Skip to main content
  • Research article
  • Open access
  • Published:

Adipose-derived mesenchymal stem cells and platelet-rich plasma synergistically ameliorate the surgical-induced osteoarthritis in Beagle dogs

Abstract

Background

The purpose of this study is to investigate the clinical effects of platelet-rich plasma (PRP) and adipose-derived mesenchymal stem cell (MSC) as the fundamental treatment of osteoarthritis (OA).

Methods

Twenty four Beagle dogs were used as cranial cruciate ligament transection models. The dogs were divided into four groups (nā€‰=ā€‰6) according to the intra-articular injection materials: the control group with phosphate-buffered saline (PBS), the PRP group with PRP, the MSC group with MSCs emerged in PBS, and the MSC and PRP co-treatment (MP) group with MSCs and PRP.

Results

Lameness score, focal compression strength, articular extracellular matrix (ECM) compositions, histopathology, and real-time PCR were used to evaluate the effects of PRP and MSCs on canine OA. In the order of MP, PRP, and MSC group, these all showed positive effects on the evaluated categories. The lameness scores were lower, and the focal compression strengths of the affected femoral articular surface cartilages were higher than those in the OA control group. Also, the inflammatory changes, when evaluated with Mankin scoring and histomorphologic examination, were significantly ameliorated with the treatment of PRP and/or MSCs. The glycosaminoglycan and collagen composition of extracellular matrix was more favorable in the test groups. The ECM-related genes significantly increased through the up-regulation, while the protein expressions of inflammatory cytokines were decreased through the inhibitory effects of PRP and MSCs on chondrocyte apoptosis and inflammatory cytokines.

Conclusions

Taken together, this study suggests that PRP and MSCs treatments have a beneficial effect on OA via the stimulation of ECM synthesis and chondrocyte proliferation and via the inhibition of inflammatory reaction.

Background

Osteoarthritis (OA) is the most common clinical syndrome of joint pain and dysfunction, accompanied by varying degrees of functional limitation and the reduced quality of life. Because it is mostly irreversible and progressive, OA joint consequently loses their cartilage layer [1].

The most ideal treatment of OA is focused on blocking the catabolic activity of cartilage and enhancing regeneration of normal cartilage. Until now, therapies for the OA commonly focused on the palliation of pain and discomfort, improvement on functional movement, and prevention of further degeneration. Thus, the primary approach in the clinical treatment of OA involves the extensive use of NSAIDs, analgesics, and hyaluronan which allows the brief symptomatic relief but provides no apparent disease-modifying effect [2ā€“4]. Therefore, there is a critical need for the development of the alternative agents that can fundamentally prevent the destruction of cartilages or stimulate its proper repair. In these aspects, various efforts have been tried to search for the effective cartilage-preserving methods with cell sources.

To sum up, the isolated chondrocyte expansion and implantation method is regarded as the fundamental solution. However, the main concern of the most cultured chondrocytes is losing the characteristics of producing hyaline-like cartilage [5, 6]. In a canine model, it has been reported that cultured autologous chondrocytes failed to return to the normal hyaline cartilage [7].

In human, the intra-articular injection of 1.0ā€‰Ć—ā€‰108 cells of adipose-derived mesenchymal stem cell (MSC) could improve the function, reduce the pain, and regenerate the hyaline-like cartilage [8]. MSCs are typically not observed in synovial fluid. However, MSCs appear in synovial fluid in the OA condition, and they are thought to play an important role in the regeneration of damaged tissue and have anti-inflammatory effect [9]. As the patient ages, the quality and quantity of MSCs decrease and there is a reduction of their ability to proliferate and differentiate. Also, the depletion of healthy MSCs is regarded as one of the reasons worsening OA [10]. For these reasons, it is believed that the direct administration of MSCs could promote the positive role of them by preventing their depletion and improving cartilage regeneration [11].

The platelet-rich plasma (PRP) is defined as the plasma with the platelet population of more than 1.0ā€‰Ć—ā€‰106 cells/Ī¼l, and it has typically four- to eightfold more platelets than the normal plasma [12]. PRP has various growth factors such as platelet-derived growth factor and transforming growth factor beta [13]. It has been well known that PRP has angiogenic, anti-inflammatory, and anti-catabolic effects [13, 14]. It has also been reported that transforming growth factor beta and fibroblast growth factor from PRP have an anabolic effect on cartilage metabolism [15]. These factors not only regulate the cell migration and proliferation but also enhance the wound healing and extracellular matrix (ECM) remodeling via the stimulation of angiogenesis [16, 17].

Based on the previous studies, it is hypothesized that the PRP could have a synergistic effect on the cartilage regeneration with the combination of MSCs. Therefore, the purpose of this study is to examine the effect of PRP and MSCs on the morphologic change and regeneration of articular cartilage in the inflammation process using canine OA model.

Methods

PRP preparation

Autologous PRP was prepared in each dog using double spin method. Fifty milliliters of fresh blood was collected with 7Ā ml of acid citrate dextrose formula A. Then, the blood was centrifuged at 1200Ā rpm for 10Ā min into three layers: plasma, buffy coat, and red blood cell. After the plasma and buffy coat were separated into a new tube, the mixture was centrifuged at 2500Ā rpm for 10Ā min. Discarding the supernatant, only the lower 20Ā % of the plasma was harvested. The collected plasma (PRP) was tested using complete blood cell count test (Cell-Dyne, Abbott Lab., USA) to make sure it had 1.0ā€‰Ć—ā€‰106 platelets/Ī¼l or more. All prepared PRP in this study were used within 6Ā h.

Adipose-derived mesenchymal stem cell isolation and culture

Approximately 15Ā g of fat tissue was aseptically collected from the flank of a dog. It was rinsed with phosphate-buffered saline (PBS) several times to remove any remaining anesthetic agent and blood. Rinsed fat tissue was then digested using 0.075Ā % collagenase type I (collagenase type 1A, Sigma-Aldrich, USA) in 37Ā Ā°C water-chamber for 2Ā h with shaking or inverting every 30Ā min. After adding the equal volumes of Dulbecco modified Eagle medium and 10Ā % fetal bovine serum, it was centrifuged at 1200Ā rpm for 10Ā min. The supernatant and digested lipids were discarded. The cell pellet was washed with PBS, and it was filtered through 100-Ī¼m nylon mesh. After centrifuging at the same condition, the cells were suspended into a 100ā€‰Ć—ā€‰20Ā mm cell culture dish with low-glucose Dulbecco modified Eagle medium and 10Ā % fetal bovine serum. After 24Ā h, non-adherent cells and debris were washed with PBS, and cell culture media was replaced twice per week. MSCs were collected and used between passage 1 and 2 in all the experiments of this paper. The flow cytometry analysis with established MSCs was performed, and obtained MSCs were negative for cluster of differentiation (CD) 34 and CD45 and strongly positive for CD29 and CD44Ā (Additional file 1: Fig.Ā S6).

The canine model of cranial cruciate ligament transection

The procedures were approved by Institutional Animal Care and Use Committees of Kyungpook National University. Twenty four physically healthy Beagle dogs were used in this experiment. The weights of dogs were 7.7ā€‰Ā±ā€‰1.1Ā kg (meanā€‰Ā±ā€‰standard deviation), and ages were between 2ā€“3Ā years old. Under the anesthetic state, the cranial cruciate ligament of a right hind limb was excised with a no. 11 scalpel blade. The connective tissues and skin were sutured with routine procedure. Analgesics (tramadol 8Ā mg/kg BID, subcutaneous) and antibiotics (enrofloxacin 5Ā mg/kg SID, subcutaneous) were administrated for 3Ā days after the surgery. After a week for soft tissue healing, each dog regularly walked for 10Ā min per day for 2Ā months. Then, treatment was given every week for 1Ā month. Another 2Ā months later, the dogs were sacrificed and stifle samples were collected.

MSC and PRP application

After the canine OA model, the subjects were treated every week for 1Ā month with an intra-articular injection with each material according to the groups: the control group with 1Ā ml of PBS, the PRP group with 1Ā ml of PRP, the MSC group with 1.0ā€‰Ć—ā€‰107 MSCs in 1Ā ml of PBS, and the MSC and PRP co-treatment (MP) group with 1.0ā€‰Ć—ā€‰107 MSCs in 1Ā ml of PRP. The contralateral stifle joint of the dogs in control group were used as a sham group in a histopathological examination, and no treatment material was given.

Evaluations

Lameness score

The lameness score was measured before the surgery and by every month after the surgery. All dogs had normal gait and no lameness before surgery. Previously described scoring system was used [18], and is as followed: 0, no detectable lameness; 1, minor lateral weight shift but no lameness at walk and trot; 2, no lameness at a walk but mild lameness at a trot; 3, mild lameness at a walk and significant lameness at trot; 4, non-weight bearing at a trot; and 5, non-weight bearing at walk and standing. As a blind test, three veterinarians assessed the grade of lameness.

The measurement of focal compressive strengths

After sacrifice and sampling, the ex vivo compression strengths of the femoral and tibial articular surfaces (0.2Ā mm) of each sample were detected with a computerized testing machine (SV-H1000, Japan Instrumentation System Co., Tokyo, Japan) as N (Newton). The measured points were central region of the medial femoral and tibial condyle.

Histological process

The articular cartilages of central region of the lateral femoral and tibial condyle were taken from knee joints of each group, and they were separately fixed in 10Ā % neutral buffered formalin (NBF), and then decalcified in decalcifying solution (24.4Ā % formic acid and 0.5Ā N sodium hydroxide) for 14Ā days (mixed decalcifying solution was exchanged once a day for 14Ā days). Each femoral and tibial articular surface cartilage was longitudinally trimmed, then embedded in paraffin, sectioned (3ā€“4Ā Ī¼m) using tungsten bladder equipped automated polycut microtome (Model RM2255, Leica, Wetzlar, Germany), and stained with Sirius red stain for cartilaginous tissues. In each prepared histological samples, the histological profiles were interpreted under a light microscope (Model Eclipse 80i, Nikkon, Tokyo, Japan) as blinds to group distribution when this analysis was made.

Analysis of ECM compositions

Some parts of the cartilage on the femoral and tibial articular surface taken from the knee joints of each group were separately lyophilized to obtain the dry weight. Then, the piece was digested and used for glycosaminoglycan (GAG) and collagen (COL) analyses [19, 20]. The concentration of GAG was determined through the di-methyl-methylene blue sulfated GAG assay using a UV/VIS spectrophotometer (Optizen Pop, Mecasys, Daejeon, Korea). Collagen content was determined by Erlichā€™s hydroxyproline assay [21]. The hydroxyproline content was converted to the collagen content using the following equation: (Ī¼g hydroxyprolineā€‰Ć—ā€‰dilution factor)/0.13ā€‰=ā€‰Ī¼g collagen, based on the fact that hydroxyproline representing approximately 13Ā % of the amino acid content of collagen in the human meniscus [22]. The concentrations of GAG and collagen were standardized to tissue dry weight and expressed as microgram per milligram to allow comparison among the experimental groups.

ECM-related chondrogenic gene mRNA expressions

The SOX9 and aggrecan messenger RNA (mRNA) expressions on the femoral and tibial articular surface cartilages were detected using real-time PCR. Briefly, RNA was extracted using Trizol reagent (Invitrogen, Carlsbad, CA, USA). The RNA concentrations and quality were determined by CFX96ā„¢ Real-Time System (Bio-Rad, Hercules, CA, USA). To remove contaminating DNA, samples were treated with recombinant DNase I (DNA-free; Ambion, Austin, TX, USA). RNA was reverse transcribed using the reagent High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, CA, USA) according to the manufacturerā€™s instructions. The cDNA strand was synthesized from the total RNA and then the mixture of primers and the cDNA products was amplified by PCR, and the conditions of PCR amplification were 58Ā Ā°C for 30Ā min, 94Ā Ā°C for 2Ā min, 35Ā cycles of 94Ā Ā°C for 15Ā s, 60Ā Ā°C for 30Ā s, 68Ā Ā°C for 1Ā min, and then 72Ā Ā°C for 5Ā min. Analysis was carried out using ABI Step One Plus Sequence Detection System (Applied Biosystems, Foster City, CA, USA), and their expression levels were calculated as relative to sham group. The expression of glyceraldehyde 3-phosphate dehydrogenase (G3PDH) mRNA was used as a control for tissue integrity in all of the samples. The sequences of the PCR oligonucleotide primers were as listed in TableĀ 1.

Table 1 Oligonucleotides for real-time PCR used in this study

BrdU uptake measurement

To assess the effects of MSC and PRP or their co-treatment (MP) on the proliferation of cells within the dog knee joints, proliferating cells were labeled by an intraperitoneal injection of 5-bromo-2ā€²-deoxyuridine (BrdU). Dogs were given intraperitoneal injections of BrdU (Sigma-Aldrich, St. Louise, MO, USA) 50Ā mg/kg, in a volume of 1Ā ml/kg and dissolved in saline, and the animals were sacrificed 72Ā h later. BrdU uptakes were detected with immunohistochemistry using an anti-BrdU antibody, as shown in histomorphometry sections.

Immunohistochemistry

Immunoreactivity for BrdU as cell proliferating marker was considered using purified primary antibody with avidin-biotin-peroxidase complex (ABC). Immunoreactivities in the prepared femoral and tibial surface cartilage tissues against caspase-3, cleaved poly(ADP-ribose) polymerase (PARP), tumor necrosis factor (TNF)-Ī±, cyclooxygenase (COX)-2, interleukin (IL)-1Ī², interferon (IFN)-Ī³, and inducible nitric oxide synthase (iNOS) were also additionally observed after treatment MSCs, PRP, or their combination. Briefly, endogenous peroxidase activity was blocked by incubated in methanol and 0.3Ā % H2O2 for 30Ā min, and non-specific binding of immunoglobulin was blocked with normal horse serum blocking solution for 1Ā h in humidity chamber after epitope retrievals by pretreatment of trypsin (Sigma-Aldrich, St. Louise, MO, USA) and 2Ā N HCl, on the prepared unstained sections. The primary antisera were treated for overnight at 4Ā Ā°C in humidity chamber and then incubated with biotinylated universal secondary antibody and ABC reagents for 1Ā h at room temperature in humidity chamber. Finally, sections were reacted with peroxidase substrate kit for 3Ā min at room temperature. All sections were rinse in 0.01Ā M PBS for three times, between each step. The primary antisera and detection kits for immunohistochemistry used in this study were described in Additional file 1: TableĀ S1.

Histomorphometry

To observe more detailed histopathological changes, the articular cartilage injuries stained with Safranin O staining were evaluated and recorded using the Mankin scoring systems referred by the other studies [23]. With this system, the higher the score, the higher the level of OA (semiquantative scores; maxā€‰=ā€‰12). The thicknesses of femur and tibia articular cartilages (micrometer per cartilage) were measured with the histomorphometrical analyses at prepared longitudinally trimmed samples, using a computer based automated image analyzer (iSolution FL ver 9.1, IMT i-solution Inc., Vancouver, Quebec, Canada). A total of six histological regions of femoral and tibial articular surface regions of each group were considered for further analysis. The cells occupied by over 20Ā % of immunoreactivities, the density, of each antibody for caspase-3, PARP, TNF-Ī±, COX-2, IL-1Ī², IFN-Ī³, and iNOS were regarded as positive, and the numbers of each immunoreactive cells were counted separately in each of the femoral and tibial articular surface regions as cells per square millimeter, under blinds condition.

Statistical analysis

The values were expressed as meanā€‰Ā±ā€‰standard deviation (SD). A multiple comparison tests were conducted for different groups. According to the result of variance homogeneity by the Levene test, one-way ANOVA test and the least-significant differences (LSD) multi-comparison test were used for parametric comparison, and Kruskal-Wallis H test was used for non-parametric comparisons, followed by Mann-Whitney (MW) U test with Bonferroni correction. Statistical analyses were conducted using SPSS for Windows (Release 14.0K, IBM SPSS Inc., USA).

Results

Although the lameness score in MP group was decreased compared to those in the other groups, there were no significant changes between groups. The lameness score was significantly decreased at 2Ā months and at 3Ā months after treatment in the PRP group and MP group when compared with the previous treatment, respectively (Fig.Ā 1).

Fig. 1
figure 1

The lameness score of the OA dogs. Control, treated with 1Ā ml of PBS; PRP, treated with 1Ā ml of PRP; MSC, treated with 1.0ā€‰Ć—ā€‰107 MSC in 1Ā ml of PBS; MP, treated with 1.0ā€‰Ć—ā€‰107 MSC cell in 1Ā ml of PRP. Asterisk indicates significantly different with day 0 within PRP group by MW test; Number sign significantly different with day 0 within MP group by MW test

The focal compressive strength of the femoral and tibial articular surface cartilages were significantly decreased in the control group as compared to that of the sham group. However, the focal compressive strength significantly increased by the treatment of all three test materials. In MP group, the focal compressive strength was higher than those in any other treated groups (Fig.Ā 2).

Fig. 2
figure 2

Focal compressive strengths on the femoral and tibial articular cartilages. Values are expressed as meanā€‰Ā±ā€‰SD of six dogs, N (Newton). a pā€‰<ā€‰0.05 as compared with sham control by LSD test; b pā€‰<ā€‰0.05 as compared with control by LSD test; c pā€‰<ā€‰0.05 as compared with MSC treated group by LSD test; d pā€‰<ā€‰0.01 and e pā€‰<ā€‰0.05 as compared with PRP treated group by LSD test

The Mankin scores of control group were significantly increased as compared with those of shame group. When compared with control group, it was found that the Mankin score significantly reduced with the treatment of all three test materials on the femoral and tibial articular cartilages. Especially, the Mankin score of MP treated dogs was lowest compared to those of MSC or PRP treated dogs (TableĀ 2).

Table 2 Mankin scores on the femur and tibia articular cartilages

The thickness of articular cartilages was higher in the MSC and PRP than control group. The more favorable effect on the articular surface was examined in MP group, compared to those of MSC and PRP groups (Fig.Ā 3).

Fig. 3
figure 3

Representative general histopathological images and the thicknesses of the femoral and tibial articular cartilages. Of sham (a, b), control (c, d), MSC (e, f), PRP (g, h), and MP (i, j). H&E and Safranin O stain. Scale barsā€‰=ā€‰90Ā Ī¼m. a pā€‰<ā€‰0.01 as compared with sham control by LSD test; b pā€‰<ā€‰0.01 and c pā€‰<ā€‰0.05 as compared with control by LSD test; d pā€‰<ā€‰0.01 as compared with MSC treated group by LSD test; e pā€‰<ā€‰0.01 and f pā€‰<ā€‰0.05 as compared with PRP treated group by LSD test

The contents of COL and GAG as the main component of ECM were significantly decreased in control group compared with those of sham group, but it was significantly increased in all treated groups compared with control group (Fig.Ā 4).

Fig. 4
figure 4

COL and GAG contents on the femoral articular cartilages. a pā€‰<ā€‰0.01 as compared with sham control by LSD test; b pā€‰<ā€‰0.01 and c pā€‰<ā€‰0.05 as compared with control by LSD test; d pā€‰<ā€‰0.01 as compared with MSC treated group by LSD test; e pā€‰<ā€‰0.01 and f pā€‰<ā€‰0.05 as compared with PRP treated group by LSD test

When the real-time PCR performed to determine the ECM-related genes, the expression of cartilage aggrecan and SOX9 was decreased as compared with those of sham group. However, those down-regulated gene expressions were significantly increased by the treatment of all three test materials, and it was most potent in MP group (Fig.Ā 5).

Fig. 5
figure 5

mRNA expressions of aggrecan and SOX9 on the femoral and tibial articular cartilages. Values are expressed as meanā€‰Ā±ā€‰SD of six dogs, relative to control/G3PDH mRNA. a pā€‰<ā€‰0.01 and b pā€‰<ā€‰0.05 as compared with sham control by LSD test; c pā€‰<ā€‰0.01 and d pā€‰<ā€‰0.05 as compared with control by LSD test; e pā€‰<ā€‰0.01 as compared with MSC treated group by LSD test; f pā€‰<ā€‰0.01 and g pā€‰<ā€‰0.05 as compared with PRP treated group by LSD test

The BrdU-positive cells in the femoral and tibial articular cartilages significantly decreased in control group comparing to that of sham group. The values were significantly increased in all treated groups compared with control group. The increase of cell proliferation on the cartilage was most significant in MP group than MSC or PRP group (Fig.Ā 6, TablesĀ 3 and 4).

Fig. 6
figure 6

Representative immunohistochemical findings of femoral and tibial articular cartilage (BrdU, TNF-Ī±, and PARP). Of sham (a, b), control (c, d), MSC (e, f), PRP (g, h), and MP (i, j). Scale barsā€‰=ā€‰90Ā Ī¼m

Table 3 Immunohistochemical analysis on the femur articular cartilages
Table 4 Immunohistochemical analysis on the tibia articular cartilages

Immunopositive cells to caspase-3 and PARP were significantly increased in the femoral and tibial cartilages of control group when compared with those of the sham group. However, these increased cells were significantly reduced by treatment of all three test materials, and the reduction was most significant in MP group (Fig.Ā 6, TablesĀ 3 and 4).

TNF-Ī±, COX-2, IL-1Ī², iNOS, and IFN-Ī³ staining cells were increased significantly in the femoral and tibial cartilages of control group compared with sham group. However, these increases of pro-inflammatory cytokines on the cartilages were diminished in treated groups (Fig.Ā 6 and Additional file 1: Fig. S1-S5, TablesĀ 3 and 4).

Discussion

Osteoarthritis (OA) is characterized by the loss of articular cartilage components with inflammation, eventually resulting in impaired joint function [24, 25]. For this reason, this study mainly focused on the evaluation of the clinical signs, the change of ECM component and articular cartilage, the gene expression related to chondrogenesis, and the articular pathologic processes such as inflammation and apoptosis with the treatment of PRP and/or MSC.

Previously, it has been reported that the intra-articular injection of PRP reduced the lameness score [26]. Similarly, there were a meaningful decrease of lameness score with the treatment of PRP and MSC mixed with PRP in this canine OA model. Based on this result, we anticipate that PRP would relieve pain and improve articular function in an arthritic condition.

It is well known that the structure of articular cartilage is altered by the disorganization of collagen network, decrease of proteoglycan contents, and disruption of the integrity of the ECM in the process of OA [27]. Therefore, we tested the compressive strength of the articular cartilage to observe whether PRP or MSCs has a protective effect on the articular damage by OA. As previous studies have reported, Mankin score is a good index of osteoarthritis [28] and it is highly associated with compressive strength of articular cartilage [27]. We evaluated Mankin score of the affected articular cartilage. In addition, the thickness of articular cartilage and the content of COL and GAG were measured to confirm the OA condition. As a result, there were decreases in Mankin score as well as increases in the thickness of cartilage and the content of COL and GAG in the PRP and/or MSC treatment. These results suggested that PRP and MSC may have protective and regenerative effects on the degenerative cartilage of OA.

Aggrecan is known as a core protein of cartilage-specific proteoglycan, and it was reported that the mRNA expression of aggrecan was down-regulated according to the severity of OA condition [29, 30]. The expression of SOX9, the essential transcription factor for the chondrogenic differentiation, was also inhibited by OA model [30]. In this study, the down-regulations of SOX-9 and aggrecan in the control group were increased by the MSC and PRP treatment. Based on these results, we can suppose that the increase of ECM-related factors such as aggrecan, SOX9, COL, and GAG may be closely associated with the recovery of damaged articular cartilage. From this aspect, it could be explained by the fact that PRP and/or MSC treatment was associated with the increase of focal compressive strengths and the decrease of Mankin score. These findings are compatible with a previous report indicated that the inhibition of SOX9 gene expression and the content of GAG might result in cartilage degeneration [30].

We next performed BrdU staining to examine whether the MSC and/or PRP treatment promoted the proliferation of chondrocytes. It was shown that BrdU-positive cells decreased in OA condition and increased with a treatment of MSC and/or PRP. We also performed immunostaining for the caspase-3 and PARP expression examine whether apoptotic change during OA could be affected by PRP and/or MSC treatment. Results showed that the number of caspase-3- and PARP-positive cells was increased in the OA condition and was reduced with a treatment of MSC and/or PARP. These results were consistent with the findings of previous study that caspase-3 led to PARP to cleavage and consequently caused the progression of OA with cell apoptosis and death [31]. Therefore, we propose that the proliferation of chondrocyte may be suppressed in the process of OA through apoptotic change of articular cartilage, and this may be partly prevented by MSC and/or PRP treatment.

We examined the protein expression of cytokines relative to inflammatory reaction in the cartilage tissue. It has been known that pro-inflammatory cytokines mediated the activation of various inflammatory pathways and played a role in the progression of OA [32]. In the present study, the numbers of TNF-Ī±-, COX-2-, IL-1Ī²-, IFN-Ī³-, and iNOS-positive cells were increased in the OA condition, and MSC and/or PRP treatment stimulated the down-regulated expression of inflammatory cytokines. This result is comparable to the previous studies that MSC increased the anti-inflammatory cytokines such as IL-10, and PRP also had anti-inflammatory effect via blockage of NF-ĪŗB cascade, which contributes to the up-regulation of pro-inflammatory cytokines [33]. It has been known that MSC played important role in the cartilage repair by direct differentiation to chondrocyte and paracrine effect [8, 34]. In addition, it also has been reported that PRP could promote the proliferation by its various growth factors rather than differentiation of MSCs [35]. Therefore, PRP might prevent the depletion of MSCs, enhance the effect of MSCs, and guide MSCs to properly differentiate into chondrocytes. Consistent with previous study, the results of this study suggested that each treated with either MSC or PRP could decrease death or apoptosis of chondrocytes and inhibit inflammatory response [31, 36] and that the co-treatment with MSC and PRP might have beneficial effects on the pathologic changes in the articular cartilage.

In summary, we have shown that MSC and/or PRP ameliorated the degeneration of articular cartilage in the surgically induced OA animal model. The proliferation of chondrocyte may be suppressed in the process of OA through apoptotic change of articular cartilage, and this may be partly prevented by MSC and/or PRP treatment. MSC and/or PRP treatment also stimulated the down-regulated expression of inflammatory cytokines such as TNF-Ī±-, COX-2-, IL-1Ī²-, IFN-Ī³-, and iNOS. It will be needed to elucidate if transplanted MSCs are engrafted and differentiated to chondrocyte in OA cartilage and how MSCs and PRP interact in intra-articular environment.

Conclusions

Taken together, this study shows that the combination of MSC and PRP has a beneficial and synergistic effect on OA via the ECM synthesis and chondrocyte proliferation and via the anti-inflammatory reaction. Therefore, the combination treatment of MSC and PRP may be very useful as an inflammatory regulator for the treatment of OA that exhibit irreversible articular degeneration.

Ethics approval and consent to participate

All procedures were approved (KNU2015-060) by the Institutional Animal Care and Use Committees of Kyungpook National University.

Abbreviations

BrdU:

5-bromo-2ā€²-deoxyuridine

CD:

cluster of differentiation

COX:

cyclooxygenase

ECM:

extracellular matrix

GAG:

glycosaminoglycan

IFN:

interferon

IL:

interleukin

iNOS:

inducible nitric oxide synthase

LSD:

least-significant differences

MSC:

mesenchymal stem cell

MW:

Mann-Whitney

OA:

osteoarthritis

PARP:

cleaved poly (ADP-ribose) polymerase

PBS:

phosphate-buffered saline

PRP:

platelet-rich plasma

TNF:

tumor necrosis factor

References

  1. Goldring MB, Goldring SR. Osteoarthritis. J Cell Physiol. 2007;213:626ā€“34.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  2. Lo GH, LaValley M, McAlindon T, Felson DT. Intra-articular hyaluronic acid in treatment of knee osteoarthritis: a meta-analysis. JAMA. 2003;290:3115ā€“21.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  3. Arrich J, Piribauer F, Mad P, Schmid D, Klaushofer K, Mullner M. Intra-articular hyaluronic acid for the treatment of osteoarthritis of the knee: systematic review and meta-analysis. CMAJ. 2005;172:1039ā€“43.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  4. Tamura T, Ohmori K. Rhein, an active metabolite of diacerein, suppresses the interleukin-1 alpha-induced proteoglycan degradation in cultured rabbit articular chondrocytes. Jpn J Pharmacol. 2001;85:101ā€“4.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  5. Lin L, Zhou C, Wei X, Hou Y, Zhao L, Fu X, et al. Articular cartilage repair using dedifferentiated articular chondrocytes and bone morphogenetic protein 4 in a rabbit model of articular cartilage defects. Arthritis Rheum. 2008;58:1067ā€“75.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  6. Marlovits S, Hombauer M, Truppe M, Vecsei V, Schlegel W. Changes in the ratio of type-I and type-II collagen expression during monolayer culture of human chondrocytes. J Bone Joint Surg (Br). 2004;86:286ā€“95.

    ArticleĀ  CASĀ  Google ScholarĀ 

  7. Nganvongpanit K, Pothacharoen P, Chaochird P, Klunklin K, Warrit K, Settakorn J, et al. Prospective evaluation of serum biomarker levels and cartilage repair by autologous chondrocyte transplantation and subchondral drilling in a canine model. Arthritis Res Ther. 2009;11:R78ā€“8.

  8. Jo CH, Lee YG, Shin WH, Kim H, Chai JW, Jeong EC, et al. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells. 2014;32:1254ā€“66.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  9. Sekiya I, Ojima M, Suzuki S, Yamaga M, Horie M, Koga H, et al. Human mesenchymal stem cells in synovial fluid increase in the knee with degenerated cartilage and osteoarthritis. J Orthop Res. 2011;30:943ā€“9.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  10. Murphy JM, Dixon K, Beck S, Fabian D, Feldman A, Barry F. Reduced chondrogenic and adipogenic activity of mesenchymal stem cells from patients with advanced osteoarthritis. Arthritis Rheum. 2002;46:704ā€“13.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  11. Barry F, Murphy M. Mesenchymal stem cells in joint disease and repair. Nat Rev Rheumatol. 2013;9:584ā€“94.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  12. Metcalf KB, Mandelbaum BR, McIlwraith CW. Application of platelet-rich plasma to disorders of the knee joint. Cartil. 2013;4:295ā€“312.

    ArticleĀ  Google ScholarĀ 

  13. Bendinelli P, Matteucci E, Dogliotti G, Corsi MM, Banfi G, Maroni P, et al. Molecular basis of anti-inflammatory action of platelet-rich plasma on human chondrocytes: mechanisms of NF-kappaB inhibition via HGF. J Cell Physiol. 2010;225:757ā€“66.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  14. El-Sharkawy H, Kantarci A, Deady J, Hasturk H, Liu H, Alshahat M, et al. Platelet-rich plasma: growth factors and pro- and anti-inflammatory properties. J Periodontol. 2007;78:661ā€“9.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  15. Danisovic L, Varga I, Polak S. Growth factors and chondrogenic differentiation of mesenchymal stem cells. Tissue Cell. 2011;44:69ā€“73.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  16. Demidova-Rice TN, Wolf L, Deckenback J, Hamblin MR, Herman IM. Human platelet-rich plasma- and extracellular matrix-derived peptides promote impaired cutaneous wound healing in vivo. PLoS ONE. 2012;7:e32146.

    ArticleĀ  PubMedĀ  CASĀ  PubMed CentralĀ  Google ScholarĀ 

  17. Zhang N, Wu YP, Qian SJ, Teng C, Chen S, Li H. Research progress in the mechanism of effect of PRP in bone deficiency healing. Sci World J. 2013.

  18. Bennett D, Eckersall PD, Waterston M, Marchetti V, Rota A, McCulloch E, et al. The effect of robenacoxib on the concentration of C-reactive protein in synovial fluid from dogs with osteoarthritis. BMC Vet Res. 2013;9:42.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  19. Warnock JJ, Baltzer WI, Duesterdieck-Zellmer K, Ott J. Minimally invasive synovium harvest for potential use in meniscal tissue engineering. Res Vet Sci. 2012;93:1472ā€“80.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  20. Warnock JJ, Spina J, Bobe G, Duesterdieck-Zellmer KF, Ott J, Baltzer WI, et al. Culture of canine synoviocytes on porcine intestinal submucosa scaffolds as a strategy for meniscal tissue engineering for treatment of meniscal injury in dogs. Vet J. 2014;199:49ā€“56.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  21. Reddy GK, Enwemeka CS. A simplified method for the analysis of hydroxyproline in biological tissues. Clin Biochem. 1996;29:225ā€“9.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  22. Ignatā€™eva NY, Danilov NA, Averkiev SV, Obrezkova MV, Lunin VV, Sobolā€™ EN. Determination of hydroxyproline in tissues and the evaluation of the collagen content of the tissues. J Anal Chem. 2007;62:51ā€“7.

    ArticleĀ  Google ScholarĀ 

  23. van der Sluijs JA, Geesink RG, van der Linden AJ, Bulstra SK, Kuyer R, Drukker J. The reliability of the Mankin score for osteoarthritis. J Orthop Res. 1992;10:58ā€“61.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  24. Qin J, Liu YS, Liu J, Li J, Tan Y, Li XJ, et al. Effect of angelica sinensis polysaccharides on osteoarthritis in vivo and in vitro: a possible mechanism to promote proteoglycans synthesis. Evid Based Complement Alternat Med. 2013.

  25. Na JY, Song KB, Kim S, Kwon YB, Kim DG, Lee JK, et al. Effects of HPL-04 on degenerative osteoarthritis. J Korean Soc Food Sci Nutr. 2014;43:30ā€“9.

    ArticleĀ  Google ScholarĀ 

  26. Kon E, Buda R, Filardo G, Di Martino A, Timoncini A, Cenacchi A, et al. Platelet-rich plasma: intra-articular knee injections produced favorable results on degenerative cartilage lesions. Knee Surg Sports Traumatol Arthrosc. 2010;18:472ā€“9.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  27. Franz T, Hasler EM, Hagg R, Weiler C, Jakob RP, Mainil-Varlet P. In situ compressive stiffness, biochemical composition, and structural integrity of articular cartilage of the human knee joint. Osteoarthr Cartil. 2001;9:582ā€“92.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  28. Pauli C, Whiteside R, Heras FL, Nesic D, Koziol J, Grogan SP, et al. Comparison of cartilage histopathology assessment systems on human knee joints at all stages of osteoarthritis development. Osteoarthr Cartil. 2012;20:476ā€“85.

    ArticleĀ  PubMedĀ  CASĀ  PubMed CentralĀ  Google ScholarĀ 

  29. Lorenz H, Wenz W, Ivancic M, Steck E, Richter W. Early and stable upregulation of collagen type II, collagen type I and YKL40 expression levels in cartilage during early experimental osteoarthritis occurs independent of joint location and histological grading. Arthritis Res Ther. 2005;7:R156ā€“65.

    ArticleĀ  PubMedĀ  CASĀ  PubMed CentralĀ  Google ScholarĀ 

  30. Yagi R, McBurney D, Laverty D, Weiner S, Horton Jr WE. Intrajoint comparisons of gene expression patterns in human osteoarthritis suggest a change in chondrocyte phenotype. J Orthop Res. 2005;23:1128ā€“38.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  31. Shakibaei M, John T, Seifarth C, Mobasheri A. Resveratrol inhibits IL-1 beta-induced stimulation of caspase-3 and cleavage of PARP in human articular chondrocytes in vitro. Ann N Y Acad Sci. 2007;1095:554ā€“63.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  32. Goldring MB. The role of the chondrocyte in osteoarthritis. Arthritis Rheum. 2000;43:1916ā€“26.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  33. Andia I, Maffulli N. Platelet-rich plasma for managing pain and inflammation in osteoarthritis. Nat Rev Rheumatol. 2013;9:721ā€“30.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

  34. Chung JY, Song M, Ha CW, Kim JA, Lee CH, Park YB. Comparison of articular cartilage repair with different hydrogel-human umbilical cord blood-derived mesenchymal stem cell composites in a rat model. Stem Cell Res Ther. 2014;5:39.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  35. Mishra A, Tummala P, King A, Lee B, Kraus M, Tse V, et al. Buffered platelet-rich plasma enhances mesenchymal stem cell proliferation and chondrogenic differentiation. Tissue Eng Part C Methods. 2009;15:431ā€“5.

    ArticleĀ  PubMedĀ  CASĀ  PubMed CentralĀ  Google ScholarĀ 

  36. Pereira RC, Scaranari M, Benelli R, Strada P, Reis RL, Cancedda R, et al. Dual effect of platelet lysate on human articular cartilage: a maintenance of chondrogenic potential and a transient proinflammatory activity followed by an inflammation resolution. Tissue Eng Part A. 2013;19:1476ā€“88.

    ArticleĀ  PubMedĀ  CASĀ  Google ScholarĀ 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Young-Sam Kwon.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authorsā€™ contributions

SY and YSK built the idea and protocols of this paper and wrote the manuscript. SKK participated in data collection and analysis. All authors approved this manuscript.

Additional file

Additional file 1:

This contains Figures S1ā€“S6 and Table S1.Ā Fig.Ā S1-S5)Ā Representative images of IL-1Ī²+,COX-2+, iNOS+, Caspase-3+ and IFN-Ī³+ cells, Fig.Ā S6)Ā Result of FACS analysis,Ā Table S1)Ā Primary antisera and detection kits for immunohistochemistry used in this study

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yun, S., Ku, SK. & Kwon, YS. Adipose-derived mesenchymal stem cells and platelet-rich plasma synergistically ameliorate the surgical-induced osteoarthritis in Beagle dogs. J Orthop Surg Res 11, 9 (2016). https://doi.org/10.1186/s13018-016-0342-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13018-016-0342-9

Keywords