Immunohistochemistry (IHC)
For IHC analysis of PFN2 or CD34, formalin-fixed paraffin-embedded (FFPE) sections were blocked with 5% bovine serum albumin (BSA) and incubated with mouse anti-PFN2 antibody (Proteintech, Rosemont, IL, USA) or mouse anti-CD34 antibody (Abcam, Cambridge, UK) overnight at 4°C. The sections were then incubated with biotinylated secondary antibodies, and detected using the Streptavidin-Peroxidase IHC assay kit and DAB (ZSGB-bio, China). Staining was evaluated by two certified pathologists, blinded to the experimental conditions, and scored according to staining intensity (negative: 0 points; weak: 1 point; moderate: 2 points; and strong: 3 points) multiplied by the percentage of stained cells (percentage of positive cells: ≤25%: 1 point; 26–50%: 2 points; 51–75%: 3 points; ≥75%: 4 points). Scores of <6 were considered low-expression, and scores of ≥6 were considered high-expression. Written informative consent was received from all participating individuals.
Lentiviral infection and transfection
Cells were plated at a density of 4 ´ 105/60 mm dish, 18 h prior to transfection. Transient transfection was performed using the Lipofectamine 2000 reagent (Invitrogen, Waltham, MA, USA) according to the manufacturer’s instructions. For stable transfection, cells were infected with lentivirus and selected using puromycin (5 mg/mL) according to the manufacturer's instructions. Transfection efficiency was determined using western blotting.
Escherichia coli containing the plasmids HA-PFN2, HA-PFN2-Mus, and Flag-PKM2 were purchased from LongQian Biotech (Shanghai, China). Lentiviral and plasmid constructs for PFN2 knockdown (hU6-MCS-Ubiquitin-EGFP-IRES-puro) were purchased from GeneChem (Shanghai, China). The shRNA targets were as follows:
shRNA#1: CGCGAAGAAATGCTCAGTGAT
shRNA#2: CCGGGAAGGTTTCTTTACCAA
Cell viability assay
Cell viability was evaluated using the cell counting kit-8 (CCK-8) assay (MedChemExpress, Monmouth Junction, NJ, USA). The cell suspension was inoculated into 96-well plates (2 × 103 cells/well) and CCK-8 solution (10 μL) added to the wells after 24 h. Following a 1 h incubation, absorbance was measured at 450 nm using a microplate reader.
Colony formation assay
For colony formation assays, cells were plated in 6-well plates (400 cells/well). After culturing for two weeks, cells were fixed with 4% paraformaldehyde and stained with 1% crystal violet. The colonies (>50 cells) were counted manually.
Tube-formation assay
Matrigel with reduced growth factor (100 μL/well) was added into 48-microwell plates and placed at 37°C for 30 min to solidify. Next, 4 × 104 human pulmonary microvascular endothelial cells (HPMECs) were resuspended in conditioned media from different cell types and added to the microwells. Cells were incubated for 6 h.
A droplet-based tumor angiogenesis model was established as previously described [37]. Tumor cells (TCs) were 3D cultured in Matrigel, whereas HPMECs were cultured as a monolayer. Both TCs and HPMECs were cultured in serum-free medium. After 24 h, TCs were transferred into the endothelial cell (EC) well, and a coculture unit was formed by fusing the two cell droplets through a reciprocating horizontal movement of the capillary between the two droplets.
Rhodamine phalloidin immunofluorescence staining used to assess tube formation. Angiogenic tube formation was quantified by calculating vessels area and total vessels length, counting the total number of junctions per field.
Tumor xenograft model
All animal experiments were approved by the Animal Care Committee of the Dalian Medical University (NO.00122713). BALB/c nude mice (7-weeks-old) were obtained from Liaoning Changsheng Biotechnology Co. Ltd. (Shenyang, China). A mixture of A549/shNC or A549/shPFN2 cell suspension in Matrigel (3:1, v/v) was injected subcutaneously into the dorsal flanks of nu/nu mice (3 × 106 cells/200 μL per mouse). Tumor xenografts were allowed to grow for 3 weeks. Tumors were measured every 3 days with calipers and calculated using the following formula: a2 × b × 0.5, where a is the smallest diameter and b is the diameter perpendicular to a. At the end of the experiment, the mice were sacrificed, and the tumors excised and weighed. All tissue samples were prepared into FFPE (4 μm) for later analysis by IHC.
Western blotting analysis
Whole cell and nuclear extracts were prepared. Protein samples were electrophoresed on SDS-PAGE and transferred onto polyvinylidene fluoride membranes (Merck, USA). The membranes were blocked with 5% skim milk in TBS containing 0.1% Tween-20 (TBST) and subsequently incubated with primary antibody, overnight at 4°C. Next, membranes were washed with TBST three times at room temperature and incubated with a horseradish peroxidase (HRP)-conjugated secondary antibody for 1 h, followed by three 10 min washes. Protein bands were visualized using an enhanced chemiluminescence (ECL) solution. Band intensities were quantified using the ImageJ software (NIH, Bethesda, MD, USA) and normalized to those of each control lane.
Immunofluorescence (IF)
Cells were cultured on glass-bottom culture dishes for 24 h and then fixed with 4% paraformaldehyde for 30 min followed by permeabilization with 0.5% Triton X-100 for 15 min. Cells were gently washed with PBS, blocked with normal goat serum for 40 min, and incubated with anti-PKM2 (1:50) antibody diluted in blocking buffer overnight at 4°C. After three 10 min washes, the cells were incubated with the corresponding fluorescence-conjugated secondary antibodies for 2 h at room temperature. After three more washes, the nuclei were stained with DAPI for 5 min. Cells were imaged using a confocal laser scanning microscope (Leica Microsystems, Wetzlar, Germany).
qRT-PCR
Total RNA was extracted using RNAiso Plus reagent (Takara Bio, Shiga, Japan) and assessed using a NanoDrop 2000 system (Thermo Fisher Scientific, Waltham, MA, USA) to determine RNA concentration and purity. cDNA was synthesized from 0.5–1 μg of total RNA using TransScript First-Strand cDNA Synthesis SuperMix (TransGen Biotech, Beijing, China). qPCR was performed using the TB Green Premix Ex Taq II (Takara Bio) on a LightCycler96 Real-Time System (Roche, Basel, Switzerland). The data were analyzed using the ΔΔCt method. All reactions were performed in triplicate, and mRNA levels were normalized to glyceraldehyde 3-phosphate dehydrogenase (GAPDH) mRNA expression. The primers used were as follows:
VEGF:
5'-TCACAGGTACAGGGATGAGACAC-3' (Forward)
5'-CAAAGCACAGCAATGTCCTGAA-3' (Reverse)
PFN2:
5'-GGCGTCTTTCAGAGCATTACG-3' (Forward)
5'-TGTCCATTGTGCAGTCACCAT-3' (Reverse)
GAPDH :
5'-ACCACAGTCCATGCCATCAC-3' (Forward)
5'-ATGACCTTGCCCACAGCCTT-3' (Reverse)
ELISA of human VEGF (hVEGF)
To compare the hVEGF secretion by A549 cells in 3D droplets and traditional Petri dishes, a 3D droplet array composed of A549 cells was generated. After culturing for 48 h, supernatants from 20 droplets were collected and diluted 50-fold with FBS-free medium in a 96-well plate. hVEGF was quantified using an ELISA kit specific for hVEGF (R&D Systems, Minneapolis, MN, USA) according to the manufacturer’s protocol.
Co-immunoprecipitation (Co-IP)
Co-IP assays were performed using the Pierce Classic Magnetic IP/Co-IP Kit (Thermo Fisher Scientific) according to the manufacturer’s protocol. Cell lysates from each sample were incubated with 10 μg of conjugated antibody, overnight at 4°C, to form the immune complex.
Liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis of PFN2-interacting proteins
To identify PFN2-interacting proteins, co-IPed proteins were visualized after separation by electrophoresis using a Coomassie Blue Staining Kit (Beyotime, Shanghai, China). The co-IP gel and its corresponding negative gel bands were excised into five parts according to the heavy and light chains and sent to Novogene (Beijing, China) for LC-MS/MS analysis. See ESM Methods for further details.
Surface plasmon resonance (SPR) assay
The binding affinity of recombinant hPFN2 (rhPFN2) to rhPKM2 was determined using an OpenSPR biomolecular interaction analyzer with a COOH chip (SEN-AU-100-10-COOH, Nicoya, Kitchener, Ontario). The rhPKM2 protein was dissolved in 10 mM acetate buffer (pH 3.5) and immobilized onto the chip. The sensor surface was activated by injecting a mixture of 50 mM N-hydroxysuccinimide (NHS) and 200 mM 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC), and blocking with 1 M ethanolamine, pH 8.5. Different concentrations of rhPFN2 (800, 400, 200, 100, 50, and 0 nM) were prepared in the running buffer (PBS). The interactions were determined at a flow rate of 20 μL/min for 240 s during the association phase, followed by 180 s for the dissociation phase at 25°C. The data were analyzed using the TraceDrawer manager software (Uppsala, Sweden). The binding kinetic parameters were calculated by global fitting of the kinetic data from various concentrations of rhPFN2 using a one-to-one analysis model.
Computational docking and molecular simulation
The crystal structures of PFN2 and PKM2 were obtained from the Protein Data Bank (PDB; http://www.rcsb.org/pdb/). The Rosetta software was used for protein docking, which consisted of two steps. Aggressive sampling was performed in the first step, using the centroid model. In the second step, an all-atom model was used for small-scale optimization. Because the binding mode between proteins was not clear, the blind docking method was adopted for the first docking between any systems, that is, random placement and random docking of two proteins. Docking results for 100 phases were obtained. First, blind molecular docking was performed on PFN2 and PKM2, and the active binding regions of the two proteins were set. The lowest docking energy among the 100 docking results between PFN2 and PKM2 was selected for further analysis.
Statistical analysis
Significance was calculated using GraphPad Prism software (GraphPad Software, San Diego, CA, USA). All statistical analyses were performed using data from at least three independent experiments. More than three means were compared using two- or one-way ANOVA with Bonferroni correction, and two means were compared using the unpaired Student’s t-test of *P<0.05 was considered significant.
For IHC, the results were analyzed using a nonparametric alternative to ANOVA. The Kruskal–Wallis H test was used to assess differences between the four independent groups; if there was a significant difference, we tested the differences between every two groups using the Mann-–Whitney U test.