2.1. Ethics Statement.
Adult male Sprague-Dawley rats (6-8 weeks old, weighing 220 ± 20g) were purchased from the Animal Centre of Nanjing First Hospital, Nanjing medical university (Nanjing, China) and raised in specific pathogen-free animal cages under constant temperature and humidity and a 12h/12h dark/light cycle with adequate food and water. All experimental protocols with regard to the use of animals were approved by the Institutional Animal Care Committee of Nanjing First Hospital. All animal experiments were performed in accordance with the guidelines of decreasing the amount of suffering, pain, and discomfort of the experimental animals.
Human clinical samples were obtained from patients who underwent total knee arthroplasties after informed consent and approval from the Ethics Committee of Nanjing First Hospital (Nanjing, China). Only patients with primary knee OA were selected, and patients with inflammatory arthritis or with a history of prior knee surgery were excluded.
Isolation of human ADSCs.
Donor-matched infrapatellar fat pad and subcutaneous fat were obtained from OA patients. Briefly, as for isolation of hADSCs-IPFP and hADSCs-ScAT, the infrapatellar fat pad and subcutaneous fat were harvested and washed in phosphate-buffered saline (PBS), and then finely diced into small pieces by using a surgical scissor. The diced tissues were digested in 0.1% type I collagenase (Sigma, America) for 10 h, and the cell suspension was filtered through a 40-μm cell strainer (BD Bioscience). The released cells were centrifuged at 500 g for 10 min and resuspended in Red Cell Lysis Buffer (Beyotime, China) at room temperature for 10 min. The cells were then centrifuged again, and resuspended in Dulbecco's modified Eagle's medium (DMEM)/F12 (Invitrogen, America) supplemented with 10% fetal bovine serum (FBS; Gibco, America) and 1% P/S. The medium was changed every other day.
Isolation of mouse ADSCs.
8-week-old C57/B6J mice were sacrificed by cervical dislocation under isofurane anaesthesia. Adipose tissue was harvested from the inguinal areas and infrapatellar fat pad used in all experiments. Mouse ADSCs (mADSCs) were isolated from each adipose tissue sample. Briefly, adipose tissue was washed with phosphate-bufered saline (PBS) (pH 7.4) and cut into 0.5–1.0 mm small fragments. The tissues were then transferred into 15-mL tubes. Type I collagenase (1.0 mg/mL was then added to the tube at an identical volume. The mixture was immediately agitated using a water bass shaker (150 rpm) at 37 °C for 30 min. The digested tissue was filtered through a 40-μm cell strainer and centrifuged at 200×g for 5 min. The supernatant was aspirated and the cell pellet was resuspended in erythrocyte lysis buffer (168 mM NH4Cl, 10 mM KHCO3, and 0.1 mM EDTA4Na; 10 mL) at 4 °C for 10 min. After erythrocyte lysis, 5 mL of medium was added, and the tube was centrifuged at 200×g for 5 min. The cell pellet was resuspended in medium and filtered through sterilized 100- and 40-μm cell strainers (Corning Inc., NY, USA). The mADSCs were cultured and used in the third passage.
Identification of ADSCs.
For detecting the classical biomarkers of ADSCs, we performed flow cytometric analysis by the following fluorescein isothiocyanate- (FITC-) conjugated or phycoerythrin-(PE-) conjugated antibodies: CD29, CD44, CD90, CD105, CD73, CD34, and CD45 (Becton Dickinson, San Jose, CA, USA). The FITC-IgG and PE-IgG isotypic immunoglobulins were detected as isotype controls. After incubation, cells were washed twice and finally suspended in FACS buffer for flow cytometry analysis (BD Biosciences).
The multipotent differentiation potential of ADSCs to differentiate to osteoblasts, adipocytes, and chondroblasts was evaluated. After subculturing to the third generation, the culture medium was replaced with osteogenic, adipogenic, or chondrogenic differentiation complete medium (Cyagen Biosciences, China). After the induction of the differentiation cultures for 14 days, the accumulation of calcium, intracellular lipids, and mucopolysaccharides was estimated by the alizarin red staining, oil red O staining, and Alcian blue staining (Sigma-Aldrich, USA), respectively.
Isolation of human-chondrocyte.
Primary human OA chondrocytes were isolated from leftover pieces of osteoarthritic cartilage from total knee replacement surgery of seven OA patients, Articular cartilage was removed aseptically from subchondral bone, cut into pieces, and washed with PBS, then digested overnight in high-glucose DMEM (Invitrogen, America) supplemented with 0.2% type II collagenase (Sigma, America) and 1% P/S. The cell suspension was filtered by a 40-μm cell strainer; and the collected cells were centrifuged at 400 g for 5 min, and then resuspended in high glucose DMEM supplemented with 10% FBS and 1% P/S. The medium was replaced every other day. Chondrocytes were induced with IL-1β (10 ng/ml) for 24 h after transfection for subsequent experiments.
Isolation of mouse-chondrocyte.
To extract chondrocytes, 3-week-old C57/B6J mice were sacrificed for collection of cartilage from knees. First, cartilage was into small pieces after washing with PBS. Second, the samples were digested in 0.25% trypsin-EDTA (Gibco, USA) solution for 5 min, and DMEM-F12 (Gibco) containing 0.2% collagenase type II (Sigma-Aldrich) for 6 h at 37 °C, successively. The released chondrocytes were seeded in T25 cell culture flasks. Cells were passaged at a ratio of 1:3 at 80% confluence. The culture medium was refreshed every 3 days. Chondrocytes were induced with IL-1β (10 ng/ml) for 24 h after transfection for subsequent experiments.
Exosome Extraction and Identification.
The Exos derived from ADSCs were isolated as previously described. In brief, culture media supplemented with exosome-depleted FBS (SBI, USA) were used for cultivating ADSCs. Subsequently, conditioned supernatants from ADSCs cultures were collected and centrifuged such to remove dead cells or debris. The collected medium was subjected to ultracentrifugation at 110,000 g for 2 h at 4℃ after filtration with a 0.22-mm filter. The exosome-containing pellet was washed and resuspended with PBS and stored at -80°C prior to further analysis. The total protein contents of the Exos were evaluated.
Analysis of particle size and intensity was conducted with nanoparticle trafficking analysis (NTA) system (NanoSight NS300). After isolation, the Exos were diluted infiltered PBS before administration. Samples were administered and recorded under controlled flow by the NanoSight syringe pump. Automatic settings were performed to measure the minimum particle size and track length. The measuring conditions were temperature 23.75 ± 0.5°C, 25 frames per second, and measuring time 60s. The detection threshold was uniform in the different groups.
For morphologic observation with transmission electron microscopy (TEM), exosome pellets were seeded on formvar carbon-coated 200-mesh copper electron microscopy grids, placed at room temperature for 5 min, and then were stained with aqueous uranyl acetate. The grids were washed with PBS and continued to semidry at room temperature prior to detection under TEM (Hitachi, H7500 TEM, Tokyo, Japan).
The CD63, CD9, TSG101 and Calnexin biomarkers were measured with western blotting analysis. Exos were collected and resolved by SDS/PAGE and then transferred to PVDF membranes (Millipore, Billerica, MA, USA). The membranes were blocked by 5% nonfat milk in TBST buffer and incubated overnight using rabbit anti-CD63 (1:300, Proteintech, China), CD9 (1:1000, Proteintech, China), TSG101(1:2000, Proteintech, China), Calnexin(1:5000, Proteintech, China) separately, then washed with TBST, and incubated continuously using HRP linked goat anti-rabbit IgG (1:5000, keygen, China), and the protein intensity was determined with the automatic imager (General Electric, Fairfield, CT, USA).
Exosome Label and Track.
To determine whether ADSCs derived-Exos can be taken up by chondrocytes, Exos were labeled with PKH26 (Sigma-Aldrich, USA) following the protocol recommended by the manufacturer. which allow us to fluorescently label isolated Exos to track cellular interaction and uptake. Exos were co-cultured with chondrocytes then fixed with 4% paraformaldehyde. The nuclei were stained with Hoechst 33342 (Beyotime, China). The cytoskeleton was stained by Actin-Tracker Green (Beyotime, China). The uptake of exosome was observed using a confocal laser scanning microscope (Zeiss LSM710, Germany).
Determination of Cell Viability.
Cell viability was quantitatively evaluated by Cell Counting Kit-8 (Beyotime, China). In brief, chondrocytes (1 × 104) were planted in 96-well microculture plates incubated overnight. Then, the viability assay was conducted, and absorbance was assessed using a microplate reader at a wavelength of 450 nm according to the manufacturer’s instructions with some modification. Three independent experiments were performed, and cell viability of different groups was evaluated as a percentage of the control.
Migration assay
Migration in conditioned chondrocytes was detected using a Transwell system. Briefly, 5 × 104 chondrocytes were placed in a 24-well transwell plate with 8 μm pore size inserts (Corning, NY, USA,). Then, 500 μL of DMEM containing 0.5% FBS and 1% PBS with 5 μg Exos was added to the lower chamber before culturing for 16 h. The cells in the upper chamber (2 × 105 cells) were then placed in 4% paraformaldehyde for 20 min and mixed with 0.5% hematoxylin and eosin for 10 min. The percentage of cell migration in each well was determined in a blinded manner.
Cell apoptosis analysis
chondrocytes were plated in 6-well plates at a concentration of 2 × 105/well and were treated with or without ADSC-Exo (5ug). Cell apoptosis was analyzed by using an Annexin V assay kit (Keygen, China) and then measured by a Beamcyte-1026® flow cytometer (BDA Inc., China).
Exosomal small RNA sequence assay
The small RNA sequence for hADSCsScAT Exos and hADSCsIPFP Exos were performed by OE Biotech Company (Shanghai, China). Three samples were processed for each type of exosome. The fragmentation mixtures were hybridized to an Agilent-Human microRNA array 21.0 (8 ×60 K, Design ID:070,156). For microarray analysis, the Affymetrix (Santa Clara, CA, USA) miRNA 4.0 platform was employed. The sample labeling, microarray hybridization and washing were performed based on the manufacturer’s instructions (Agilent Technologies Inc., Santa Clara, California, USA). Differentially expressed miRNAs were identified using a fold change cut off value of ≥1.5 set for both up- and down-regulated genes.
Luciferase Reporter Assay.
Luciferase vectors including the 3′-UTR of ADAMTS4 containing the ADAMTS4-miR-99b-3p response elements and the mutant were obtained from GenePharma. Either miR-99b-3p mimic and negative control was then transfected into the human embryonic kidney (HEK) 293 cells in the presence of either the wild-type or the mutant reporter plasmid. Luciferase activity was determined with the Dual-Luciferase reporter system (Vazyme), and Renilla luciferase activity was set as internal control.
miRNA expression and transfection, miRNA abundant Exos isolation
miR-99b-3p-mimic, miR-mimic-NC, were constructed by RiboBio (Guangzhou, China). The ADSCs and chondrocytes were transfected with mimics at a density of 4 × 105 each well in six-well plates using Lipofectamine 2000 reagent (Invitrogen).
The Exos derived from miR-99b-3p-mimic transfected ADSCs were isolated as previously described.
Plasmid transfection assay
The pcDNA3.1 vector and pcDNA3.1 vector for ADAMTS4 overexpression commercially were prepared by General Biosystems (GenePharma, China). These plasmids were transfected into chondrocytes with the Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA).
Synthesis of HB-PEGDA polymers
Vinyl terminated hyperbranched PEG polymers (HB-PEGDA) were prepared via an in situ reversible addition−fragmentation chain transfer polymerization (RAFT) strategy.[37] Briefly, using 2’-azobis(2-methylpronitrile) (AIBN) as the initiator and tetraethylthiuram disulfide (DS) as the RAFT agent precursor, the polymerisation of polyethylene glycol diacrylate (PEGDA, average Mn = 700) (0.4 mol·L−1) was performed in 75 mL of butanone at 70 °C with a feed ratio of [PEGDA] : [DS] : [AIBN] at 25:1:1.8. Gel permeation chromatography (GPC, Agilent 1260 Infinity II) was used to monitor the evolution of polymer molecular weight during the polymerization. Unless otherwise stated, the following hydrogel syntheses in this work were performed using HB-PEGDA polymer with a molecular weight of 15 kDa. NMR data were collected by a Bruker AVANCE NEO 400 MHz NMR spectrometer.
Synthesis of SH-HA
SH-HA was synthesized using a previously described method.[40] Briefly, HA (500 mg, 1.25 mmol) was dissolved in 10 mL MES buffer (pH 4.75, 0.1 M) with DTP (595 mg, 2.5 mmol) and EDCI (480 mg, 2.5 mmol). The mixture was stirred for 5 h before 1 M NaOH was added to adjust the pH to 7.0 for quenching the reaction. Then, DTT (501 mg, 3.25 mmol) was added, after which the pH of the solution was raised to 8.5 using 1 M NaOH. The mixture was stirred for 24 h. At the end of the reaction, the pH of the solution was adjusted to 3.5 by 1 M HCl. The acidified solution was transferred to dialysis tubes (Mw cutoff = 12 kDa) and dialyzed against dilute HCl (pH 3.5) containing 100 mM NaCl for 4 times. Finally, the solution was centrifuged, and the supernatant was lyophilized to obtain the white fluffy product.
Preparation of HMPs through the microfluidic droplet technique
HMPs were prepared by a Michael addition reaction between HB-PEGDA and SH-HA within discrete droplets produced by a microfluidic droplet maker.[41] Briefly, in a typical experiment, aqueous dispersions containing 1 % (w/v) SH-HA and 3.33 % (w/v) HB-PEGDA were intersected by the fluorinert TM FC-40 oil containing 2 wt% Pico-SurfTM in the microfluidic droplet maker to generate uniform microdroplets. The as-obtained microdroplets were left at room temperature for 1 hour to ensure adequate gelation. Then, to purify the HMPs, 20% v/v 1H, 1H, 2H, 2H-perfluoro-1-octanol (PFO) in Novec 7500 oil was added into the samples to remove the surfactant. The purified HMPs were soaked and washed three times with PBS buffer, and then dispersed in the PBS buffer for later use.
Hydrogel Rheological Characterization
Rheological properties of the samples were assessed using a stress-controlled rheometer (HAAKE RheoStress 1, Thermo Scientific, USA) fitted with an 8 mm diameter flat plate (2 mm gap) at 25℃. Prior to rheological testing, the granular hydrogel prepared by centrifuging HMP dispersion at 104 rpm for 15 min were carefully pipetted into the plate gap. Dynamic oscillatory strain amplitude sweep measurement was conducted by varying % strain from 0.1 to 1000% at a frequency of 1 Hz, and oscillatory frequency sweeps were conducted ranging from 0.1 to 100 rad/s at 1% strain amplitude.
Exos encapsulation efficiency
The amount of remaining Exos in the solution was detected by BCA protein assay. The following formulas were used for calculating the Exos encapsulation efficacy (EE):
Cell cytotoxicity and compatibility
Chondrocytes were seeded in a 96-well plate at a density of 1×104 cells/well and cultured overnight, HMPs, HMPs@ExosScAT, HMPs@ExosScAT-99b-3p were added to each well. After 24 h, cell viability was conducted using the Cell Counting Kit-8 (CCK-8, Beyotime, China) assay.
In addition, the cyto-compatibility was assessed by Calcein AM/PI staining assay. chondrocytes at passage three were seeded in 24-well plates at a density of 5×104 cells/well and cultured overnight prior to the addition of 1-5 mg/mL. After 1, 2, and 3 days, the viability of the cells was analyzed using the Calcein/PI Cell Viability/Cytotoxicity Assay Kit (Beyotime, China).
Immunofluorescent Staining.
For cell immunofluorescence, chondrocytes were incubated in complete culture medium with various Exos for an additional three hours. The cells were then washed in phosphate-buffered saline (PBS) and fixed with 4% paraformaldehyde for 15 min. The fixed cells were permeabilised with 0.1% Triton X-100 for 5 min and washed with PBS three times. Then, the cells were treated with primary ADAMTS4, ACAN, COMP antibodies (Abcam, Cambridge, UK) overnight at 4°C in PBS supplemented with 1% bovine serum albumin.
Immunohistochemistry staining
Immunohistochemical analysis was performed as described previously [31]. Briefly, samples were fixed in 4% paraformaldehyde (Sigma-Aldrich, St. Louis, MO, USA), decalcified, embedded in paraffin, and cut into 5-μm sections that were deparaffinized, rehydrated, and then incubated with primary ADAMTS4, ACAN, COMP antibody (Abcam, Cambridge, UK) overnight at 4 °C. Sections were incubated with secondary antibody at 37 °C for 1 h and with 3,3-diaminobenzidine (DAB) for 3 min.
Evaluation of an in Vivo Imaging System (IVIS)
To determine the residence time of Exos, ExosScAT-99b-3p, HMPs@Exos and HMPs@ExosScAT-99b-3p in the articular cavity. Exos were labelled with PKH26. Exos (50μg), ExosScAT-99b-3p (50ug), HMPs@exo (50ug Exos/), HMPs@ExosScAT-99b-3p were injected into right knee of mice, respectively. Then, the mice were scanned by the IVIS Lumina II system (PerkinElmer, USA) to observe the retention time of Exos in all groups.
Quantitative Real-Time Polymerase Chain Reaction (RT-qPCR).
After harvesting the chondrocytes and separating the total RNA by TRIzol Reagent (Invitrogen), and reverse transcribed into complementary DNA according to the manufacturer’s protocol of the reverse transcription kit (R312, Vazyme, China). qPCR was carried out following the instructions provided by the Taq Pro Universal SYBR qPCR Master Mix (Q712, Vazyme, China) using a real-time fluorescence quantitative PCR appliance (QuantStudio®5, ThemoFisher Scientific, USA). Otherwise, the expression of miRNAs was quantified by miRNA Universal SYBR qPCR Master Mix (Vazyme, China) and miRNA 1st Strand cDNA Synthesis Kit (Vazyme, China) was applied for the single stranded cDNA synthesis. GAPDH and U6 were taken as internal reference genes. All the primers for RNAs are listed in Additional file: Table 1.
Table 1 Primer sequence for RT-qPCR
Genes
|
Primer sequence
|
miR-381-3p
|
F:CGGGCTATACAAGGGCAAGC
R:CAGCCACAAAAGAGCACAAT
|
miR-224-5p
|
F:CGGGCTCAAGTCACTAGTGGTTC
R:CAGCCACAAAAGAGCACAAT
|
miR-99b-3p
|
F:CGGGCCAAGCTCGTGTCTGT
R:CAGCCACAAAAGAGCACAAT
|
miR-27a-5p
|
F:CGGGCAGGGCTTAGCTGCTT
R:CAGCCACAAAAGAGCACAAT
|
miR-889-3p
|
F:CGGGCTTAATATCGGACAA
R:CAGCCACAAAAGAGCACAAT
|
miR-424-3p
|
F:CGGGCCAAAACGTGAGGCG
R:CAGCCACAAAAGAGCACAAT
|
miR-744-5p
|
F:CGGGCTGCGGGGCTAGGGCT
R:CAGCCACAAAAGAGCACAAT
|
miR-483-5p
|
F:CGGGCAAGACGGGAGGAAAG
R:CAGCCACAAAAGAGCACAAT
|
miR-222-3p
|
F:CGGGCAGCTACATCTGGCT
R:CAGCCACAAAAGAGCACAAT
|
miR-503-5p
|
F:CGGGCTAGCAGCGGGAACAGT
R:CAGCCACAAAAGAGCACAAT
|
miR-29a-5p
|
F:CGGGCACTGATTTCTTTTGG
R:CAGCCACAAAAGAGCACAAT
|
U6
|
F: 5′-GGTCGGGCAGGAAAGAGGGC-3′
R: 5′-GCTAATCTTCTCTGTATCGTTCC-3′
|
Western Blot Analysis
The proteins were harvested and lysed in RIPA buffer. Equal amounts of protein extracts (30 μg) were loaded per lane and resolved by SDS/PAGE. Subsequently, polypeptides were separated and transferred to PVDF membranes. The membranes were blocked and then treated overnight with rabbit anti-Col2a1 (1:1000, Abcam, UK), rabbit anti-MMP13 (1:1000, Cell Signaling Technology), rabbit anti-ACAN (1:1000, Abcam, UK), rabbit anti-COMP (1:1000, Abcam, UK), rabbit anti-ADAMTS4 (1:1000, Abcam, UK), and mouse anti-GAPDH (1:1000, Beyotime) antibodies, respectively. After being washed with TBST three times, the membranes were incubated using the corresponding HRP-conjugated secondary antibodies (1:1000, Beyotime) with blocking solution at room temperature for 2 h. Finally, the expression levels of protein were measured by enhanced chemiluminescence kit (Millipore). The protein bands were normalized by GAPDH and quantified as the ratio of the optical density.
Induction of OA
All animal experiments were in accordance to the Animal Research Committee regulations of Nanjing first hospital, Nanjing medical university. OA was induced in 8-week-old male C57BL/6 mice by surgical destabilization of the medial meniscus (DMM) of the right knee. Specifically, the DMM surgery was performed by surgical sectioning of the medial meniscotibial ligament and the sham operation was performed by incision of the cutaneous and muscular planes at baseline.
Intra‐articular injection
In the animal section 1 in Fig. 2, all mice underwent DMM surgery or sham surgery were randomly divided into three groups: (1) PBS group (control); (2) ExosScAT group; (3) ExosIPFP group; (4) ExosScAT-NC group; (5) ExosScAT-99b-3p group. The mice were given intra-articular injections of 10μl PBS or 10μl Exos (5ug) twice a week for 4 weeks with a 30‐gauge needle and a micro syringe (Hamilton). Afterward the mice were sacrificed, and cartilage tissue was dissected for histological assessment (H&E, Safranin O/fast green and toluidine blue staining) and immunofluorescent staining (ADAMTS4, ACAN, COMP, Col2a1).
In the animal section 2 in Fig. 6, all mice underwent DMM surgery were randomly divided into five groups: (1) PBS group (Control); (2) ExosScAT-99b-3p group; (3) HMPs group (4) HMPs@ExosScAT group; (5) HMPs@ExosScAT-99b-3p group. The mice were injected with 20μl PBS, 20μl Exo@ScAT-99b-3p (20ug), 30μl HMPs, 30μl HMPs@Exos (containing 20ug Exos), 30μl HMPs@ExosScAT-99b-3p (containing 20ug Exos) at the first day. After 4 weeks, the mice were sacrificed, and cartilage tissue was dissected for histological assessment (H&E, Safranin O/fast green and toluidine blue staining) and immunofluorescent staining (ADAMTS4, ACAN, COMP, Col2a1).