Inhibition of chondrocyte apoptosis in a rat model of osteoarthritis by exosomes derived from miR‑140‑5p‑overexpressing human dental pulp stem cells

  • Authors:
    • Tianji Lin
    • Nan Wu
    • Linhong Wang
    • Ruipeng Zhang
    • Ruolang Pan
    • Yun-Fang Chen
  • View Affiliations

  • Published online on: December 30, 2020     https://doi.org/10.3892/ijmm.2020.4840
  • Article Number: 7
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Osteoarthritis (OA) is a common joint disorder, and the restoration of the impaired cartilage remains a main concern for researchers and clinicians. MicroRNAs (miRNAs or miRs) play crucial roles in the pathogenesis of OA. The present study examined the therapeutic efficacy of exosomal‑miR‑140‑5p for the treatment of OA using dental pulp stem cells (DPSCs). The findings indicated that the exosomal burden of miR‑140‑5p was substantially increased following the transfection of DPSCs with miR‑140‑5p mimic. The administration of DPSC‑derived exosomes promoted chondrocyte‑related mRNA expression, including aggrecan, Col2α1 and Sox9, in interleukin (IL)‑1β‑treated human chondrocytes. This effect was substantially enhanced by miR‑140‑5p‑enriched exosomes. The results further revealed that miR‑140‑5p‑enriched exosomes induced a more significant reduction in IL‑1β‑induced chondrocyte apoptosis than the DPSC‑derived exosomes. Mechanistically, it was found that miR‑140‑enriched DPSC‑derived exosomes exerted anti‑apoptotic effects, probably by regulating the expression levels of apoptosis‑related proteins. Furthermore, multiple administrations of miR‑140‑5p‑enriched exosomes substantially improved knee joint conditions in a rat model of OA. Collectively, the data of the present study suggest that exosomes derived from genetically modified DPSCs may prove to be a potential strategy for the treatment of OA.

Introduction

As determined in the mid-eighteenth century, articular cartilage, once destroyed, cannot be repaired due to the minimal regeneration capacity of articular cartilage. Small defects or injuries may cause severe pain and joint disability owing to secondary osteoarthritis (OA) (1-4). Numerous surgical techniques have been developed over the past 2 centuries, which may help to partly alleviate symptoms; however, these cannot regenerate tissue similar to native articular cartilage (5-7). One strategy is to repair tissue with properties identical to native cartilage, which integrates with the tissue to encircle the lesion. An alternative approach would be to improve the self-regeneration of articular cartilage using cells and/or other biomaterials. For this strategy, autologous mesenchymal stromal/stem cells (MSCs) have become an excellent choice. MSCs are plastic-adherent fibroblast-like cells that can be isolated from an array of mesenchymal tissues, including bone marrow, adipose tissues, umbilical cord, placenta and dental pulp (8-12). They can also differentiate into adipocytes, osteoblasts, myocytes and chondrocytes both in vivo and in vitro (13-17).

In recent years, accumulating evidence has suggested that MSCs can migrate to injured sites and affect the local microenvironment by secreting different bioactive factors, including interleukin (IL)-6, IL-10, hepatocyte growth factor, transforming growth factor-β, matrix metalloproteinases and tissue inhibitors of metalloproteinases (18-24). Moreover, the paracrine effects of MSCs are mediated by the secretion of extracellular vesicles (EVs) (13,25,26). Exosomes are a subtype of EVs, approximately 30-100 nm in diameter, containing a particular set of protein families from intracellular compartments (27,28). MSC-derived exosomes have been investigated in different disease models and have exhibited therapeutic potential in managing stroke, Parkinson's disease and OA (29-33). MicroRNAs (miRNAs or miRs), including miR-222, miR-140 and miR-381, regulate chondrogenesis and cartilage degeneration (34-36). Previous studies have demonstrated that miR-140 plays essential roles in regulating cartilage homeostasis and development by enhancing the expression of Sox9 and aggrecan (ACAN) (37-39). Therefore, the role of miR-140 in the repair of cartilage injury in a model of OA is worthy of investigation. In the present study, miR-140-5p in was overexpressed in dental pulp stem cells (DPSCs) and an improved version of exosomes was produced from these cells, which promoted the recovery of articular cartilage injuries (Fig. S1).

Materials and methods

Human DPSC isolation and culture

Human DPSCs were provided by S-Evans Biosciences. The Ethics Committee of S-Evans Biosciences approved the collection of the human samples (no. 2020-01). Briefly, regular human third molars were extracted from adult donors (age, 18-28 years) during orthodontic treatment or due to impaction after obtaining written informed consent. Subsequently, the pulp tissue was lightly harvested and then digested in a collagenase type I (2 mg/ml) and dispase (4 mg/ml) solution for 1 h at 37°C. After passing through a 70-µm strainer (Falcon, BD Biosciences), the cell suspension was seeded into tissue culture plates at a density of 1×105/well with the α modification of Eagle's medium (Gibco; Thermo Fisher Scientific, Inc.), supplemented with 15% fetal bovine serum (FBS, Gibco; Thermo Fisher Scientific, Inc.), 2 mM l-glutamine, 100 units/ml penicillin and 100 µg/ml streptomycin. The plates were then incubated at 37°C in 5% CO2 using a humidified incubator and passaged by 0.25% trypsin-ethylenediaminetetraacetic acid (EDTA, Gibco; Thermo Fisher Scientific, Inc.) digestion at a 1:5 ratio.

Characterization of DPSCs and flow cytometric analysis

Osteogenic, adipogenic and chondrogenic inductions were conducted in vitro, as previously described, to evaluate the multilineage differentiation capacity of the DPSCs (40). To evaluate mineralization, the cultures were stained with Alizarin Red S according to the manufacturer' instructions (Cyagen Biosciences, Inc.). Briefly, cells were fixed with 4% paraformaldehyde (Beyotime Institute of Biotechnology) for 30 min and then stained with Alizarin Red S for 5 min at room temperature. Lipid droplets formed following adipogenesis induction were visualized using Oil Red O staining according to the manufacturer' instructions (Cyagen Biosciences, Inc.). Briefly, cells were fixed with 4% paraformaldehyde for 30 min followed by Oil Red O staining for 30 min at room temperature. Successful chondrogenic differentiation was tested with the use of Alcian blue staining according to the manufacturer' instructions (Cyagen Biosciences, Inc.). Briefly, cells were fixed with 4% paraformaldehyde for 30 min followed by Alcian blue staining for 30 min at room temperature.

The phenotype of the DPSCs was examined at the third passage by flow cytometry (BD FACSVerse) using antibodies, including anti-CD14 (BD Biosciences, cat. no. 555397, at a dilution of 1:5 and incubation at room temperature for 15 min), anti-CD34 (BD Biosciences, cat. no. 555822, at a dilution of 1:5 and incubation at room temperature for 15 min), anti-CD73 (BioLegend, cat. no. 344004, at a dilution of 1:20 and incubation at room temperature for 15 min), anti-CD105 (BioGems, cat. no. 17111-60, at a dilution of 1:20 and incubation at room temperature for 15 min), anti-HLA-DR (BD Biosciences, Cat. no. 555811, at a dilution of 1:5 and incubation at room temperature for 15 min) and anti-CD90 (BioLegend, cat. no. 328110, at a dilution of 1:5 and incubation at room temperature for 15 min) according to the manufacturer' instructions. The suitable isotype-matched antibody (PE; BD Biosciences, cat. no. 555749; FITC, BD Biosciences, cat. no. 555573, at dilution of 1:5 and incubation at room temperature for 15 min) was utilized as a negative control. The data were analyzed using BD FACSuite software (1.0.5.3841).

Isolation and identification of exosomes

Exosome isolation was performed using ultracentrifugation as previously described (41). In brief, culture supernatants of DPSCs were centrifuged at 3,000 × g for 30 min and then at 10,000 × g for a further 30 min at room temperature. Finally, exosomes were pelleted following centrifugation at 64,000 × g for 110 min at 4°C using an SW28 rotor (Beckman Coulter, Inc.) and washed once with 0.32 M sucrose. Nanosight 2000 analysis and transmission electron microscopy (FEI) were utilized for the identification of exosomes that were resuspended in phosphate-buffered saline.

Cell transfection

DPSCs at the third passage were transfected with miR-140-5p mimic (20 µg/ml) or negative control (20 µg/ml) (Guangzhou RiboBio Co., Ltd.). The Amaxa Nucleofection system was adopted according to the instructions of the manufacturer of the Human Mesenchymal Stem Cell Nucleofector TM kit (Lonza Group, Ltd.). Briefly, DNA and cells were mixed in the Amaxa cuvette (Lonza Group, Ltd.) and placed in the nucleofector device. Nucleofection was conducted using the U-23 program (Lonza Group, Ltd.), which was optimized and performed as recommended by the manufacturer. Subsequently, the cuvette was rinsed with culture medium and the cells were transfer into the dish for further culture.

RNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted using the Total Exosome RNA and Protein Isolation kit (Invitrogen; Thermo Fisher Scientific, Inc., cat. no. 4478545) according to the manufacturer's protocol, and the amount of RNA isolated was quantified using a NanoDrop spectrophotometer (NANO-100; Hangzhou Allsheng Instruments Co.,Ltd.). RT reactions were performed using 100 ng total RNA with the TaqMan miRNA RT kit (Applied Biosystems; Thermo Fisher Scientific, Inc., cat. no. 4366596) according to the manufacturer's instructions. qPCR reactions were conducted on a 7900 System (Applied Biosystems; Thermo Fisher Scientific, Inc.) using the Taq-Man miR-140 probe and gene-specific primers (Thermo Fisher Scientific, Inc., Assay ID 001187). Homo sapiens snRNA U6 qPCR Primer (Thermo Fisher Scientific, Inc., Assay ID 001973) were used as an internal control. qPCR was performed under the following conditions: 10 min at 95°C, followed by 40 cycles of 95°C for 10 sec, 20 sec at 60°C and 10 sec at 72°C.

Analysis of relative gene expression in cartilage cells

Cartilage cells subjected to IL-1β (10 ng/ml; R&D Systems, Inc.) and DPSC-derived exosome (5×108 particles/ml) treatment for 48 h were collected and total RNA was isolated using TRIzol reagent (Invitrogen, cat. no. 15596026) and reversed transcribed into first-strand cDNA using reverse transcriptase (Takara Bio, Inc.; cat. no. H2640A). RT-qPCR was performed using a SYBR-Green Master Mix (Takara Bio, Inc.; cat. no. RR820A) under the following conditions: 95°C for 4 min, 40 cycles of 95°C for 30 sec, 58°C for 30 sec, 72°C for 30 sec, followed by melting curve analysis of 95°C for 30 sec, 72°C for 30 sec, and 95°C for 30 sec. Relative gene expression was calculated using the 2−ΔΔCq (42) method and the data were normalized against GAPDH. The primers used are listed in Table I. Each PCR trial was performed in triplicate and repeated at least 3 times.

Table I

Primers used for reverse-transcription polymerase chain reaction.

Table I

Primers used for reverse-transcription polymerase chain reaction.

GenesForwardReverse
Aggrecan TGAGCGGCAGCACTTTGAC TGAGTACAGGAGGCTTGAGG
Col2a1 TCAGGAATTTGGTGTGGACATA CCGGACTGTGAGGTTAGGATAG
Sox9 ATGAAGATGACCGACGAGCA CAGTCGTAGCCTTTGAGCAC
GAPDH GGCACAGTCAAGGCTGAGAATG ATGGTGGTGAAGACGCCAGTA
Animal experiments

Specific pathogen-free (SPF) male Sprague-Dawley (SD) rats (approximately 8 weeks old, weighing 200±20 g) were procured from the Shanghai SLAC Laboratory Animal Co., Ltd. [Certificate no. SCXK (Shanghai) 2017-0005] and maintained in the SPF laboratory of a local animal facility. The experimental protocol was approved by the local Medical Animal Experiment Ethics Committee of Zhejiang Provincial People's Hospital (no. 2019-034).

A total of 24 SD rats were randomly divided into the blank, OA model, exosome treatment and improved-exosome treatment groups, with 6 animals per group. Subsequently, 50 µl of 25 mg/ml iodoacetic acid (Sigma-Aldrich; Merck KGaA) was administered to the double knee joints to induce OA. The same amount of saline was administered into the knee joint cavities of the rats in the blank group. After 1 week, the exosome treatment group and improved-exosome treatment group were injected with 50 µl of exosomes (5×1010 particles/ml, isolated from the culture supernatants of DPSCs as described above) and miR-140-enriched exosomes (5×1010 particles/ml, isolated from the culture supernatants of miR140-transfacted DPSCs as described above), respectively, in the knee joint cavity of both hind limbs once a week; a total of 4 such injections were administered. The rats in the blank group and OA model group were administered the same amount of saline. The rats were sacrificed by an overdose of pentobarbital (150 mg/kg) intraperitoneally at 1 week after the end of treatment in accordance with a previous study (43).

Gross morphological analysis of the knee joints

At 1 week after the end of treatment, 3 rats were randomly selected from each group and anesthetized with 3% pentobarbital sodium (60 mg/kg) by intraperitoneal injection. An X-ray examination of the knee joint structure was then performed, and the grade of knee joint degradation of digital radiography films (PerkinElmer, Inc., CLS136341/F) was scored using a modified Kellgren-Lawrence scoring system for rats as previously described (44,45).

Histopathological examination of the knee joints

The rats were sacrificed, and their knee joints were isolated and fixed in 10% neutral-buffered formalin (Sangon Biotech) for 3 days and then decalcified for 2 weeks in buffered 12.5% EDTA (as previously described with some modifications) (46,47) and formalin solution. The tissues were then embedded in paraffin and were sliced along the longitudinal axis of the lower extremity at a thickness of 5 µm. They were subsequently stained with Safranin O-Fast Green (Sangon Biotech) and hematoxylin and eosin (H&E; Sangon Biotech). For Safranin O-Fast Green staining, sections were deparaffinized and stained with Safranin O for 5 min followed by Fast Green staining for 5 min at room temperature. For H&E staining, sections were deparaffinized and stained with hematoxylin staining solution for 10 min and then counterstained in eosin staining solution for 1 min at room temperature. The OARSI score was used to evaluate histological changes as previously described (48).

Primary isolation and culture of human chondrocytes

Normal human cartilage tissues were obtained from donation with written informed consent, and the protocol was approved by the Ethics Committee of Zhejiang Provincial People's Hospital in Hangzhou, China (2018 KY 012). Samples were obtained aseptically and digested using a standard protocol. Briefly, the tissues were cut into <1 mm3 sections; they were then transferred to a culture flask, mixed with 5-fold volumes of 0.25% trypsin, and placed in a 37°C, 5% CO2 incubator for 30 min. The trypsin digestion solution was then discarded and 5-fold volumes of 0.05% type II collagenase (Sigma-Aldrich; Merck KGaA) were added to the tissue fragments. Subsequently, cells were collected every 4 h until the tissue block was digested. These chondrocytes were then cultured in DMEM supplemented with 20% FBS in a 37°C, 5% CO2 incubator.

Effects of DPSC-derived exosomes on IL-1β-treated chondrocytes in vitro

Normal human chondrocytes were treated with IL-1β to induce the inflammatory, in vitro model of OA as previously described with some modifications (49). The cells were divided into 4 different groups as follows: i) Normal culture; ii) IL-1β (10 ng/ml, R&D Systems); iii) IL-1β (10 ng/ml) + DPSC-derived exosomes (5×108 particles/ml); and iv) IL-1β (10 ng/ml) + miR-140 enriched exosomes (5×108 particles/ml). Cells were collected for use in western blot analysis and apoptosis analysis at different time points.

Western blot analysis

Western blot analysis was performed as previously described (50). Briefly, exosomes or chondrocytes were added to cold lysis buffer (Beyotime Institute of Biotechnology, cat. no. P0013B) and incubated for 30 min on ice. The lysates were then centrifuged at 13,000 × g for 10 min at 4°C. Subsequently, the supernatants were collected and quantified using the BCA protein assay kit (Beyotime Institute of Biotechnology, cat. no. P0011), and samples (20 µg) were separated on 10% polyacrylamide gels and transferred to Hybond-P membranes. 5% BSA (Beyotime Institute of Biotechnology, cat. no. ST025) was used for blocking at room temperature for 45 min prior to incubation with antibodies. The following antibodies were used: Rabbit anti-human CD63 (ProteinTech Group, Inc., cat. no. 25682-1-AP, at a dilution of 1:500), CD9 (ProteinTech Group, Inc., cat. no. 20597-1-AP at a dilution of 1:500) and Bcl-2 (ProteinTech Group, Inc., cat. no. 12789-1-AP at a dilution of 1:1,000), as well as rabbit anti-human Bax (Beyotime Institute of Biotechnology, cat. no. AF0057, at a dilution of 1:500) and Bad (Beyotime Institute of Biotechnology, cat. no. AF1009, at a dilution of 1:500). The primary antibodies were incubated at room temperature for 45 min. GAPDH (Beyotime Institute of Biotechnology, cat. no. AF1186, at a dilution of 1:2,000) served as the loading control. The blots were then incubated with HRP-conjugated goat anti-rabbit secondary antibody (Beyotime Institute of Biotechnology, cat. no. A0208, at a dilution of 1:1,000) for 45 min at room temperature and then processed for analysis using an ECL chemiluminescence kit (Beyotime Institute of Biotechnology). ImageJ (National Institutes of Health, v1.8.0) was further employed to analyze the grayscale values obtained by western blotting.

Apoptosis analysis

Apoptosis was determined using the Annexin V-propidium iodide (PI) binding assay as previously reported (51). The cells were harvested and labeled with Annexin V-FITC and PI for 15 min using a FITC Annexin V apoptosis detection kit I (BD Pharmingen). Apoptosis was evaluated using flow cytometry (Navios, Beckman Coulter, Inc.) within 30 min of staining, and the data were analyzed using FlowJo software v11.0. The extent of apoptosis was quantified as the percentage of Annexin V+ cells. All experiments were performed in triplicate.

Statistical analysis

Data are presented as the means ± standard deviations of the results of at least 3 independent studies. Student's t-tests were utilized to identify differences among groups, as suitable. One-way ANOVA with the Bonferroni test was performed for multiple group comparisons. A P-value <0.05 was considered to indicate a statistical significant difference.

Results

Isolation and characterization of DPSCs

DPSCs were isolated from the pulp tissue of extracted human third molar teeth without caries, periodontal diseases, or infections. These cells are closely associated with MSCs found in the stromal compartment of various tissues, including the bone marrow. As shown in Fig. 1A, the cells exhibited a typical spindle-shaped morphology. DPSCs followed the criteria that defined multipotent MSCs, as recommended by the International Society for Cellular Therapy guidelines in 2006 (52). Following culture in a specific differentiation medium for 2-3 weeks, the isolated DPSCs successfully differentiated into mesenchymal derivatives, including osteoblasts (identified by Alizarin Red labeling), adipocytes (identified by Oil Red O labeling) and chondrocytes (identified by Alcian blue) (Fig. 1A). Furthermore, as revealed by flow cytometric assay, these cells were positively stained for the common MSC-associated markers, CD73, CD90 and CD105, and did not exhibit an expression of the hematopoietic markers, CD14, CD34 and HLA-DR (Fig. 1B).

Isolation and identification of exosomes from dental pulp stem cell lines

To examine the effects of miR-140-5p-transfected DPSCs on articular cartilage injuries, miR-140-5p mimic or negative control oligonucleotide were transfected into third-passage DPSCs via electroporation. The cells were collected 48 h later for total RNA extraction. RT-qPCR revealed that the cells transfected with the mimic exhibited a 10-fold increase in the miR-140-5p level compared with the negative control cells (Fig. 2A). Exosomes were collected from both types of DPSCs by centrifugation. Nanosight analysis indicated that the diameter of the majority of the DPSC-derived exosomes was approximately 134±29 nm (Fig. 2B). As shown in Fig. 2C, western blot analysis revealed that the DPSC-derived exosomes expressed exosomal markers, including CD9 and CD63. No significant differences in the expression levels of CD9 and CD63 between exosomes from the DPSCs with and without miR-140-5p transfection were observed (P>0.05). Finally, the results of RT-qPCR revealed a 4-fold increase in the miR-140-5p levels in exosomes derived from the miR-140-5p-overexpressing DPSCs than those from the control exosomes.

Effect of DPSC-derived exosomes on mRNA levels in chondrocytes

Subsequently, normal human chondrocytes were treated with IL-1β (10 ng/ml) with or without DPSC-derived exosomes. As shown in Fig. 3, IL-1β stimulation substantially decreased the chondrocyte ACAN, Col2α1 and Sox9 mRNA levels. However, the use of DPSC-derived exosomes elevated the corresponding mRNA levels, particularly the miR-140-5p-enriched exosomes.

DPSC-derived exosomes inhibit the IL-1β-induced apoptosis of chondrocytes

Chondrocytes from normal humans were treated with IL-1β (10 ng/ml) with or without DPSC-derived exosomes. The cells were trypsinized for apoptosis analysis at various time points. As shown in Fig. 4A and B, when exposed to IL-1β for 48 h, the DPSC-derived exosome groups (DPSC-derived exosomes and miR-140-5p-enriched exosomes) exhibited a decreased apoptosis compared with the control group (16.29±0.1% and 12.32±0.2% vs. 21.91±0.3% Annexin V+ cells at 24 h; 24.91±0.1% and 18.14± 0.1% vs. 34.58±0.5% Annexin V+ cells at 48 h; all P<0.01). The miR-140-5p-enriched exosome group exhibited a more significant reduction in cell apoptosis than the DPSC-derived exosome group (P<0.01) (Fig. 4A and B). Likewise, western blot analysis indicated a significant inhibition of the expression of apoptosis-related proteins in chondrocytes treated with miR-140-5p-enriched exosomes (Fig. 4C and D).

Influence of exosome treatments on knee joint X-ray scores in a rat model of OA

Subsequently, the rat model of OA was treated with the 2 above-mentioned types of DPSC-derived exosomes and then assessed using X-rays to determine the effects of the treatments (Fig. 5). In comparison with the sham-operated group, the OA model group exhibited evident stenosis, with visible osteophytes and osteosclerosis. This was in line with previous findings (53,54), indicating the generation of the rat model of OA was successfully established. Both DPSC-derived exosomes reduced the grade and severity of knee joint damage in rats with OA, as determined by X-ray score, improved the joint cavity structure, and reduced the formation of osteophytes, articular surface irregularity and osteosclerosis. Moreover, these effects were substantially enhanced following miR-140-5p-enriched exosome treatment.

Effect of exosome treatments on the pathology of the knee joint in rats with OA

To further assess the effects of exosome treatment on the knee condition in rats with OA, the histological sections of rat cartilage in all 4 groups were examined. As observed following Safranin O-Fast Green and H&E staining (Figs. 6A and S2A), the knee joint cartilage was uneven in the rat model OA group, and the thickness of the cartilage was reduced with visible apoptotic bodies. Furthermore, the lower edge of the cartilage was uneven, and pathological changes, including inflammatory cell infiltration, could be observed. The multiple administration of DPSC-derived exosomes improved the pathology of the rat knee joints to varying degrees, and the miR-140-5p-enriched exosomes substantially reduced the total pathological OARSI grading score (Figs. 6B and S2B).

Discussion

OA is a chronic degenerative process that is a common cause of disability in middle-aged and older adults. Its common clinical phenotypes include progressive cartilage deterioration, subchondral bone remodeling, loss of joint space, marginal osteophytosis, and loss of joint function, and it affects >80% of individuals aged of ≥55 years (55). The majority of patients with OA are treated with a combination of non-pharmacological (such as physiotherapy) and pharmacological treatments to reduce pain and inflammation. Although some patients exhibit temporary relief, the efficacy of these interventions is not consistent, and studies assessing the efficacy of these interventions are still being conducted. In recent years, it has been proposed that MSCs improve chondrocyte regeneration and maintain articular cartilage in patients with OA (56).

MSCs are multipotent stromal cells that can differentiate into various cell types, including osteoblasts, adipocytes and chondrocytes (13-17). Apart from their ability to differentiate, the immune regulating function of MSC has received more attention in the past decades. Indeed, MSCs are used to treat various clinical conditions, particularly autoimmune diseases, including Crohn's disease, multiple sclerosis, lupus, COPD, Parkinson's and OA (32,57-60). However, these cells do not directly cure these conditions; the efficacy of MSC-based therapeutic strategies is ascribed more to paracrine secretion (61). MSCs influence the local microenvironment by secreting different bioactive factors. EVs are critical mediators of cell-to-cell communication in these situations. MSC-derived exosomes play a crucial role in regulating cell migration, proliferation, differentiation, and matrix synthesis. Moreover, MSC-derived exosomes have been shown to shuttle many bioactive components (proteins, lipids, mRNAs, miRNAs, lncRNAs, circRNAs and DNA) (31,62). Therefore, MSC exosomes may represent a new therapeutic alternative for OA. DPSCs are type of MSCs derived from dental pulp, which have become one of the best cells for periodontal tissue engineering owing to their excellent growth and differentiation capacity in ex vivo cultures (63,64). Reportedly, cultured DPSCs have a pale, round, or oval central nucleus with multiple nucleoli, showing active DNA transcription, and RNA synthesis. Numerous cytoplasmic vacuoles also show exuberant cellular secretory vesicles (65).

Consistent with previously published data, the current study proposed that miR-140 plays a role in chondrogenesis. miR-140 has been reported to play essential roles in regulating cartilage homeostasis and development by enhancing the expression of SOX9 and ACAN (37-39). In addition, miR-140-5p can regulate the proliferation and differentiation of DPSC and inhibit the differentiation of DPSCs into odontoblastic cells (66,67). In the present study, miR-140-5p was first overexpressed in DPSCs and the increased burden of miR-140-5p in exosomes isolated from DPSC culture supernatant was validated. The data indicated that DPSC-derived exosomes promoted the expression of chondrocyte-related mRNAs, including AGAN, Col2α1 and Sox9, in cultured normal human chondrocytes. Sox9 is the master regulator of a transcription factor for chondrogenesis. In particular, it has been shown to activate Col11A1, Col2A1 and ACAN, all of which play pivotal functions during chondrogenesis. Col11A1 has been proven to be essential in cartilage architecture during chondrogenesis by modulating chondrocyte behavior (68). ACAN can improve chondrogenesis by delaying hypertrophic chondrocyte maturation (69). Additionally, the effect of DPSC-derived exosomes on the expression of chondrocyte-related mRNAs was substantially enhanced by miR-140-5p enriched exosomes. The findings from Annexin V-PI flow cytometry and western blot analysis suggested that the miR-140-5p-enriched exosomes significantly inhibited the IL-1β-induced apoptosis of primary cultured human chondrocytes. Furthermore, the effects of DPSC-derived exosomes were explored in a rat model of OA, and the results were consistent with the in vitro findings.

In conclusion, the findings presented herein demonstrate that the overexpression of exosomal-miR-140-5p substantially attenuates OA both in vitro and in vivo. Although previous stem cell-related studies (70,71) have also indicated a protective effect of stem cell-derived exosomes in models of OA, the present study determined that exosomes from miR140-overexpressing DPSCs relieved symptoms of OA by inhibiting the apoptosis of chondrocytes and promoting cartilage repair. Hence, the present study provides a more efficient, safer and stem cell-independent strategy for the treatment of OA. To the best of our knowledge, this is the first study to adopt genetically modified DPSCs to produce miRNA-140-enriched exosomes for the treatment of OA. Nonetheless, further studies are required to clarify the specific function of exosomal-miR-140-5p in models of OA prior to the clinical application of exosome-based therapy in OA.

Supplementary Data

Funding

The present study was supported by the Social Development Project of Public Welfare Technology Application in Zhejiang Province (grant no. LGF18H060007), and the Key Technologies R&D Program of Zhejiang Province (grant no. 2019C03041).

Availability of data and materials

All data generated or analyzed in the current study are included in this published article or relevant supplementary material, or are available from the corresponding author upon reasonable request.

Authors' contributions

TL participated in the study design. NW performed the experiments. LW and RZ analyzed the data. YFC contributed to the study design and data acquisition, and provided valuable advice and drafted the manuscript. RP supervised the study, performed the experiments and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The Ethics Committee of S-Evans Biosciences approved the collection of human samples (no. 2020-01). The Ethics Committee of Zhejiang Provincial People's Hospital approved the experiments using human cartilage (2018 KY 012). All patients provided written informed consent. The experimental protocol was approved by the local Medical Animal Experiment Ethics Committee of Zhejiang Provincial People's Hospital (no. 2019-034).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that there are no competing interests. The company 'S-Evans Biosciences', with which RP is affiliated, had no role in the study design or outcomes.

Acknowledgments

Not applicable.

References

1 

Bhosale AM and Richardson JB: Articular cartilage: Structure, injuries and review of management. Br Med Bull. 87:77–95. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Buckwalter JA, Anderson DD, Brown TD, Tochigi Y and Martin JA: The roles of mechanical stresses in the pathogenesis of osteoarthritis: Implications for treatment of joint injuries. Cartilage. 4:286–294. 2013. View Article : Google Scholar

3 

Dieppe PA and Lohmander LS: Pathogenesis and management of pain in osteoarthritis. Lancet. 365:965–973. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Van Osch GJ, Brittberg M, Dennis JE, Bastiaansen-Jenniskens YM, Erben RG, Konttinen YT and Luyten FP: Cartilage repair: Past and future-lessons for regenerative medicine. J Cell Mol Med. 13:792–810. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Sgaglione NA, Miniaci A, Gillogly SD and Carter TR: Update on advanced surgical techniques in the treatment of traumatic focal articular cartilage lesions in the knee. Arthroscopy. 18(Suppl 1): S9–S32. 2002. View Article : Google Scholar

6 

Temenoff JS and Mikos AG: Review: Tissue engineering for regeneration of articular cartilage. Biomaterials. 21:431–440. 2000. View Article : Google Scholar : PubMed/NCBI

7 

Simon TM and Jackson DW: Articular cartilage: Injury pathways and treatment options. Sports Med Arthrosc Rev. 26:31–39. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Phinney DG and Prockop DJ: Concise review: Mesenchymal stem/multipotent stromal cells: The state of transdifferentiation and modes of tissue repair-current views. Stem Cells. 25:2896–2902. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Park D, Spencer JA, Koh BI, Kobayashi T, Fujisaki J, Clemens TL, Lin CP, Kronenberg HM and Scadden DT: Endogenous bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and regeneration. Cell Stem Cell. 10:259–272. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P and Hedrick MH: Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 13:4279–4295. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Qiao C, Xu W, Zhu W, Hu J, Qian H, Yin Q, Jiang R, Yan Y, Mao F, Yang H, et al: Human mesenchymal stem cells isolated from the umbilical cord. Cell Biol Int. 32:8–15. 2008. View Article : Google Scholar

12 

Valtieri M and Sorrentino A: The mesenchymal stromal cell contribution to homeostasis. J Cell Physiol. 217:296–300. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Romanov YA, Darevskaya AN, Merzlikina NV and Buravkova LB: Mesenchymal stem cells from human bone marrow and adipose tissue: Isolation, characterization, and differentiation potentialities. Bull Exp Biol Med. 140:138–143. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Timper K, Seboek D, Eberhardt M, Linscheid P, Christ-Crain M, Keller U, Müller B and Zulewski H: Human adipose tissue-derived mesenchymal stem cells differentiate into insulin, somatostatin, and glucagon expressing cells. Biochem Biophys Res Commun. 341:1135–1140. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Sonomoto K, Yamaoka K, Oshita K, Fukuyo S, Zhang X, Nakano K, Okada Y and Tanaka Y: Interleukin-1β induces differentiation of human mesenchymal stem cells into osteoblasts via the Wnt-5a/receptor tyrosine kinase-like orphan receptor 2 pathway. Arthritis Rheum. 64:3355–3363. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Liu ZJ, Zhuge Y and Velazquez OC: Trafficking and differentiation of mesenchymal stem cells. J Cell Biochem. 106:984–991. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Yu DA, Han J and Kim BS: Stimulation of chondrogenic differentiation of mesenchymal stem cells. Int J Stem Cells. 5:16–22. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Caplan AI: MSCs: The sentinel and safe-guards of injury. J Cell Physiol. 231:1413–1416. 2016. View Article : Google Scholar

19 

Anton K, Banerjee D and Glod J: Macrophage-associated mesenchymal stem cells assume an activated, migratory, pro-inflammatory phenotype with increased IL-6 and CXCL10 secretion. PLoS One. 7:e350362012. View Article : Google Scholar : PubMed/NCBI

20 

Di Gh, Liu Y, Lu Y, Liu J, Wu C and Duan HF: IL-6 secreted from senescent mesenchymal stem cells promotes proliferation and migration of breast cancer cells. PLoS One. 9:e1135722014. View Article : Google Scholar : PubMed/NCBI

21 

Qu X, Liu X, Cheng K, Yang R and Zhao RC: Mesenchymal stem cells inhibit Th17 cell differentiation by IL-10 secretion. Exp Hematol. 40:761–770. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Noh MY, Lim SM, Oh KW, Cho KA, Park J, Kim KS, Lee SJ, Kwon MS and Kim SH: Mesenchymal stem cells modulate the functional properties of microglia via TGF-beta secretion. Stem Cells Transl Med. 5:1538–1549. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Mias C, Lairez O, Trouche E, Roncalli J, Calise D, Seguelas MH, Ordener C, Piercecchi-Marti MD, Auge N, Salvayre AN, et al: Mesenchymal stem cells promote matrix metalloproteinase secretion by cardiac fibroblasts and reduce cardiac ventricular fibrosis after myocardial infarction. Stem Cells. 27:2734–2743. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Lozito TP and Tuan RS: Mesenchymal stem cells inhibit both endogenous and exogenous MMPs via secreted TIMPs. J Cell Physiol. 226:385–396. 2011. View Article : Google Scholar

25 

Rani S, Ryan AE, Griffin MD and Ritter T: Mesenchymal stem cell-derived extracellular vesicles: Toward cell-free therapeutic applications. Mol Ther. 23:812–823. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Lo Sicco C, Reverberi D, Balbi C, Ulivi V, Principi E, Pascucci L, Becherini P, Bosco MC, Varesio L, Franzin C, et al: Mesenchymal stem cell-derived extracellular vesicles as mediators of anti-inflammatory effects: Endorsement of macrophage polarization. Stem Cells Transl Med. 6:1018–1028. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Raposo G and Stoorvogel W: Extracellular vesicles: Exosomes, microvesicles, and friends. J Cell Biol. 200:373–383. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Théry C, Zitvogel L and Amigorena S: Exosomes: Composition, biogenesis and function. Nat Rev Immunol. 2:569–579. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Bang OY and Kim EH: Mesenchymal stem cell-derived extracellular vesicle therapy for stroke: Challenges and progress. Front Neurol. 10:2112019. View Article : Google Scholar : PubMed/NCBI

30 

Zhang ZG, Buller B and Chopp M: Exosomes-beyond stem cells for restorative therapy in stroke and neurological injury. Nat Rev Neurol. 15:193–203. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Yu B, Zhang X and Li X: Exosomes derived from mesenchymal stem cells. Int J Mol Sci. 15:4142–4157. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Cosenza S, Ruiz M, Toupet K, Jorgensen C and Noël D: Mesenchymal stem cells derived exosomes and microparticles protect cartilage and bone from degradation in osteoarthritis. Sci Rep. 7:162142017. View Article : Google Scholar : PubMed/NCBI

33 

Liu Y, Lin L, Zou R, Wen C, Wang Z and Lin F: MSC-derived exosomes promote proliferation and inhibit apoptosis of chondrocytes via lncRNA-KLF3-AS1/miR-206/GIT1 axis in osteoarthritis. Cell Cycle. 17:2411–2422. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Dong S, Yang B, Guo H and Kang F: MicroRNAs regulate osteogenesis and chondrogenesis. Biochem Biophys Res Commun. 418:587–591. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Shang J, Liu H and Zhou Y: Roles of micro RNA s in prenatal chondrogenesis, postnatal chondrogenesis and cartilage-related diseases. J Cell Mol Med. 17:1515–1524. 2013. View Article : Google Scholar

36 

Liu H, Sun Q, Wan C, Li L, Zhang L and Chen Z: MicroRNA-338-3p regulates osteogenic differentiation of mouse bone marrow stromal stem cells by targeting Runx2 and Fgfr2. J Cell Physiol. 229:1494–1502. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Miyaki S, Sato T, Inoue A, Otsuki S, Ito Y, Yokoyama S, Kato Y, Takemoto F, Nakasa T, Yamashita S, et al: MicroRNA-140 plays dual roles in both cartilage development and homeostasis. Genes Dev. 24:1173–1185. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Buechli ME, Lamarre J and Koch TG: MicroRNA-140 expression during chondrogenic differentiation of equine cord blood-derived mesenchymal stromal cells. Stem Cells Dev. 22:1288–1296. 2013. View Article : Google Scholar

39 

Karlsen TA, Jakobsen RB, Mikkelsen TS and Brinchmann JE: microRNA-140 targets RALA and regulates chondrogenic differentiation of human mesenchymal stem cells by translational enhancement of SOX9 and ACAN. Stem Cells Dev. 23:290–304. 2014. View Article : Google Scholar

40 

Hilkens P, Gervois P, Fanton Y, Vanormelingen J, Martens W, Struys T, Politis C, Lambrichts I and Bronckaers A: Effect of isolation methodology on stem cell properties and multilineage differentiation potential of human dental pulp stem cells. Cell Tissue Res. 353:65–78. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Greening DW, Xu R, Ji H, Tauro BJ and Simpson RJ: A protocol for exosome isolation and characterization: Evaluation of ultracentrifugation, density-gradient separation, and immunoaffinity capture methods. Methods Mol Biol. 1295:179–209. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

43 

Yan L, Zhou L, Xie D, Du W, Chen F, Yuan Q, Tong P, Shan L and Efferth T: Chondroprotective effects of platelet lysate towards monoiodoacetate-induced arthritis by suppression of TNF-α-induced activation of NF-KB pathway in chondrocytes. Aging (Albany NY). 11:2797–2811. 2019. View Article : Google Scholar

44 

Jay GD, Elsaid KA, Kelly KA, Anderson SC, Zhang L, Teeple E, Waller K and Fleming BC: Prevention of cartilage degeneration and gait asymmetry by lubricin tribosupplementation in the rat following anterior cruciate ligament transection. Arthritis Rheum. 64:1162–1171. 2012. View Article : Google Scholar

45 

Teeple E, Elsaid KA, Jay GD, Zhang L, Badger GJ, Akelman M, Bliss TF and Fleming BC: Effects of supplemental intra-articular lubricin and hyaluronic acid on the progression of posttraumatic arthritis in the anterior cruciate ligament-deficient rat knee. Am J Sports Med. 39:164–172. 2011. View Article : Google Scholar

46 

Gao X, Jiang S, Du Z, Ke A, Liang Q and Li X: KLF2 protects against osteoarthritis by repressing oxidative response through activation of Nrf2/ARE signaling in vitro and in vivo. Oxid Med Cell Longev. 2019:85646812019. View Article : Google Scholar :

47 

Inomata K, Tsuji K, Onuma H, Hoshino T, Udo M, Akiyama M, Nakagawa Y, Katagiri H, Miyatake K, Sekiya I, et al: Time course analyses of structural changes in the infrapatellar fat pad and synovial membrane during inflammation-induced persistent pain development in rat knee joint. BMC Musculoskelet Disord. 20:82019. View Article : Google Scholar : PubMed/NCBI

48 

Pritzker KP, Gay S, Jimenez SA, Ostergaard K, Pelletier JP, Revell PA, Salter D and van den Berg WB: Osteoarthritis cartilage histopathology: Grading and staging. Osteoarthritis Cartilage. 14:13–29. 2006. View Article : Google Scholar

49 

Castro Martins M, Peffers MJ, Lee K and Rubio-Martinez LM: Effects of stanozolol on normal and IL-1β-stimulated equine chondrocytes in vitro. BMC Vet Res. 14:1032018. View Article : Google Scholar

50 

Fang QX, Zheng XC and Zhao HJ: L1CAM is involved in lymph node metastasis via ERK1/2 signaling in colorectal cancer. Am J Transl Res. 12:837–846. 2020.PubMed/NCBI

51 

Crowley LC, Marfell BJ, Scott AP and Waterhouse NJ: Quantitation of apoptosis and necrosis by Annexin V binding, propidium iodide uptake, and flow cytometry. Cold Spring Harb Protoc. 2016:2016.

52 

Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop DJ and Horwitz E: Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy. 8:315–317. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Kellgren JH and Lawrence JS: Radiological assessment of osteo-arthrosis. Ann Rheum Dis. 16:494–502. 1957. View Article : Google Scholar : PubMed/NCBI

54 

Kohn MD, Sassoon AA and Fernando ND: Classifications in brief: Kellgren-lawrence classification of osteoarthritis. Clin Orthop Relat Res. 474:1886–1893. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Loeser RF, Goldring SR, Scanzello CR and Goldring MB: Osteoarthritis: A disease of the joint as an organ. Arthritis Rheum. 64:1697–1707. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Qi Y, Feng G and Yan W: Mesenchymal stem cell-based treatment for cartilage defects in osteoarthritis. Mol Biol Rep. 39:5683–5689. 2012. View Article : Google Scholar

57 

Voswinkel J, Francois S, Simon JM, Benderitter M, Gorin NC, Mohty M, Fouillard L and Chapel A: Use of mesenchymal stem cells (MSC) in chronic inflammatory fistulizing and fibrotic diseases: A comprehensive review. Clin Rev Allergy Immunol. 45:180–192. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Bai L, Lennon DP, Caplan AI, DeChant A, Hecker J, Kranso J, Zaremba A and Miller RH: Hepatocyte growth factor mediates mesenchymal stem cell-induced recovery in multiple sclerosis models. Nat Neurosci. 15:862–870. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Liu S, Liu D, Chen C, Hamamura K, Moshaverinia A, Yang R, Liu Y, Jin Y and Shi S: MSC transplantation improves osteopenia via epigenetic regulation of notch signaling in lupus. Cell Metab. 22:606–618. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Gu W, Song L, Li XM, Wang D, Guo XJ and Xu WG: Mesenchymal stem cells alleviate airway inflammation and emphysema in COPD through down-regulation of cyclooxygenase-2 via p38 and ERK MAPK pathways. Sci Rep. 5:1–11. 2015.

61 

Lee JW, Fang X, Krasnodembskaya A, Howard JP and Matthay MA: Concise review: Mesenchymal stem cells for acute lung injury: Role of paracrine soluble factors. Stem Cells. 29:913–919. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Deng H, Sun C, Sun Y, Li H, Yang L, Wu D, Gao Q and Jiang X: Lipid, protein, and microRNA composition within mesenchymal stem cell-derived exosomes. Cell Reprogram. 20:178–186. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Ashri NY, Ajlan SA and Aldahmash AM: Dental pulp stem cells: Biology and use for periodontal tissue engineering. Saudi Med J. 36:1391–1399. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Graziano A, d'Aquino R, Laino G and Papaccio G: Dental pulp stem cells: A promising tool for bone regeneration. Stem Cell Rev. 4:21–26. 2008. View Article : Google Scholar : PubMed/NCBI

65 

Merckx G, Hosseinkhani B, Kuypers S, Vanspringel L, Irobi J, Michiels L, Lambrichts I and Bronckaers A: Extracellular vesicles from human dental pulp stem cells as proangiogenic strategy in tooth regeneration. J Extracellular Vesicles. 7:134. 2018.

66 

Lu X, Chen X, Xing J, Lian M, Huang D, Lu Y, Feng G and Feng X: miR-140-5p regulates the odontoblastic differentiation of dental pulp stem cells via the Wnt1/β-catenin signaling pathway. Stem Cell Res Ther. 10:2262019. View Article : Google Scholar

67 

Sun DG, Xin BC, Wu D, Zhou L, Wu HB, Gong W and Lv J: miR-140-5p-mediated regulation of the proliferation and differentiation of human dental pulp stem cells occurs through the lipopolysaccharide/toll-like receptor 4 signaling pathway. Eur J Oral Sci. 125:419–425. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Li A, Wei Y, Hung C and Vunjak-Novakovic G: Chondrogenic properties of collagen type XI, a component of cartilage extracellular matrix. Biomaterials. 173:47–57. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Caron MMJ, Janssen MPF, Peeters L, Haudenschild DR, Cremers A, Surtel DAM, van Rhijn LW, Emans PJ and Welting TJM: Aggrecan and COMP improve periosteal chondrogenesis by delaying chondrocyte hypertrophic maturation. Front Bioeng Biotechnol. 8:10362020. View Article : Google Scholar : PubMed/NCBI

70 

Mianehsaz E, Mirzaei HR, Mahjoubin-Tehran M, Rezaee A, Sahebnasagh R, Pourhanifeh MH, Mirzaei H and Hamblin MR: Mesenchymal stem cell-derived exosomes: A new therapeutic approach to osteoarthritis? Stem Cell Res Ther. 10:3402019. View Article : Google Scholar : PubMed/NCBI

71 

Zhu Y and Wang Y, Zhao B, Niu X, Hu B, Li Q, Zhang J, Ding J, Chen Y and Wang Y: Comparison of exosomes secreted by induced pluripotent stem cell-derived mesenchymal stem cells and synovial membrane-derived mesenchymal stem cells for the treatment of osteoarthritis. Stem Cell Res Ther. 8:642017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2021
Volume 47 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin T, Wu N, Wang L, Zhang R, Pan R and Chen Y: Inhibition of chondrocyte apoptosis in a rat model of osteoarthritis by exosomes derived from miR‑140‑5p‑overexpressing human dental pulp stem cells. Int J Mol Med 47: 7, 2021
APA
Lin, T., Wu, N., Wang, L., Zhang, R., Pan, R., & Chen, Y. (2021). Inhibition of chondrocyte apoptosis in a rat model of osteoarthritis by exosomes derived from miR‑140‑5p‑overexpressing human dental pulp stem cells. International Journal of Molecular Medicine, 47, 7. https://doi.org/10.3892/ijmm.2020.4840
MLA
Lin, T., Wu, N., Wang, L., Zhang, R., Pan, R., Chen, Y."Inhibition of chondrocyte apoptosis in a rat model of osteoarthritis by exosomes derived from miR‑140‑5p‑overexpressing human dental pulp stem cells". International Journal of Molecular Medicine 47.3 (2021): 7.
Chicago
Lin, T., Wu, N., Wang, L., Zhang, R., Pan, R., Chen, Y."Inhibition of chondrocyte apoptosis in a rat model of osteoarthritis by exosomes derived from miR‑140‑5p‑overexpressing human dental pulp stem cells". International Journal of Molecular Medicine 47, no. 3 (2021): 7. https://doi.org/10.3892/ijmm.2020.4840