BMSC isolation, culturing and identification
Tibia and femur from BALB/c mice were extracted under sterile conditions to expose bone marrow cavity, which we rinsed with saline. The bone marrow filtrate was gained and centrifuged at 225 × g for five minutes. Supernatant was discarded and we resuspended cells in low-glucose (LG)-Dulbecco's modified Eagle's medium (DMEM) (LG-DMEM; HyClone, Logan) at 1×106 cells per 100 μl. Cell suspension was added gradually to mice lymphocyte separation medium (Sigma-Aldrich) with 1:1 ratio and centrifuged at 1000×g for twenty minutes. Milky turbid mononuclear cell layer was obtained. Technician resuspended cells in LG-DMEM medium without FBS at 1×106 cells per 100 μl, which were centrifuged at 225 × g for five minutes. Pelleted cells were gained. Technician resuspended cells in LG-DMEM complete medium containing 10% FBS under 5% CO2 and saturated humidity at 37 °C. Culture medium was altered every 3d, which we subcultured with 1:3 ratio when cell confluency reached 80~90%. BMSCs at passages 3~4 were utilized for the following experiment. For phenotypic analysis, we used phycoerythrin (PE). The following marker expressions were studied: CD29, CD105, CD44, CD90, and von Willebrand factor (vWF). Immunoglobulin G-matched isotype served as internal control for every antibody.
Multilineage BMSC differentiation
To identify the BMSC multilineage differentiation capacity, we cultivated 3rd passage mouse BMSCs. For adipocyte differentiations, we cultured BMSCs in adipogenic differentiation medium. After 14 days, technician conducted oil red O staining to confirm adipocyte differentiation. For osteoblast differentiation, we cultured MSCs in osteogenic differentiation medium. After 3 weeks, we stained cells with alizarin red or ALP.
ADSC-Exo isolation and identification
Upon achieving 80~90% confluency, we rinsed BMSCs utilizing phosphate-buffered saline (PBS) and cultured them in EGM-2MV media without FBS and supplemented with 1× serum replacement solution (PeproTech, New Jersey, USA) for another 2d. Conditioned medium from BMSCs was gained and centrifuged at 300×g for ten minutes and 2000×g for 10 min to eliminate cellular debris and dead cells. Lastly, after centrifugation at 12, 000×g for 0.5 h. All processes were done at 4 °C. Exo protein content was defined employing Pierce BCA Protein Assay Kit (Thermo Fisher Scientific, MA, USA). BMSC-Exos were maintained at -80 °C or immediately utilized for downstream experiments. We applied transmission electron microscopy, NTA assay and western blotting to characterize Exos that collected.
Strand-specific NGS lib preparations
Our lab obtained total RNA from BMSC Exos or PBS-treated diabetic ulcer tissue utilizing TRIzol reagent (Invitrogen, Carlsbad, CA, USA). Technician used ~3μg total RNA from every specimen through VAHTS Total RNA-seq (H/M/R) Library Prep kits from Illumina (Vazyme Biotech Co., Ltd, Nanjing, China) to eliminate ribosomal RNA while retaining other RNAs such like ncRNAs and mRNAs. Our team treated RNA through 40 U RNase R (Epicenter) at 37 °C for 3 h, followed by TRIzol purification. RNA-seq libraries were prepared through KAPA Stranded RNA-Seq Library Prep kits (Roche, Basel, Switzerland), which we employed for NGS (Illumina HiSeq 4000 at Aksomics, Inc., Shanghai, China).
Bioinformatics analyses
Interactional correlations among RNAs were calculated via the website-based tool.
Cell culture and transfection
Our team obtained EPCs cells from ScienCell. We cultured cells in DMEM (HyClone) and 10% FBS (Gibco, Carlsbad, CA), which we stored in humidified incubator at 37 °C under 5% CO2. SLC7A11 gene and circ-Snhg11 overexpression vector were constructed by inserting circ-Snhg11 cDNA into the pcDNA3.1 vector. miR-144-3p mimics and SLC7A11 siRNA were synthesized by Genepharma (Suzhou, China). Cell transfection was made through Lipofectamine 2000 (Invitrogen) following protocols.
RNA isolation and real-time PCR
Total RNA was obtained with TRIzol Reagent (Invitrogen), followed by cDNA synthesis utilizing TransScript All-in-One First-Strand cDNA Synthesis SuperMix (Transgen Biotech, Beijing, China), which we made following [24]. PCR was made utilizing Bio-Rad PCR instrument (Bio-Rad, Hercules, CA, U.S.A.) using 2× Taq PCR Master Mix (Solarbio, Beijing, China) following protocols. Fold changes were computed by relative quantification means from 2−△△Ct method. The PCR primers are:
circ-Snhg11: forward 5′-GTTCTGTGATGGTTCCTC-3′ and reverse 5′-CAGCAGCGGAGTCCACG-3′; miR-144-3p: forward 5′-TCATGTAGTAGATATGACAT-3′ and reverse 5′-GGCAGGGTCCGAGGTATTC-3′; SLC7A11: forward 5′-ATACGCTGAGTGTGGTTTGC-3′ and reverse 5′-CTTCATCCACTTCCACAGCG-3′; GPX4: forward 5′-ATACGCTGAGTGTGGTTTGC-3′ and reverse 5′-CTTCATCCACTTCCACAGCG-3′; U6: forward 5′-CTCTCGCTTCGGCAGCACA-3′ and reverse 5′-ACGCTTCACGAATTTGCGT-3′; GAPDH forward 5′-GCAAGGATGCTGGCGTAATG-3′ and reverse 5′-TACGCGTAGGGGTTTGACAC-3′.
Dual-luciferase reporter assay
SLC7A11 and circ-Snhg11 3′UTR including binding sites that predicated for miR-144-3p were amplified by PCR. We inserted fragments into multiple cloning sites in pMIR-REPORT luciferase miRNA expression reporter vector (Ambion, Austin, U.S.A.). We co-transfected EPCs cells with 0.1 μg luciferase reporter vectors including WT or MUT types of the SLC7A11 or circ-Snhg11 3′UTR. The same to miR-144-3p mimic or miR-control using Lipofectamine 2000 (Invitrogen). Relative luciferase activity was computed by normalizing firefly luminescence to Renilla luminescence employing Dual-Luciferase Reporter Assay System (Promega, Madison, WI, U.S.A.) following protocols at 2d post-transfection.
Reactive oxygen species (ROS) analysis
Our lab captured ROS production in EPCs or scar skin employing 2',7'-dichlorofluorescin diacetate (DCF-DA, Molecular Probes, Eugene, OR, USA). Technician seeded EPCs (1×106 cells) in 100-mm culture dishes that pretreated through DAPI for 0.5~1 h and incubated with DCF-DA (20 mM) for 10 min at 37 ºC in dark. After 10 min incubation under normoxia or hypoxia, we examined cell fluorescence images to detect their ROS productions through fluorescence microscope (ECLIPSE E600, Nikon, Tokyo, Japan). Fluorescence intensity regarding DCF-DA was detected and computed by flow cytometry (BD Biosciences, San Jose, CA, USA). For tissues, the ROS were determined immediately after skin was excised from the mice.
Tubule formation assay
We assayed in vitro neovascularization in fibrin matrices. Following treatments, serum-starved EPCs in endothelial basal medium were seeded onto plates coated with Matrigel (105 cells / well into 6 wells) (BD Biosciences, Franklin Lakes, NJ, USA) and incubated at 37 °C for 0.5 d. Technician photographed tubular structures that generated in Matrigel by phase-contrast microscopy. Lengths of the newly formed tubes in 10 randomly selected fields in each well were detected.
Diabetic wound induction
We applied BALB/c mice (male) for diabetes induction through single intraperitoneal injection w.r.t. 60 mg/kg streptozotocin (STZ) dissolved in 0.1 M citrate buffer (pH=4.5). At 3d post STZ administration, diabetes was verified by fasting blood glucose level measurements regarding blood obtained from tail vein. Mice with fasting blood glucose levels >250 mg/dL were regarded as diabetic, which we kept for 1 month and for next-step studies once the blood glucose had stabilized. Following diabetes confirmations, rats were anesthetized by intramuscular injection of a ketamine hydrochloride and xylazine cocktail at 80 and 10 mg/kg, respectively. Hair was erased from dorsal leg region and the area sterilized with povidone iodine solution. A 4 mm full-thickness excisional wound was made utilizing sterile biopsy punch. Our lab randomly allocated mice for 200 μg subcutaneous injection BMSC Exos in 100 μL PBS or the equal volume of PBS alone at 4 sites around the wound (25 μL / site). We euthanized mice after 15 d and harvested skin specimens for histopathological validations.
Immunohistochemical data
Our team fixed tissue samples with 4% paraformaldehyde solution, which we embedded in paraffin. We obtained sections, which we cultured with primary antibodies against CD31 or GPX4 over the night at 4 °C with secondary antibodies (Abcam) for 1h at 37 °C. We stained sections with 3,3-diaminobenzidine, which we counter-stained via hematoxylin.
Statistics analyses
Continuous variables were represented by means ± SD and one-way variance analysis was utilized to compare by GraphPad Prism (GraphPad, La Jolla, USA). P ≤ 0.05 informed statistical significance.