Reagents
PEI (25 kDa) was purchased from Sigma (St. Louis, MO, USA). Dextran (70 kDa) and N,N-carbonyldiimidazole (CDI) were purchased from Aladdin Reagent Co., Ltd. (Shanghai, China). Other chemical reagents were purchased from Sangon Biotech (Shanghai, China) unless otherwise stated.
Cationic polymer preparation
Cationic polymers were synthesized according to a previously reported method [17, 36]. Briefly, for cDex, 0.3 g of dextran dissolved in DMSO (30 ml) was incubated with 0.9 g of CDI for activation. Two hours later, 2.5 ml of anhydrous ethylenediamine was cautiously added dropwise for the cross-linking reaction. Then, the mixture was collected 24 h later and dialyzed against deionized water for 3 days. For DETA-Dex, the reaction was similar to that with cDex, only ethylenediamine was replaced with diethylenetriamine (DETA). After vacuum drying, the obtained cDex/DETA-Dex was kept in a dryer. The cationic polymers were characterized by 1H NMR (AVANCE III HD 400, Brook Corporation, USA), elemental analysis (Vario MICRO cube, Elementar, Germany), and Fourier transform infrared spectroscopy (NEXUS 870., NICOLET, USA) in the scanning range of 4000–400 cm−1.
To detect the degradability of the cationic polymers, cationic polymer (50 mg) was incubated with fresh mouse serum (5 ml) for 24 h. Then, the protein was removed by the Sevag method, and the cationic polymers were precipitated with acetone (PEI) or absolute ethanol (cDex and DETA-Dex). The cDex and DETA-Dex precipitates were dissolved in H2O (3 mg/ml) and passed through a 0.45 μm syringe filter for homogeneity and molecular weight analysis by the HPLC–RID method. The chromatographic conditions were as follows: an Agilent 1200 series LC system (Agilent Technologies, Palo Alto, CA, USA) was equipped with a TSK-GEL G4000PWXL column (300 mm × 7.8 mm, 10 μm) and a differential refractive index detector (RID; G1362A, Agilent Technologies, Palo Alto, CA, USA). During the experiment, the flow rate was maintained at 0.4 ml/min, and the column temperature was kept at 40 °C. For all separations, the mobile phase was ultra-pure H2O. Data acquisition and analysis were carried out using Astra software (version 6.0.2, Wyatt Technology Co., Santa Barbara, CA, USA).
In vitro phase separation assay
Fluorescent staining of RNA was performed using a standard protocol. Briefly, 4T1 cells were grown on glass coverslips to 80% confluence and then treated with 10 μg/ml cationic polymer (PEI, cDex, and DETA-Dex) in RPMI-1640 for the indicated times (1 h, 2 h, 4 h, 6 h, or 48 h). Cells were washed three times with cold PBS and fixed with 70% ethanol. Next, the cells were washed twice, and the detection reagent (500 nM SYTO RNAselect Green and 5 μg/ml DAPI nuclear stain in 1 × PBS) was added for 30 min of incubation at RT in the dark, followed by an additional three washes. After staining and sealing, the cells were analyzed under a Zeiss LSM980 microscope with a Zeiss Plan-Apochromat 63 × /1.4 oil objective. The progress of phase separation in vitro was recorded with a digital camera. Briefly, 200 μg of 4T1 mRNA in 100 μl of H2O was placed in a 0.2 ml polypropylene PCR tube, 20 μg of cationic polymer was added, and videos were recorded with a digital camera. To study the kinetics of the phase transition, the OD value at 600 nm was measured using a UV‒Vis spectrophotometer (UV-2550, Shimadzu). For cytoplasm RNase or protease treatment, 4T1 cell cytoplasm extract was treated with RNase A (20 μg/ml) at 37 °C and proteinase K (50 μg/ml) at 37 °C for 30 min, and then the supernatants were separated by centrifugation (12,000 rpm, 4 °C for 20 min). Each concentration consisted of triplicate samples, and each experiment was performed at least twice. The protein, RNA, and DNA contents in the droplets in the cytoplasm of the 4T1 cells were determined with a BCA protein quantitation kit and an iQuant™ High Sensitivity Quantitation Kit (GeneCopoeia).
In vitro and in vivo FRAP assays
For the in vitro RNA FRAP experiment, the droplets were photobleached with 100% laser power for 5 s using 488 nm lasers. Time-lapse images of the sample were collected after 100 ms of exposure time at 1 frame every 5 s using a Zeiss Plan-Apochromat 20 ×/1.0 water objective. Cellular FRAP was performed on a Zeiss LSM880 confocal microscope system at 37 °C in a live-cell imaging chamber. The RNA droplets were fully photobleached with 100% laser power for 2 s using a 488 nm laser. Time-lapse images were acquired over 5 min after bleaching. Images were processed by Zen Blue 3.1 software, and FRAP data were fitted to a single exponential model using GraphPad Prism 8.
mRNA sequencing microarray analysis
4T1 cells treated with cationic polymer for 24 h were flash-frozen in liquid nitrogen and stored at − 80 °C for microarray expression analysis. Total RNA from the cells was isolated using TRIzol according to the manufacturer’s instructions, and RNA concentration and purity were measured using a NanoDrop ND-1000 spectrophotometer (NanoDrop Technologies Inc., USA). The integrity of the RNA was determined by denaturing agarose gel electrophoresis. RNA samples were further purified, converted into cDNA, and labeled according to the Agilent One-Color Microarray-Based Gene Expression Analysis protocol (Agilent Technology). After hybridization and washing, the chips were scanned with an Agilent DNA Microarray Scanner. Agilent Feature Extraction software (version 11.0.1.1) was used to analyze the acquired array images. Quantile normalization and subsequent data processing were performed using the GeneSpring GX v12.1 software package (Agilent Technologies). After quantile normalization of the raw data, genes for which at least 1 out of 5 samples with detected flags (“all targets value”) were chosen for further data analysis. Differentially expressed genes between the two samples were identified through fold change filtering. Hierarchical clustering was performed using R scripts. GO analysis and pathway analysis were performed using the standard enrichment computation method. To determine whether specific biological pathways were differentially affected by the cationic polymers, we analyzed our microarray dataset using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database (http://www.genome.jp/kegg/pathway.html). The Jvenn (http://jvenn.toulouse.inra.fr/app/index.html) online tool was used for cross-analysis to obtain lists of the exclusive and common genes among the cationic polymers [37].
Dot blot hybridization
For RNA droplet preparation, 50 μg of 4T1 mRNA and 5 μg of cationic polymer were incubated for 30 min at 37 °C and then isolated by centrifugation at 15,000 rpm for 15 min. The precipitates were washed briefly three times with DEPC-treated ultrapure water and resuspended in 50 μl of water. For membrane preparation, the nylon membranes were soaked in 2 × SSC (1 × SSC comprising 0.15 M sodium chloride and 0.015 M sodium citrate, pH 7.0). Fifty microliters of RNA droplets were added to each dot with micropipette tips, and the membranes were dried for 30 min at room temperature and immobilized by UV exposure in a UV crosslinker. Then, the membranes were immersed in hybridization tubes with 10 ml of prehybridization solution preheated at 42 °C for 12 h in a shaking water bath. Portions (µg) of the probes were denatured in a boiling water bath for 10 min and then immediately placed in an ice bath for 5 min. The prehybridization solution was removed and replaced with 10 ml of hybridization solution containing 6 × SSC, 20% formamide, 5 × Denhardt’s solution, 0.5% SDS (w/v), 500 µg/ml yeast tRNA, and 30 ng/ml biotin-labeled DNA probes. The hybridization tubes were resealed and incubated for 3 h at 42 °C. The hybridization solution was then removed, and the membranes were washed briefly three times with washing buffer (2 × SSC-0.1% (w/v) SDS) for 10 min at room temperature. The membranes were then incubated in 10 ml of HRP-streptavidin (1:500) for 30 min at 42 °C and rinsed briefly three times with 10 ml of washing buffer for 15 min at room temperature. Enhanced ECL chemiluminescence detection solution (2 ml) was added to the tubes in the darkroom for analysis by scanning.
Mouse model
BALB/c mice and BALB/c nude mice (female, 6 weeks of age; Vital River Laboratory Animal Technology Co. Ltd., Beijing China) were housed in a specific pathogen-free (SPF) animal facility with controlled light (12-h light/dark cycles), temperature, and humidity conditions, and were fed a standard chow diet with water available. All experimental procedures involving animals were approved by the Institutional Animal Care and Use Committee of Nanjing University.
The mouse mammary carcinoma cell line 4T1 was obtained from ATCC (CRL-2539). Cells were grown and maintained in RPMI-1640 medium supplemented with 10% FBS. To generate the heterotopic tumor model, 1 × 106 cells were injected subcutaneously into the left armpits of the animals. We measured the tumor size with a Vernier caliper and weighed the tumor samples upon harvest. The tumor volumes were calculated as follows: tumor volume (mm3) = 0.5 × length × width × height. The antitumor activities of the cationic polymers were examined in heterotopic 4T1 tumor-bearing mice. When the tumor size reached approximately 0.5 cm in diameter, the tumor-bearing mice were injected with PBS, PEI, cDex, DETA-Dex, or Dex (3 mg/kg body weight) every 2 days. After drug administration, the tumor sizes were examined every 2 days. Fourteen days after the first administration of cationic polymer, all tumors were excised and weighed, and all tumor tissues were sectioned for histopathological and immunofluorescent analyses. Once the tumor size reached 1.3 cm, the study was terminated, and the mice were sacrificed. To evaluate the toxicity, healthy BALB/c mice were given the therapeutic agents via the caudal vein (5 mg/kg, i.v. injection), and the survival rate was recorded 30 min after administration.
To analyze the effect of the combination of the CPs and anti-PD-1 treatment, tumor model mice were randomly assigned to four groups (10 mice per group) via random lottery: the PBS group, DETA-Dex group, anti-PD-1 antibody group, or combined DETA-Dex and anti-PD-1 antibody group. Mice were injected with 3 mg/kg DETA-Dex and 5 mg/kg anti-PD-1 antibody (clone RMP1-14, Bioxcell) via intratumoral injection. On Day 14, we measured tumor sizes with calipers and weighed the tumor samples upon harvest.
Flow cytometry analysis
To prepare single-cell suspensions for flow cytometry, tumors were dissected into fragments and then digested with a protease cocktail containing collagenase IV (2 mg/ml) and DNase I (0.5 mg/ml) at 37 °C for 20 min and gently dissociated under a Miltenyi gentleMACS™ Dissociator and strained through 70 μm cell strainers. After red blood cell lysis, tumor-infiltrating leukocytes (TILs) were isolated using a tumor-infiltrating tissue leukocyte separation kit (WBC1092Z, TBD sciences, Tianjin, China) and blocked in 100 ml of 1% BSA for 30 min on ice.
Cells were incubated with FITC anti-mouse CD45 (#103,108, BioLegend, 2.5 μg/ml), BV421 anti-mouse CD4 (#100,438, BioLegend, 1.25 μg/ml), PE anti-mouse CD3 (#100,206, BioLegend, 2.5 μg/ml), APC anti-mouse CD8 (#100,712, BioLegend, 2.5 μg/ml), Brilliant Violet 711 anti-mouse CD3 (#100,241, BioLegend, 2.5 μg/ml), and APC anti-mouse CD25 (#102,012, BioLegend, 2.5 μg/ml). Cells were incubated with BV605 anti-mouse IFN-γ (#505,840, BioLegend, 2.5 μg/ml), BV711 anti-mouse IL-4 (#504,133, BioLegend, 5 μg/ml), and PE/Cyanine 7 anti-mouse IL17a (#506,922, BioLegend, 2.5 μg/ml) after using an Intracellular Staining Permeabilization Wash Buffer kit (#421,002, BioLegend). Cells were incubated with PE anti-mouse Foxp3 antibody (#320,008, BioLegend, 10 μg/ml) after using the True-Nuclear™ Transcription Factor Buffer kit (#424,401, BioLegend). Then, the cells were analyzed with a flow cytometer (Attune NxT device, Thermo Fisher Scientific). Examples of the gating strategy for flow cytometry analysis are shown in Additional file 1: Fig. S6. All antibodies and their isotype control antibodies were obtained from BioLegend (San Diego, CA, USA).
PI staining and flow cytometry
The cells were stained with propidium iodide (PI; Beyotime Biotechnology) by modification of a protocol described previously [16]. 2.5 × 105 4T1 cells was seeded in 6-well plates and incubated at 37 °C under 5% CO2 for 24 h. For PI staining, 1 ml of CPs/RPMI1640 solution was added after removal of cell culture media followed by addition of 5 μl of 10 mg/ml PI/H2O solution. After the incubation, cells were trypsinized with trypsin—EDTA and centrifuged at 300 g for 5 min. Resulting cell pellets were then resuspended in PBS with 0.1% BSA and then the cells were analyzed with a flow cytometer.
RNA isolation, qPCR, Western blot, and enzyme-linked immunosorbent assays
Total RNA from cells and tissues was extracted by using TRIzol reagent (Life Technologies). qPCR was performed by using ChamQ SYBR qPCR Master Mix (Vazyme Biotech Co., Ltd.) in a Step one™ Real-time PCR System (Applied Biosystems). Each sample was analyzed in triplicate and repeated in three independent assays. The level of each gene was normalized to that of GAPDH. The primers were synthesized by Sangon Biotech, and the primer sequences are shown in Additional file 1: Table S1.
Western blot analysis was performed according to the standard protocol. The protein samples were isolated with radioimmunoprecipitation assay (RIPA) buffer containing 1% protease inhibitor (Beyotime Biotechnology, Shanghai, China), and the total concentrations were detected using a bicinchoninic acid (BCA) protein assay kit (MicroBCA Kit, Thermo Scientific, USA). The proteins were mixed with SDS loading buffer and separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS–PAGE) and then transferred to polyvinylidene difluoride membranes (Bio-Rad, USA). The membranes were blocked with 5% skim milk and then incubated with primary antibodies (anti-TGFβ1, Ab ab179695, Abcam, USA; anti-β-actin, BM0627, Boster, China) overnight at 4 °C. The membranes were incubated with HRP-conjugated secondary antibodies (Life Technologies, USA) after three washes with PBST (PBS with 0.1% v/v Tween-20). After washing, the positive signals were visualized with fluorography using an enhanced chemiluminescence system (Cell Signaling Technology, USA).
Fresh tumor tissues were excised from the tumor-bearing mice 21 days after the tumors were established and then mechanically disrupted in PBS (0.1 g tissue/ml PBS) with EDTA-free protease inhibitor. Then, the tissues were homogenized in a Bead Beater apparatus (Tissuelyser-24, Jingxin Industrial Development Co., Ltd, Shanghai, China) with 5 mm beads for 30 s × 4 (60 Hz). The supernatants were separated by centrifugation (12,000 rpm, 4 °C for 20 min). The levels of the cytokines in the homogenates (TNFα, IL10, IL12, and TGFβ1) were then immediately measured with ELISA kits (eBioscience) following the manufacturer’s instructions.
H&E staining and immunofluorescent staining
Tumors were processed as described and then fixed in 4% paraformaldehyde, embedded in paraffin, and sectioned into 5 μm sections. The slides were counterstained with hematoxylin and eosin (H&E) for histologic analyses. Rabbit anti-mouse TGFβ1 was first applied to the sections, followed by FITC-labeled donkey anti-rabbit secondary antibodies (KPL, Gaithersburg, MD, USA). Slides were imaged with a Zeiss LSM980 microscope with a Plan-Apochromat 10 × /0.45 objective.
Statistical analysis
The results are expressed as the mean ± standard error of the mean (SEM). N refers to the number of animals per group. Differences between two groups were analyzed by two-tailed unpaired t test, and differences between groups were analyzed by one-way ANOVA with Dunnett’s tests. Two-way ANOVA with Dunnett’s multiple comparisons test and two-way ANOVA with Sidak’s multiple comparisons test were also used in this study. A value of p ≤ 0.05 was considered significant, ns, not significant. All statistical analyses were performed in GraphPad Prism 8.