or IONPs@CG for 72 hours; the supernatant was collected and used in the ELISA following the manufacturer’s procedures. Optical density was assessed at 450 nm and 570 nm. In the in vivo experiment, the supernatants from tumor homogenates were centrifuged in 3K centrifuge tubes (Pall Corporation) and analyzed with mouse TNF-alpha (TNF-α), IFN-gamma (IFN-γ), IL-12, IL-4, IL-10, and IL-13 DuoSet ELISA kits (R&D Systems) according to the manufacturer’s guidelines.
In vitro blood–brain barrier model
bEnd.3 brain-derived endothelial cells were seeded in the upper Transwell chamber (Franklin Lakes, NJ, USA) at a density of 1×105/well with 500 μl DMEM overnight to form monolayer cells. GL261 cells were seeded in the lower chamber at a density of 1×105/well with 500 μl MEM for one day. On the next day, FITC-CG or FITC-IONPs@CG NPs were added to the upper chamber and incubated for 48 hours41. After treatment, the GL261 cells were imaged under a fluorescence microscope (ECHO Revolve).
Multicellular tumor spheroid (MCTS)
GL261 cells were seeded in a MCTS chip at a density of 1×106 cells/well with 1 ml DMEM for one day and treated with different concentrations of CG NPs or IONPs@CG (40 ppm) for 24 hours. After treatment, the cells were fixed with 4% formaldehyde at room temperature for 15 min. Then, the cells were permeabilized with 0.1% Triton X-100 for 10 min and stained with 1 μg of DAPI in 300 μl of PBS for 20 min in the dark at room temperature42. Then, to hyalinize the cells, 300 μl of SCALEVIVEW (FUJIFILM) was added to the cells for 4 hours and incubated in a 37°C incubator. Finally, chips were photographed using a confocal system (ANDOR, Dragonfly 200) and quantified by ImarisViewer and ImageJ version 1.5.
Seahorse assay
GL261 cells were seeded at 2×104 cell/well in 24-well plates with 1 ml DMEM for 24 hours and treated with different concentrations of ONPG NPs (0 and 40 ppm) for 0 and 48 hours, and in 24-well plates, A1, B4, C3, and D6 cells were not seeded, as the temperature background correction on the machine. Then, following the procedure of the XF Mito Stress Test Kit, oligomycin (1 mM), FCCP (1 mM and 2 mM), and rotenone/antimycin A (0.5 mM) were added. Finally, the Seahorse XFe24 Extracellular Flux Analyzer was used to obtain the results43.
RNA sequencing
RNA was isolated from each tumor sample after treatment to identify the involved genes, molecules and mechanistic pathways. cDNA was prepared for sequencing based on the Geo-seq protocol. Sequencing libraries were generated by the TruePrep DNA Library Prep Kit V2 from Illumina (Vazyme) and evaluated by a Bioanalyzer (DNA HS kit, Agilent). RNA-seq data were then mapped to the GRCm38 murine genome by HISAT2 (version 2.1.0) with default parameters for murine samples. Differential expression and pathway analyses were conducted, with a focus on immune regulation. Gene set enrichment analysis (GSEA) was performed by GSEA software developed by UC San Diego and the Broad Institute44.
T-cell proliferation and activation analysis
Mouse PBMCs were cocultured with GL261 cells pretreated with CG NPs (40 ppm) or IONPs@CG (40 ppm) for 5 days. PBMCs were stained with CellTrace™ Violet reagent (Thermo Fisher) before coculture. The culture medium contained IL-2 (3.3 ng/ml) to promote T-cell proliferation. PBMCs supplemented with a CD3/CD28 stimulator were used as an activation positive control. Suspended immune cells were finally stained with CD8, CD4, CD62L, and CD44 to determine the proliferation of cytotoxic T cells or memory T cells. The percentages of these cell types were obtained using FACSCalibur flow cytometry, and data were analyzed by FlowJo software.
In vitro and in vivo phantom MRI
Based on our previously published work, IONPs@CG and cell phantoms were imaged using a Bruker PharmaScan 7T MRI scanner and a volume coil (inner diameter =72 mm, Bruker Biospin, Billerica, MA, USA). Localizers were used to verify the phantom and animal positioning in the center of the images. Multiecho T2*-weighted images with the same geometry were acquired using the fast, low-angle shot (FLASH) sequence with 30-35 echo sequences. The sequence parameters were as follows: repetition time (TR), 300 ms; first echo time (TE), 3.02 ms; echo spacing, 3.44 ms; in-plane matrix size, 256 × 256; and flip angle, 15°. All MRI data were processed using customized MATLAB scripts (The Mathworks Inc., Natick, MA, USA). We also calculated ΔR2* values using MR images. A region of interest (ROI) was drawn within the GBM margin in the visible tumor area of all tissue samples. The mean intensity levels of the T2* signal, weighted by area, were averaged across the measured samples for each pre- and post-IONPs@CG (3000 ppm) treatment image. The reciprocals of pre- and posttreatment T2* values were subtracted to obtain the ΔR2* value.
Perfusion-weighted MRI (PW MRI) analysis
Cerebral blood volume (CBV) was measured using T2* dynamic susceptibility-weighted contrast-enhanced perfusion-weighted magnetic resonance imaging (PW MRI). A gradient-echo echo-planar imaging sequence was used with the following parameters: FOV= 20 × 20 mm2, matrix size 128 × 128, TR/TE = 800/12 ms. A total of 75 time points were collected in one minute. The signal intensity change in the time series was converted into the relaxation rate by the equation ∆S(t)=TE-1{-ln[S(t)/S0]}, where S(t) is the signal intensity at time t and S0 is the precontrast signal intensity45. The effects of recirculation and contrast material leakage were reduced by fitting a gamma-variate function to the measured ∆S(t) curve46,47. Whole-brain CBV was calculated by fitting individual voxel time courses to a gamma variate function. To minimize the possible intersubject differences in those PW MRI-derived parameters caused by factors, such as brain topography and temperature, that might affect the absolute PW MRI-derived parameters, the change in CBV was expressed as a ratio of the lesion to the contralateral area (i.e., relative CBV, rCBV). A consistent ROI area was drawn in the healthy contralateral white matter, and the corresponding mean value of CBV in the ROI was calculated as the reference standard. On a pixel-by-pixel basis, the CBV maps were calculated first and then normalized by dividing every pixel CBV value by the reference value of the CBV calculated from the unaffected area to minimize variances in an individual animal.
Systemic and local immune alterations after CG NP or IONPs@CG treatment
After 21 days of treatment, immune cells from the bone marrow, spleen, tumor-draining lymph nodes (TDLNs) and infiltrating lymphocytes (TILs) were isolated from GBM model mice and stained with different markers to determine the proportions of myeloid-derived suppressor cells (MDSCs, CD11b+/Gr-1+), regulatory T cells (Tregs, CD4+/CD25+/FOXP3), dendritic cells (DCs, CD11c+/CD24+/MHCII+), natural killer (NK) cells (CD3-/NK1.1+/CD49b+), M1/M2 macrophages (CD11b+/CD86 or CD206) and cytotoxic T cells (CD8+/IL-2+ or CD8+/IFN-g+) and the systemic immune status. Local immune status was also validated by IF analysis of mouse tumor cryosections. For IF staining, brain tissue from mice was fixed with 4% formaldehyde, dehydrated with 30-50% sucrose, frozen at -80°C, embedded in OCT compound, and sectioned with a cryostat microtome. Brain tissue sections were then be stained for CD8, CD86, CD206, IFN-g, CD4, and FOXP3 and imaged by a fluorescence microscope (TissueFAXS-S PLUS).
Establishment of orthotopic Pan02-Luc pancreatic tumor model
All animal treatments and surgical procedures were performed following the guidelines of the National Cheng Kung University (NCKU) Laboratory Animal Center. C57BL/6 mice (8 weeks, male) were anesthetized using an intraperitoneal Zoletil 100 (Virbac) injection and placed in a face-up position. Then, 2 × 106 Pan02-Luc cells in a solution containing 10 μL of DMEM and 10 μL of Basement Membrane Matrix (BD Biosciences) were surgically implanted into the pancreas using BD 30 G 3/10 cc insulin syringes (BD Biosciences). The wound was sutured using CT204 Chromic Gut (20 mm, 75 cm, Unify Surgical Sutures Mfg. Co.) and NC193 monofilament nylon (19 mm, 45 cm, Unify Surgical Sutures Mfg. Co.) sutures, and the mice were allowed to rest until they fully recovered. The mice with orthotopic Pan02-Luc pancreatic tumors were treated with 100 μL of sterilized PBS (control) or 100 μL 1500 ppm CG NPs in sterilized PBS through intravenous injection once a week. Simultaneously, 12.5 mg/kg InVivoPlus anti-mouse PD-1 mAb (RMP1-14, BioXCell) was administered through intraperitoneal injection twice a week. Afterward, the body weight of the mice was measured, and the tumor growth of Pan02-Luc pancreatic cancer cells was monitored by the Xenogen IVIS Spectrum Noninvasive Quantitative Molecular Imaging System (Caliper Life Sciences) every 3 to 4 days. The maximum tumor burden permitted by the Institution Animal Care and Use Committee (IACUC) of NCKU was that the IVIS bioluminescence should not exceed 1×107 photon/sec/cm2/sr without ascites formation. All experimental mice with orthotopic pancreatic tumors were sacrificed before reaching the maximum tumor burden.
Establishment of the GL261-bearing GBM animal model and treatment procedure
Six-week-old male C57BL/6JNarl mice (20-25 g) were purchased from the National Animal Center and housed at the Animal Center of China Medical University. To establish an immunocompetent model, 1×105 GL261 cells intracerebrally implanted into the mice. Six-week-old male BALB/c nude mice were purchased from the National Animal Center to establish an immunodeficient model. Then, 5×106 GL261 cells were mixed with Matrigel phosphate-buffered saline (7:3) in a total volume of 100 μl and inoculated into the subcutaneous region of the right flank. After the tumor size reached 5 mm3 (C57BL/6JNarl mice) or 100 mm3 (nude mice), the mice were divided into 5 groups: control (0.1% DMSO), CG NP treatment (1500 ppm/treat/intravenous injection), 10F.9G2 (anti-PD-L1, 100 mg/treat/intraperitoneal injection), temozolomide (TMZ) treatment (50 mg/treat/gavage), IONPs@CG treatment (1500 ppm/treat/intravenous injection), CG NPs combined with 10F.9G2 and IONPs@CG combined with 10F.9G2 for 21 days. Additional anti-MCT-4 (0.5 mg/treat/twice per week) was used in combination with CG plus 10F.9G2.
Biodistribution test of IONPs@CG by radioactive isotope
125I (1 mL) was added to the IONPs@CG, and the reaction was performed on a 300 rpm shaker at room temperature for 30 min to produce 125I-IONPs@CG. Next, purification was performed by centrifugation in a 10k centrifuge tube (Sartorius Vivaspin®, Göttingen, Germany) at 12,000 rpm for 20 min. Afterward, the supernatant was removed, and the particles were suspended in normal saline. The process was repeated once. The radiolabeling yield and radiochemical purity were determined by an instant thin-layer chromatography system (AR-2000 radio-TLC Imaging Scanner, Bioscan Inc., Poway, CA, USA). Mice were anesthetized with isoflurane and intravenously injected with 125I-IONPs@CG (3.7 MBq). After 24 and 48 hours of injection, mice were sacrificed by CO2. The organs were excised, weighed, and assayed for radioactivity by a gamma counter (1470 Wizard, PerkinElmer, Waltham, MA, USA). The tissue activity was displayed as a percentage injected dose (ID) per gram of tissue (%ID/g).
Statistical analysis
Statistical analysis was performed using GraphPad Prism 7.0 software. One-way analysis of variance (ANOVA) with a Tukey's post hoc test was used to determine significant differences between more than two groups. The Student’s t test was used to determine significant differences between two groups. To ensure the reliability and consistency of our results, each group of cells or mice was independently tested with 3~6 separate samples.