Animals
Male Sprague-Dawley rats weighing 80-100 g (3 weeks old) and male ICR mice weighing 25-30 g (8-10 weeks old) were obtained from SIPPR/BK Laboratory Animal (Shanghai, China). The animals were kept at stable temperature (22 ± 2 °C) and humidity (60 ± 5%) condition, with 12-h light/dark cycle and free access to water and food. All animal care and experimental procedures were approved by the Zhejiang Chinese Medical University Animal Welfare Ethics Committee (reference number: ZSLL-2017-058), and performed in accordance with the NIH guidelines for the Care and Use of Laboratory Animals [29].
Cell culture
BMSCs were isolated based on our previously described method [30]. Briefly, the bone marrow was flushed with prechilled DMEM/F12 containing 1% (v/v) penicillin and streptomycin (Haotian Biological Technology, Hangzhou, China) from the femurs and tibias of rats. Then, centrifuged the bone marrow aspirates and suspended the cells in DMEM/F12 supplemented with 10% fetal bovine serum (FBS) (Gibco, Thermo Fisher Scientific, MD, USA). The medium was replaced 2 days after and every 3 days thereafter. The cells were passaged when 80-90% confluence was reached with a ratio of 1:2. The passage 3-4 BMSCs were used for the following experiments. To identify the BMSCs, cells were detected by flow cytometry using specific antibodies against cell surface markers, including CD34 (Santa Cruz, CA, USA), CD29, CD45 and CD90 (Biolegend, San Diego, CA,USA).
Human umbilical vein endothelial cells (HUVECs) obtained from American Type Culture Collection (ATCC, Manassas, VA, USA) were cultured in RPMI-1640 medium (Hyclone, UT, USA) containing 10% FBS and 1% antibiotics. Humidified condition at 37 °C with 5% CO2 was maintained. HUVECs were passaged every two or three days.
BMSC-Exos isolation, purification and identification
BMSC-Exos were isolated and purified by differential ultracentrifugation method [31]. Briefly, upon reaching 70-80% confluence, BMSCs were rinsed three times with PBS and cultured in fresh medium containing 10% Exos-free FBS medium (SBI Biosciences, CA, USA). The supernatants were collected after an additional 48 h incubation, and sequentially centrifuged at 300 × g and 2000 × g for 10 min to remove the dead cells. Then, the supernatant was centrifuged at 10,000 × g for 30 min at 4°C to clear the residual cellular debris and filtered with a 0.22-μm filter (Millipore, MA, USA). Afterwards, the filtrates were ultracentrifuged at 4°C and 100,000 × g for 2 h, followed by washing with PBS and ultracentrifuged at 100,000 × g for 2 h. At last, the pelleted Exos were resuspended in 100 μL PBS and quantified by total proteins using a Micro BCA Protein Assay kit (Thermo Scientific, IL, USA), then stored at -80°C.
For identification of Exos, the morphology, particle concentration, size distribution, and specific surface markers (CD9, CD63 and TSG101) of isolated Exos were detected by transmission electron microscope (TEM, Hitachi, Japan), nanoparticle tracking analysis (NTA), and western blot analysis respectively.
Focal cerebral ischemia model in mice
Focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) as previously described [32]. Briefly, male ICR mice were intraperitoneally anesthetized with 10% chloral hydrate at a final dose of 350 mg/kg. Then, a tip blunted and poly-L-lysine coated 6-0 nylon monofilament suture was advanced approximately 10 mm distal to the right carotid bifurcation to occlude the origin of the MCA. After 60 min of ischemia, the suture was carefully and slowly withdrawn. Sham-operated mice received identical surgery without inserting a suture. During the surgery, the rectal temperature of mice was maintained at 37°C with a homeothermic pad.
BMSC-Exos administration and BrdU labeling
To evaluate the therapeutic effects of BMSC-Exos, mice were allocated to four cohorts: sham operation group, MCAO group, 25 μg BMSC-Exos group and 50 μg BMSC-Exos group, and then were administered with Exos in 100 μL PBS or PBS alone by tail vein injection at 24 h after ischemia. To observe cell proliferation, mice were administered 50 mg/kg of 5-bromo-2-deoxyuridine (BrdU, Sigma-Aldrich, MO, USA) by initial intraperitoneal injection 24 h after ischemia, followed by daily consecutive injection for up to14 days.
Neurological function evaluation
The neurological deficit score was evaluated 1, 3, 7, and 14 days after stroke according to the Zea Longa score [33]: 0, no deficit; 1, failure to fully extend left forepaw; 2, circling to the left; 3, paresis to the left; 4, depressed level of consciousness and no spontaneous walking. The corner test was carried out as previously described [34]. Briefly, two 30 cm×20 cm×1 cm boards were attached to each other at an angle of 30° and with a small opening between the two boards. A mouse was placed into the central square facing the corner. When both sides of the vibrissae were stimulated by boards, the mouse then reared forward and upward, after which it turned back to face the open end. Each mouse was tested for ten trials, and the selected turning sides were recorded.
Infarct volume assessment
Mice were euthanized by overdose anesthesia with 10% chloral hydrate, and brains were moved and frozen immediately at -20°C for approximately 5 min, and then dissected into 1 mm-thick coronal slices. The slices were stained with 2% 2,3,5-triphenyltetrazolium chloride (TTC) for 15 min, and fixed with 4% paraformaldehyde for 24 h. The infarct volume was evaluated by Image J software.
Immunofluorescence staining
Mice were transcardially perfused with normal saline followed by 4% paraformaldehyde solution for 10 min at 14 days after MCAO. Brains were fixed overnight at 4°C, and then soaked in 30% sucrose solution. The brains were frozen and cut into 10-μm-thick frozen sections (Leica, Wetzlar, Germany). BrdU/von Willebrand factor (vWF) was detected by double immunofluorescence staining as described in our previous study [30].
BMSC-Exos uptake by HUVECs
To label Exos with green fluorescent dye, Exos were firstly resuspended with the PKH67 dye (Sigma-Aldrich, Munich, Germany) and incubated for 5 min, then terminated by 2% bovine serum albumin. Next, Exos were ultracentrifuged at 100,000 × g for 1 h to clear unbound dyes. HUVECs were incubated with PKH67-labelled Exos for 12 h followed by 4% paraformaldehyde fixation for 15 min and 4’,6-diamidino-2-phenylindole (DAPI) stain for 5 min. The internalization of PKH67-labeled Exos by HUVECs was observed using a fluorescence microscope (Leica, Wetzlar, Germany).
MiR-21-5p inhibitor transfection
The miR-21-5p inhibitor and the negative control (NC) were synthesized by RiboBio (Guangzhou Ribobio, Guangzhou, China). BMSCs at 80% confluence were transfected with 100 nM miR-21-5p inhibitor or NC, which were performed using Lipofectamine 2000 and Opti-MEM medium (Invitrogen, CA, USA) according to the manufacturer’s protocol. After 6 h, the transfection mixture was replaced by DMEM/F12 containing 10% exosome-free FBS. Conditioned medium of transfected cells was collected and centrifuged as described above.
MTT assay
The proliferation of HUVECs was measured using the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay (Beyotime, Shanghai, China). HUVECs plated on 96-well plates (1×104 cells/well) were cocultured with Exos (25 and 50 μg/mL) for 12, 24, 36, and 48 hours, and then 10 μL of MTT tetrazolium salt solution (5 mg/mL) was added into each well. After incubation for another 4 h, the formazan crystals were dissolved by adding 150 μL of dimethyl sulfoxide (Sigma, MO, USA). The optical density (OD) value of each well was measured at 490 nm by microplate reader (Tecan Austria GmbH, Grodig, Austria). Each experimental group was performed in six replicate wells.
Scratch wound healing assay
HUVECs were seeded into 6-well plates (5×105 cells/well) for confluence, followed by scratching with 200 μL pipette tip when reaching 90% confluence. Then, 2 mL of serum-free RPMI-1640 medium supplemented with Exos was added to each well. Images were captured at 0 h and 24 h after wounding. The migration rate (%) was calculated as follows: migration rate (%)= (initial wound area (t=0 h)–residual area (t=24 h))/ initial wound area (t=0 h)×100%. Each experiment was repeated three times.
Transwell migration assay
Transwell migration assay was carried out using 24-well chambers (8 μm, Corning, NY, USA). 500 μL of the RPMI-1640 containing 1% FBS was added into the lower chamber, and HUVECs (6×104 cells/well) suspended in 100 μL FBS-free medium were seeded in the upper chamber in the presence or absence of Exos. After 8 h for migration, nonmigratory cells were removed from the top of the insert membrane using humidified cotton swabs. The migrated cells at the bottom surface of membrane were fixed in 4% paraformaldehyde and stained with 0.1% crystal violet. The migrated cells were imaged and counted at 5 random fields. Three duplicates were set in each group.
Tube formation assay
After thawed overnight at 4 °C, 50 μL per well matrigel matrix (BD Biosciences, CA, USA) was added into precooled 96-well plates and incubated at 37°C to polymerize for 30 min. Next, HUVECs (2 × 104 cells/well) in FBS-free RPMI-1640 containing Exos were seeded onto matrigel-coated plates. Capillary-like tubular structures were captured after 6 h incubation. The total tube lengths from five random microscopic fields were calculated using Angiogenesis Analyzer Image J software.
Quantitative reverse transcription-polymerase chain reaction (qRT-PCR)
Total RNA from brain tissues of ischemic boundary region or cells was extracted using miRNeasy Mini Kit (Qiagen, Hilden, Germany). cDNA was produced from the total RNA using Mir-XTM miRNA First-strand Synthesis Kit (TaKaRa, Dalian, China). Subsequently, the product from reverse transcription was amplified with the SYBR Premix Ex Taq Kit (TaKaRa, Dalian, China) on an iQ5 real-time PCR detection system (Bio-Rad, CA, USA). Relative expression levels of miRNA were calculated by the 2−ΔΔCt and were normalized to U6. Each sample repeated 3 times and at least three samples obtained from independent experiments were examined. All primers used in this study are listed in Table 1.
Table 1 Primer sequences for qRT-PCR
Primers
|
Sequences
|
miR-21-5p
|
5’-CCGCGTAGCTTATCAGACTCAGACTGATGTTGA -3’
|
miR-22-3p
|
5’- CGAAGCTGCCAGTTGAAGAACTGT -3’
|
miR-486
|
5’- TCCTGTACTGAGCTGCCCC -3’
|
let-7i-5p
|
5’- GCGTGAGGTAGTAGTTTGTGCTGTT -3’
|
Note: The forward and reverse primers of U6, and the mRQ 3’Primer used as the universal reverse primer for above miRs, were supplied in the Mir-X miRNA First-strand Synthesis kit (Cat No. 638313)
Western blot analysis
Total protein was extracted with RIPA lysis buffer containing protease inhibitor PMSF ( Beyotime, Shanghai, China),and quantified using BCA protein assay kit (Beyotime, Shanghai, China). Firstly, equal amounts of protein were separated by 10% sodium dodecyl sulfate-polyacrylamide gel (SDS-PAGE) and transferred to PVDF membranes (Millpore, CA, USA). After being blocked with 5% skim milk in TBST, the membranes were incubated with the following primary antibodies overnight at 4°C: CD9 (1:1000; Bioworld, MN, USA), CD63 (1:1000; Bioworld, MN, USA), TSG101 ( 1:1000; Abcam, MA, USA ), VEGF (1: 500; Santa Cruz, CA, USA), VEGFR (1:500; Abcam, MA, USA ), Ang-1 (1:1000; Santa Cruz, CA, USA), Tie-2 (1:1000; Santa Cruz, CA, USA) and GAPDH (1:1000; Santa Cruz, CA, USA), followed by incubation with corresponding secondary antibodies at room temperature for 1 h. Signals were visualized by enhanced chemiluminescence detection kit (Millpore, CA, USA). Relative expression levels were normalized to levels of GAPDH.
Statistical analysis
Data were analyzed by SPSS software (version 25.0, SPSS, IL, USA) and presented as mean ± standard deviation (SD). Neurological deficit and corner test data were analyzed by nonparametric Kruskal-Wallis H test. One-way analysis of variance (ANOVA) (Student-Newman-Keuls) statistical differences between groups was used for analyzing all other data. P<0.05 was considered statistically significant.