2.1. Isolation and culture of tonsil-derived mesenchymal stem cells
Tonsil-derived mesenchymal stem cells (ToMSCs) were isolated following a slightly modified version of a previously established protocol. Briefly, discarded tonsil fragments were obtained from children who had undergone tonsillectomy at the Department of Otolaryngology of Bundang CHA Hospital (Seongnam-si, Korea). Written informed consent was obtained from the parents of the children who provided tonsil tissue. The study protocol was approved by the institutional review board of Bundang CHA Hospital (approval number: 2021-03-016). Isolated tonsillar tissue was washed twice with 1× phosphate-buffered saline (PBS) (Welgene, Gyeongsan-si, Korea). The tissue was then cut into small pieces and digested in RPMI 1640 medium (Invitrogen, Waltham, MA, USA) containing 210 U/mL collagenase type 1 (Invitrogen) and 10 µg/mL DNase I (Sigma-Aldrich, St. Louis, MO, USA) for 30 minutes at 37°C. The digested tissue was filtered through a wire mesh, and the collected cells were washed twice with RPMI 1640/20% fetal bovine serum (FBS) (Gibco, Waltham, USA), followed by an additional wash with RPMI 1640/10% FBS. Mononuclear cells were isolated from the cell suspension using Ficoll-Paque (GE Healthcare, Little Chalfont, UK) density gradient centrifugation, and 1×108 cells were seeded in a T-125 culture flask containing Dulbecco modified Eagle medium F12 (Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% FBS, 100 µg/mL streptomycin (Invitrogen), and 100 U/mL penicillin (Invitrogen). After 48 hours, unattached cells were discarded, and the adherent cells were expanded with fresh medium.
2.2. Flow cytometry analysis
To characterize the phenotype of ToMSCs, flow cytometry analysis was performed. A minimum of 30,000 cells were suspended in 100 µL of 1×PBS containing 2% FBS and stained with fluorochrome-conjugated antibodies. The antibodies used were APC-αCD44, PE-αCD73, PE-αCD90, APC-αCD105 (all from Miltenyi Biotec, Teterow, Germany), PE-αHLA-DR (BD Biosciences, Franklin Lakes, NJ, USA), and FITC-αHLA-ABC (Thermo Fisher Scientific).
2.3. Plasmid construction
2.3.1. pAAVS1-puro-CAG-SDF1α-6H
To obtain the SDF1α-6H (6His) DNA, polymerase chain reaction (PCR) was conducted using pBABE puro SDF-1 alpha (Addgene, Watertown, MA, USA) as a template, which produced a DNA fragment containing Sal I and Mlu I sites at the 5'- and 3'-ends, respectively. The PCR product was then cloned into the pTA vector (Real Biotech Corp, Banqiao City, Taiwan). Subsequently, the SDF1α-6H DNA fragment was isolated from the pTA-SDF1α-6H plasmid through Sal I/Mlu I double digestion. This fragment was inserted into a Sal I/Mlu I-digested pAAVS1-puro-CAG-EGFP plasmid (Addgene). The resulting plasmid, pAAVS1-puro-CAG-SDF1α, was prepared on a medium scale (midiprep) and confirmed by sequencing.
2.3.2. pAAVS1-puro-CAG-TGFβ1-6H
The pAAVS1-puro-CAG-TGFβ1-6H plasmid was generated using the same approach as the pAAVS1-puro-CAG-SDF1α-6H plasmid. In this instance, TGFβ1-6H complementary DNA (cDNA) was acquired through PCR, utilizing TGFB1_pLX307 (Addgene) as a template.
2.3.3. pAAVS1-puro-CAG-mCherry
The pAAVS1-puro-CAG-mCherry plasmid, used as a negative control, was generated using the same protocol as the pAAVS1-puro-CAG-SDF1α-6H and pAAVS1-puro-CAG-TGFβ1-6H plasmids. For this cloning process, mCherry DNA was PCR-amplified from pCDNA3-FlipGFP-Casp3-T2A-mCherry (Addgene).
2.3.4. pAAVS1-puro-Tetoff-TGFβ1-IGF1-BMP7-CAG-tTA-Advanced
The Tetoff promoter and tTA-Advanced gene were obtained from pTRE-TIGHT (Takara Bio, Shiga, Japan) and pAAV-Tetoffbidir-Alb-luc (Addgene), respectively. TGFβ1, IGF1, and BMP7 cDNAs were derived from TGFB1_pLX307 (Addgene), pT7T3Pac-IGF1 (Korea Human Gene Bank, Daejeon, Korea), and pTetON-BMP2/7 (Addgene), respectively. The three genes were fused using P2A and T2A sequences and expressed as a monocistronic gene cassette under the Tetoff promoter. Following this, all DNA components were subcloned into the pAAVS1-puro-CAG plasmid, resulting in the creation of pAAVS1-puro-Tetoff-TGFβ1-IGF1-BMP7-CAG-tTA-Advanced.
2.4. CRISPR/Cas9-mediated knock-in gene editing
The adeno-associated virus integration site 1 (AAVS1) locus, a safe harbor site, was utilized as the integration site for the knock-in gene cassettes using the CRISPR/Cas9 system [31, 32]. Transfections were performed on ToMSCs (passage 2) with 6.25 µg of Cas9 protein, 12 µg of sgRNA, and 1 µg of donor DNA using the Neon Transfection System (Invitrogen) with parameters set at 990 V, 40 ms, and 2 pulses. The donor DNAs used in this study included pAAVS1-Puro-CAG-TGFβ1-6H, pAAVS1-puro-CAG-SDF1α-6H, and pAAVS1-puro-Tetoff-TGFβ1-IGF1-BMP7-CAG-tTA-Advanced. The resulting stable ToMSC lines were designated as ToMSC-TGFβ1-6H, ToMSC-SDF1α-6H, and ToMSC-Tetoff-TGFβ1-IGF1-BMP7, respectively.
Following electroporation, cells were treated with 0.7 µg/mL puromycin (Sigma-Aldrich) 48 hours later to select for the knock-in colonies. The chosen clones were then cultured in the presence of 0.14 µg/mL puromycin.
2.5. Immunofluorescent detection of transgene expression
Cells were washed with ice-cold 1× PBS and fixed using 4% paraformaldehyde/1×PBS (Biosolution, Seoul, Korea). The fixed cells were washed four times with 1× PBS and subsequently permeabilized with 0.1% Triton-X (Sigma-Aldrich) for 10 minutes. Subsequently, the cells were blocked with 5% normal goat serum (Thermo Fisher Scientific) and incubated overnight at 4°C with primary antibodies, under constant shaking. The primary antibodies used included anti-His tag antibody (1:2,000) (ABM, Richmond, BC, Canada) for ToMSC-SDF1α-6H and ToMSC-TGFβ1-6H, as well as anti-BMP7 antibody (1:500) (Thermo Fisher Scientific) for ToMSC-Tetoff-TGFβ1-IGF1-BMP7. After incubation with the primary antibodies, cells were washed three times with 1×PBS and incubated with a goat anti-rabbit Alexa Fluor 594 IgG antibody at room temperature for 1 hour (1:200) (Invitrogen). Following four washes with 1×PBS, the cells were counterstained with 4',6-diamidino-2-phenylindole and dihydrochloride (DAPI) (1:2,000) (Sigma-Aldrich) and observed under a microscope.
2.6. Western blots
Total protein was extracted from cells using RIPA buffer (Thermo Fisher Scientific), and 30 µg of protein per lane was separated via 12% SDS-PAGE. The proteins were then transferred onto nitrocellulose membranes (Sigma-Aldrich). Subsequently, the membranes were blocked in 5% non-fat milk for 1 hour at room temperature with constant shaking. This was followed by incubation with primary antibodies, including anti-His tag antibody (1:2,000) (abm), anti-BMP7 antibody (1:500) (Thermo Fisher Scientific), and anti-β-actin (1:1,000) (Santa Cruz Biotechnology, Dallas, TX, USA) at 4°C overnight. The membranes were then washed three times with PBST (1×PBS, 0.2% (v/v) Tween-20) and incubated with the corresponding secondary antibodies (goat anti-rabbit or goat anti-mouse IgG H&L (HRP) (LI-COR, Lincoln, NE, USA) at room temperature for 2 hours. Afterward, the membranes were washed another three times with 1×PBST, then finally analyzed using an Odyssey CLx Imager (LI-COR).
2.7. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR)
Total RNAs were extracted from samples of ToMSC-Tetoff-TGFβ1-IGF1-BMP7, ToMSC-TGFβ1, ToMSC-SDF1α, and ToMSC-mCherry using an RNA purification kit made by MACHEREY-NAGEL (MACHEREY-NAGEL GmbH & Co., Düren, Germany), and cDNAs were synthesized using 100 ng of each RNA using a cDNA synthesis kit (TOYOBO, Osaka, Japan). Quantitative RT-PCR was performed using a power SYBR green master mix (Thermo Fisher Scientific).
2.8. Enzyme-linked immunosorbent assay
The level of TGFβ1 protein secreted into the medium from cultured cells was measured using a human TGFβ1 DuoSet enzyme-linked immunosorbent assay (ELISA) kit (R&D Systems, Minneapolis, MN, USA) according to the manufacturer’s instructions.
2.9. Animal surgical procedures
Thirty-five female Sprague-Dawley rats, 10 weeks old and weighing between 230 and 250 g, were purchased from Orient Bio. Inc. (Seongnam, Korea). The rats were acclimatized for 1 week on a 12/12 h light/dark cycle, with a temperature of 22°C ± 1°C and a relative humidity of 50%±1%. During this period, the animals were given free access to food and water.
The animal study was approved by the Institutional Animal Care and Use Committee of CHA University (IACUC220190) and was conducted in accordance with the committee’s guidelines.
Prior to surgery, the peritoneal site of the rat was sterilized with 70% alcohol, followed by the administration of general anesthesia via intraperitoneal injection of Zoletil (50 mg/kg; Virbac Laboratories, Carros, France) and Rompun (10 mg/kg; Bayer, Seoul, Korea). The tail skin was then sterilized with 70% alcohol and treated with povidone-iodine. A dorsal longitudinal skin incision was made along the tail to expose the coccygeal disc. The coccygeal discs (Co6/7, Co7/8) were punctured with a 21-G sterile spinal needle, from the dorsal to the ventral side of the tail, perpendicular to the tail skin. The needle was subsequently rotated 360º twice and held for 30 seconds [33–35]. The needle was bent at a length of 5 mm, limiting the puncture depth to that distance. The skin was then sutured with Nylon 4 − 0 and sterilized with povidone-iodine. Throughout the experiment, the rats were kept on healing pads to prevent hypothermia. Postoperatively, an analgesic (ketoprofen, 1 mg/kg; SCD Pharm. Co. Ltd., Seoul, Korea) and an antibiotic (Cefazolin, 15 mg/kg, CKD Pharmaceuticals, Seoul, Korea) were administered at appropriate doses for 3 days. All rats were housed individually under a 12/12 h light/dark cycle at a controlled temperature of 22ºC ± 1ºC and a relative humidity of 50%±1%.
2.10. Experimental design in a rat disc degeneration model
Degeneration of the coccygeal disc (Co6/7 and Co7/8) was induced in 35 rats by puncturing the center of the disc using a 21-G spinal needle. The rats were then randomly divided into four groups: (1) ToMSC-mCherry (n = 7), (2) ToMSC-TGFβ1 (n = 9), (3) ToMSC-SDF1α (n = 9), and (4) ToMSC-Tetoff-TGFβ1-IGF1-BMP7 (n = 10). Two weeks after the induction of disc degeneration, ToMSC-mCherry, ToMSC-TGFβ1 (1 µL of 2 × 104 cells + 1 µL hydrogel), ToMSC-SDF1α (1 µL of 2 × 104 cells + 1 µL hydrogel), and ToMSC-Tetoff-TGFβ1-IGF1-BMP7 (1 µL of 2 × 104 cells + 1 µL hydrogel) plasmids were implanted into the center of the punctured Co7/8 disc in each group using a Hamilton syringe with a 30-G needle. Six weeks post-implantation, the rats were euthanized via carbon dioxide asphyxiation, and the coccygeal discs were removed for radiologic and histologic analysis. The healthy IVDs at Co5/6 in each group were used as the normal control group, and the degeneration-induced Co6/7 discs in each group were used as the injury-control group.
2.11. Quantitative behavioral nociception assay
The von Frey test for behavioral nociception was performed 1 day prior to implantation and then on days 7, 14, 21, 28, 35, and 42 post-implantation [36]. To minimize the potential misinterpretation of exploratory activities as a positive response, the rats were individually placed in small cages with mesh floors and lids to acclimate to the environment for 20 minutes. Subsequently, a 2-g filament was applied with sufficient force to the ventral surface of the tail for a maximum of 6 seconds. A response was considered positive if the rat exhibited nocifensive behaviors, such as flinching, licking, withdrawing, or shaking the base of the tail, immediately or within 6 seconds. In the event of a positive response, the rat was tested with a lighter filament. However, if no response was detected, testing continued with a heavier filament until five responses were recorded in five attempts for two consecutive filaments. The von Frey analysis was conducted by two independent observers who were blinded to the treatment conditions of the specimens.
2.12. Magnetic resonance imaging
Six weeks post-implantation, changes in disc structure and the degree of coccygeal disc degeneration were evaluated using a 9.4-T magnetic resonance imaging (MRI) spectrometer (Bruker BioSpec, Billerica, MA, USA). T2-weighted imaging was performed for the coronal plane with the following parameters: time to repetition (TR) of 5,000 ms, time to echo (TE) of 30 ms, 150 horizontal × 150 vertical matrix, 15 horizontal × 15 vertical fields of view, and 0.5-mm slices with 0-mm spacing between each slice. For the sagittal plane, imaging was conducted with parameters including TR of 5,000 ms, TE of 30 ms, 150 horizontal × 150 vertical matrix; 15 horizontal × 15 vertical field of view, and 0.5-mm slices with 0-mm spacing between each slice. The MRI index (calculated as the area of the NP multiplied by the average signal intensity) was used as an outcome measure to evaluate the extent of rat coccygeal disc degeneration [37, 38]. The high signal intensity area in the mid-sagittal plane of the T2-weighted image, which delineates the NP, was considered the region of interest. The signal intensity of this region was measured using ImageJ (version 1.50b, National Institutes of Health, Bethesda, MD, USA; https://imagej.nih.gov/ij/) and compared to that of a normal rat coccygeal disc. The MRI index of each group was assessed by two independent observers who were blinded to the implantation in the rat coccygeal discs (n = 7 for group 1, n = 9 for group 2, n = 9 for group 3, and n = 10 for group 4).
2.13. Histological analysis
At 6 weeks post-implantation, the rats were euthanized using excessive carbon dioxide inhalation, and coccygeal disc tissues, along with the adjacent vertebral body, were preserved in 10% neutral buffered formalin for 1 week. Following this, the tissues underwent decalcification in Rapid Cal Immuno (BBC Biochemical, Mount Vernon, WA, USA) for 2 weeks. The disc tissues were then embedded in paraffin and sectioned to a thickness of 6 µm using a microtome (Leica, Wetzlar, Germany). Next, the sections were dewaxed, rehydrated, and stained with Safranin O (Sigma-Aldrich) to evaluate the distribution and quantity of proteoglycan. Finally, the sections were mounted using mounting media and scanned with a C-mount camera adapter (U-TVO.63XC; Olympus, Tokyo, Japan).
Histological scoring was performed using a comprehensive 16-point scale to assess the IVD based on Safranin O staining [38, 39]. This scale was divided into five subcategories, focusing on NP morphology, NP cellularity, AF morphology, endplate morphology, and the boundary between the NP and AF. Degenerative features in NP and AF morphology were given high importance, as were changes in notochordal cell morphology in degenerative rat IVDs; thus, these categories were weighted twice. Detailed observations included NP shape and total NP area; NP cell number and cellular morphology; appearance of the NP-AF border; lamellar organization and any tears or disruptions in the AF region; and disruptions, microfractures, osteophytes, or ossifications in the endplate. These observations were scored on a scale of 0 (non-degenerative) to 2 (severe degenerative changes), with total scores ranging from 0 (normal) to 16 (most severe). Two independent observers, blinded to the sample information, conducted the histological analysis of all samples.
Additionally, the acquired samples were dewaxed, rehydrated, and stained with hematoxylin and eosin (H&E) for the analysis of tissue morphology and the number of NP cells. The disc NP cell count and H&E-positive area were quantified using ImageJ.
2.14. Immunofluorescence and immunohistochemistry
Immunohistochemical analysis was conducted for aggrecan and type II collagen, along with immunofluorescence analysis for calcitonin gene receptor protein (CGRP), Tie2, Brachyury, matrix metalloproteinase-13 (MMP-13), human nuclei antibody, tumor necrosis factor-alpha (TNF-α), and interleukin-1-beta (IL-1β). For immunostaining, the sections were dewaxed, rehydrated, and stained with primary antibodies against aggrecan (1:1,000) (ab36861; Abcam, Cambridge, UK), type II collagen (1:100) (ab34712; Abcam), CGRP (1:200) (ab47027; Abcam), Tie2 (1:200) (NBP2-20636; Novus Biologicals, Littleton, CO, USA), Brachyury (1:200) (sc-166962; Santa Cruz Biotechnology), human nuclei antibody (1:200) (MAB1281; Sigma-Aldrich), TNF-α (1:200) (ab6671; Abcam), MMP-13 (1:200) (ab39012; Abcam), and IL-1β (1:200) (AF-501-NA; Novus Biologicals). After 24 hours of incubation, the sections were rinsed with 1×PBS containing 0.1% Tween 20 and subsequently incubated with the secondary antibody anti-Rb horseradish peroxidase (Roche Diagnostics Ltd., Basel, Switzerland), as well as Alexa Fluor 488- (1:400) (A11034; Invitrogen), Alexa Fluor 488- (1:400) (A11029; Invitrogen), Alexa Fluor 568- (1:400) (A10042; Invitrogen), and Alexa Fluor 647 (1:400) (A21469; Invitrogen)-conjugated secondary antibodies. Following additional washes, the sections were counterstained with 4',6-diamidino-2-phenylindole (DAPI) (1:1,000) (Abcam) for 10 minutes. The prepared sections were then examined using a fluorescence microscope (Zeiss 880, Oberkochen, Germany and Leica SP5). The percentages of the area positive for aggrecan and type II collagen, as well as the number of positive cells relative to DAPI for CGRP, Tie2, Brachyury, MMP-13, human nuclei antibody, TNF-α, and IL-1β, were quantified using ImageJ.
2.15. Statistical analysis
Statistical analysis was performed using GraphPad Prism (version 5.01; GraphPad Software, San Diego, CA, USA) and ImageJ. All data were represented as mean ± standard deviation. To determine statistical significance, one-way analysis of variance (ANOVA) with the Tukey post-test or two-way ANOVA was utilized. The threshold for statistical significance was established at p < 0.05.