Skip to main content

Therapeutic potential of mesenchymal stem cell based therapy for osteoarthritis

Abstract

Osteoarthritis (OA) is a chronic degenerative disease affecting articular cartilage in joints, and it is a leading cause of disability in the United States. Current pharmacological treatment strategies are ineffective to prevent the OA progression; however, cellular therapies have the potential to regenerate the lost cartilage, combat cartilage degeneration, provide pain relief, and improve patient mobility. One of the most promising sources of cellular regenerative medicine is from mesenchymal stem cells (MSCs). MSCs can be isolated from adipose tissue, bone marrow, synovial tissue, and other sources. The aim of this review is to compile recent advancement in cellular based therapy more specifically in relation to MSCs in the treatment of osteoarthritis.

Introduction

Osteoarthritis (OA) is a chronic degenerative disease of articular cartilage that is the leading cause of joint disease in the United States. OA is characterized by the inability of chondrocytes to produce adequate functional matrix to compensate for matrix damage and depletion. Comorbidities such as aging, obesity, heart disease, diabetes, and mechanical stress become prevalent concerns in patients with osteoarthritis; in 2013, the center for disease control and prevention (CDC) found that 52.5 million adults over the age of 18 had self-reported physician-diagnosed arthritis, which is 22.7 % of the adult population [1, 2]. Treatment costs for solely knee OA are estimated to be $185.5 billion per year [3]. Conventional pharmacological interventions are not effective to prevent the OA progression. Recent advances in cell therapeutics offer potential methods to treat OA.

Current therapies

OA is a chronic degenerative condition with no cure. Patients often experience pain, stiffness, swelling, loss of mobility, loss of flexibility, and weight gain secondary to reduced mobility/activity. Conventional OA therapeutics are directed toward symptomatic treatment, mainly pain management. Current treatment modalities for OA such as exercise, anti-inflammatory medication, and surgery are summarized below in Table 1. Current traditional therapies for OA have numerous downfalls in being perfect treatment strategies. Most importantly, these therapies fail to regenerate degenerated cartilages and prevent further degenerative processes. Recent advancements in molecular biology, regenerative, and reparative medicine offer new hope to develop novel therapeutic agents for OA like conditions.

Table 1 Current traditional pharmacological therapies for osteoarthritis

Cellular therapies

Advancement in the field of cellular therapy for osteoarthritis is an exciting and quickly evolving area of research and medicine. Current cellular therapies are summarized in Table 2 and Fig. 1. One example of a cell based treatment that has improved over the past 20 years is a technique called autologous chondrocyte implantation (ACI). ACI is the only cellular based treatment with FDA approval and works by surgically obtaining autologous cartilage (i.e. the patient’s own cartilage) from a non-weight bearing area of the affected joint, isolating the chondrocytes via collagenase, expanding the chondrocytes in vitro, and finally injecting the cultured chondrocytes into the periosteum of the affected joint, with a graft to hold the cells in the desired location. [15, 19, 20]. The grafts that hold the cells in place have evolved from periosteal flaps and collagen I/III covered membranes to the latest method, matrix-induced ACI (MACI) [19]. While ACI has shown a success rate in patient improvement from 76 to 86 % (with Viste et al. showing the highest success), numerous problems and questions have been raised surrounding the procedure, including de-differentiation of the chondrocytes. ACI is also limited only the site of cartilage damage and not for generalized OA treatment [1921].

Table 2 Therapeutic potentials and limitations of cell based therapies in osteoarthritis
Fig. 1
figure 1

Schematic diagram illustrating the current clinical approaches to cell-based therapy for cartilage tissue engineering

Recent development in stem cell tissue engineering has created a lot of excitement in the field of cartilage regeneration biology. Stem cells are progenitor cells which differentiate into various cell types including osteoblasts, osteocytes, adipocytes, and cartilage [3740]. Because of this, stem cells are being investigated for their abilities to regenerate cartilage in OA patients. These cells also have demonstrated the capability to inhibit T cell growth, thus showing that they have the ability to down-regulate the natural inflammatory response in OA [41]. While stem cells can both differentiate into new cartilage cells as well as suppress inflammation, recent studies have found that stem cells can also combat OA through paracrine mechanisms. They release important cytokines such as epidermal growth factor (EGF), transforming growth factor beta (TGFB), vascular endothelial growth factor (VEGF), as well as other cytokines and new cartilage proteins that are essential in combating OA and degenerative processes. It has also been suggested that stem cells could release cytokines and proteins that could help combat neurogenic pain, which would have numerous benefits in treating OA pain [40, 42]. Further research needs to be done in order to better understand stem cells mechanism of action in regard to their immunomodulatory, differentiating, paracrine, regenerative, and anti-inflammatory abilities as well as their cellular trafficking mechanisms.

Two types of stem cells being investigated are embryonic stem cells (ESCs, captured from embryonic mammalian cells) and induced pluripotent stem cells (iPSCs). Both cells possess the pluripotent ability to differentiate into chondrocytes or any type of cell; ESCs have been found to improve cartilage repair in animal models, and Wei et al. have generated iPSCs from human OA chondrocytes and subsequently induced the cells into chondrocytic differentiation [15, 25]. While there is some promise for both ESCs and iPSCs to differentiate into human cartilage to treat OA, many problems exist such as both cell types tend to cause teratoma growth as well as immunogenicity [26].

The other type of stem cell currently being investigated for its ability to treat OA is the mesenchymal stem cell (MSC). MSCs are a heterogeneous group of stromal cells that can come from a variety of sources including adipose tissue, bone marrow, and synovium; numerous studies have proven MSCs’ abilities to differentiate into chondrocytes and regeneratively treat OA [20]. While MSCs have a limited proliferative potential in comparison to pluripotent stem cells, many advantages exist for MSC based therapies [31]. MSCs and iPSCs offer the most realistic and best potential for viable regenerative cell treatment of OA; however, MSC based therapies have less risks associated with them and an easier means of production. The source of MSCs for treatment of OA is an important factor in cartilage tissue engineering and each cell type has its pros and cons (Table 2).

Adipose-derived Mesenchymal Stem Cells (ADSCs)

Adipose-derived mesenchymal stem cells (ADSCs) can be harvested from the patient’s own adipose tissue most commonly via surgical resection or liposuction; specifically, infrapatellar fat pads (IFPs) provide cells with higher chondrogenic potential in comparison to other sources [43]. ADSC therapy for OA in animal studies is well documented, and the process of isolating ADSCs is not overly invasive [19]. Toghraie et al. demonstrated that ADSCs derived from IFPs in rabbits given to rabbits with OA-induced knees had less cartilage destruction, less subchondral sclerosis, less osteophyte buildup, as well as better cartilage overall than the control group [43]. Desando et al. also demonstrated that autologous ADSC therapy decreased the progression of degeneration in cartilage and in the synovial membrane. Furthermore, autologous ADSC therapy improved meniscal repair. Desando et al. suggested these findings could be due to the release of growth factors and cytokines. The regenerative effect of autologous ADSCs is dose and time dependent [44]. Another group also reported cartilage regeneration following autologous ADSC therapy in a surgically induced osteoarthritic sheep model. Autologous ADSCs were labeled and intra-articularly injected, leading to the cells populating the area of damaged cartilage as well as a decreased progression of OA [45]. While these animal models showed the promise of ADSCs in OA therapy, more studies like these need to be done in order to better understand the mechanisms so that they can be practiced in a routine clinical setting.

Several clinical studies also prove ADSCs’ efficacy in treating OA in human patients. Koh et al. treated patients with knee OA undergoing arthroscopic debridement with injected autologous ADSCs derived from IFPs and prepared in platelet rich plasma (PRP). Treated patients demonstrated improved mobility and function in the affected knees, reduced pain levels, and better clinical prognoses in a 1 year follow up [46]. In a 2 year follow up, Koh et al. found the patients had significantly improved Western Ontario and McMaster Universities Osteoarthritis (WOMAC) pain scores, VAS pain scores, and cartilage regeneration as confirmed by MRI [47]. This study suggests that these intra-articular injections are safe, and more clinical trials like these should be done to improve surgical and clinical outcomes. In a different proof-of-concept clinical trial, autologous ADSCs were injected in patients with knee OA. The high dose injection group had increased WOMAC scores 6 months after injections, decreased cartilage defects in affected areas, and increased cartilage volumes with thick, hyaline cartilage-like regeneration [48]. These results further proved the promising efficacy of autologous ADSCs in treating OA in humans, as well as demonstrated its safety as there were no adverse events. In 2011, Pak demonstrated the potential of ADSCs in osteonecrosis of the hip and OA in the knees of several patients. Results revealed bone formation in the osteonecrosis patients as well as cartilage regeneration in the OA knee patients. These patients’ MRIs had increased meniscus cartilage volume and thickness due to the ADSCs injections [49]. More similar clinical trials are necessary to further prove ADSCs’ efficacy and safety for routine use in the human OA setting.

Bone marrow-derived mesenchymal stem cells (BM-MSCs)

Another source of MSCs for the treatment of OA is bone marrow-derived MSCs (BM-MSCs). BM-MSCs have a higher chondrogenic capability than ADSCs [50], and they have been studied more extensively than ADSCs [19]. Numerous animal models have demonstrated the potential therapeutic value of BM-MSCs, including one by Chiang et al. in 2016. They used allogenic BM-MSCs in combination with hyaluronic acid to treat knee OA-in a rabbit model, with the contralateral osteoarthritic knee only receiving hyaluronic acid. These treated rabbits were compared to untreated OA-induced rabbits. The joints treated with BM-MSCs and hyaluronic acid underwent less cartilage loss, fewer surface abrasions, and improved cartilage content [51]. This study showed that allogenic BM-MSCs can reduce the progression of OA. In a sheep OA model, autologous BM-MSCs were intra-articularly injected into the knees of sheep with arthroscopically-caused medial femorotibial condylar and meniscal defects. In comparison to the control group, the treated sheep showed signs of regeneration in their articular cartilage and menisci. The treatment group exhibited signs of statistically significant improvement in respect to both microscopic and histological guidelines [52]. These animal studies demonstrate the capability of BM-MSCs to combat OA.

In a human trial, Orozco et al. used autologous BM-MSCs to treat OA knee patients who were unresponsive to conservative treatments. BM-MSCs were intra-articularly injected, and their results indicated strong clinical efficacies such as improved cartilage quality in T2 mapping in 11 of the 12 patients and pain relief without hospitalization or surgery [53]. These findings suggest that BM-MSCs can be safely implemented in treatment strategies for treatment-resistant OA patients. Another study tested the efficacy of autologous BM-MSCs in treating patients with knee, hip, or ankle OA. Each patient received one autologous BM-MSC injection after the cells were isolated and cultured, and they were followed for 30 months. All patients enjoyed increased walking distances, improved WOMAC scores, and improved cartilage regeneration as demonstrated on MRI [54]. This study once again presented the regenerative potential of BM-MSCs in OA joints with minimal side effects. A randomized control trial in 2015 demonstrated the efficacy of allogenic BM-MSCs in treating knee OA. The study treated OA patients who had chronic knee pain and were unresponsive to conservative OA treatments with intra-articularly injected BM-MSCs in comparison to the control group who only received intra-articularly injected hyaluronic acid. The treatment group demonstrated decreases in poor cartilage areas, improved cartilage quality, and pain relief [55]. These findings are promising because it demonstrates BM-MSCs’ ability to inhibit the progression of OA; however, more clinical and basic science research, including human trials, must be done to understand molecular mechanisms and how the cells can better prevent the progression of OA in humans. Also, more studies must be done in order to effectively compare the chondrogenic abilities of different classes of stem cells (i.e. ADSCs and BM-MSCs) [5659].

Synovial-derived mesenchymal stem cells (S-MSCs)

Several human studies have been conducted on ADSCs and BM-MSC treatments for OA, but less has been done in recent times with synovial-derived MSCs (S-MSCs). In a rat knee OA model, S-MSCs injected weekly, rather than at a single time, were found to have migrated into the synovium and retained their undifferentiated S-MSC properties. The S-MSCs increased genetic expression of chondroprotective proteins such as BMP-2 and an anti-inflammatory gene, TSG-6 [60]. This suggests that periodic injections of S-MSCs can allow the MSCs to retain their MSC properties as well as inhibit the advancement of OA through genetic transcription. In a microminipig model, S-MSCs demonstrated the ability to enhance repair of longitudinally torn menisci in avascular areas. The group treated with S-MSCs had significantly improved meniscal healing at 12 weeks in comparison to the control group macroscopically, histologically, and by T1rho mapping [61]. In 2014, Hatsushika et al. further demonstrated the potential of S-MSCs by intra-articularly injecting them into the knees of pigs that had underwent medial meniscal resections. These damaged menisci regenerated cartilage significantly better than the control group in respect to MRI and histology. The treatment group’s cartilage was better preserved, and new synovial tissue filled the area of meniscal resection at 2 weeks [62]. This study, along with the previously mentioned studies, demonstrate that S-MSCs can provide a real answer in preventing the progression of OA as well as promoting regeneration of cartilage.

Stem cell-derived exosomes

Although MSCs have demonstrated the unique ability and promise to combat degenerative diseases such as OA, they possess another mechanism of regenerative abilities not previously discussed: their exosomal products. Exosomes are packaged microvesicles that can contain proteins, lipids, factors, and/or genetic material that can be released in times of cellular stress [63, 64]. Furthermore, recent studies demonstrated transfer of genetic (miRNA, mRNA) material and protein through exosomal machinery [64, 65]. Large scale exosomes can be produced in the laboratory from stem cells and these exosomes can be used to combat the disease progression [64, 66]. If MSCs can be grown in culture and an environment similar to one of a chondrocyte undergoing osteoarthritic changes (via osteoarthritic cytokines), then the MSCs will release chondroprotective exosomes in response to the stress; these exosomal products could then be screened for, packaged in exosomes, and given to OA patients to promote cartilage regeneration [64]. Some miRNAs have already been identified in being involved in chondrogenesis and cartilage degeneration, including miRNA-101, miRNA-140, and miRNA 455 [67, 68]. Xu et al. found that human BM-MSCs release exosomes containing miRNAs that upregulate the Wnt pathway, leading to osteogenic differentiation [69]. These exosomal products can help explain how MSCs perform their regenerative abilities in combatting OA in a paracrine fashion, although it is impossible to say at this time how much of the healing is solely due to exosomes, and they may only offer a one-time relief instead of continuous relief that MSC-based treatment has demonstrated. More research is required to better understand how MSC-derived exosomal products mechanistically work, how they can be identified, and how they can be produced in Good Manufacturing Practices (GMP) in order to prevent the progression of OA in clinical medicine.

Conclusions

Current treatment strategies for OA are inadequate and costly. Due to the increasing incidence and prevalence of OA, more innovative and effective therapeutic modalities need to be investigated, including MSCs. More randomized clinical trials need to be completed in order to demonstrate the efficacy, safety, and benefits of MSCs in treating patients with OA. Most of MSC research on humans only involves knee OA, and additional analysis should include clinical trials for ankle OA, shoulder OA, hip OA, and elbow OA. MSC-based cellular therapy has the potential and opportunity to effectively combat OA, but more extensive clinical trial and animal studies are required to understand the basic molecular mechanisms of MSC dependent cartilage regeneration. Further research is also necessary to better understand the potential of MSC-derived exosomes in the treatment of OA.

References

  1. Qin J, Theis KA, Barbour KE, Helmick CG, Baker NA, Brady TJ (2015) Centers for disease control and prevention (cdc). impact of arthritis and multiple chronic conditions on selected life domains—United States, 2013. MMWR Morb Mortal Wkly Rep 64(21):578–582

    PubMed  Google Scholar 

  2. Centers for Disease Control and Prevention (CDC) (2013) Prevalence of doctor-diagnosed arthritis and arthritis-attributable activity limitation—United States, 2010–2012. MMWR Morb Mortal Wkly Rep 62(44):869–873

    Google Scholar 

  3. Field T (2016) Knee osteoarthritis pain in the elderly can be reduced by massage therapy, yoga and tai chi: a review. Complement Ther Clin Pract 22:87–92

    Article  PubMed  Google Scholar 

  4. Laev SS, Salakhutdinov NF (2015) Anti-arthritic agents: progress and potential. Bioorg Med Chem 23(13):3059–3080

    Article  CAS  PubMed  Google Scholar 

  5. Dockerty T, Latham SK, Smith TO (2016) Why don’t patients take their analgesics? A meta-ethnography assessing the perceptions of medication adherence in patients with osteoarthritis. Rheumatol Int 36(5):731–739

    Article  CAS  PubMed  Google Scholar 

  6. Kongtharvonskul J, Anothaisintawee T, McEvoy M, Attia J, Woratanarat P, Thakkinstian A (2015) Efficacy and safety of glucosamine, diacerein, and NSAIDs in osteoarthritis knee: a systematic review and network meta-analysis. Eur J Med Res 13(20):24

    Article  Google Scholar 

  7. Vučković S, Vujović KS, Medić B, Srebro D, Mostić D. Prevention of renal complications induced by non-steroidal anti-inflammatory drugs. Curr Med Chem. 2016

  8. Lee T, Lu N, Felson DT, Choi HK, Dalal DS, Zhang Y, Dubreuil M (2016) Use of non-steroidal anti-inflammatory drugs correlates with the risk of venous thromboembolism in knee osteoarthritis patients: a UK population-based case–control study. Rheumatology 55(6):1099–1105

    Article  PubMed  Google Scholar 

  9. Yu SP, Hunter DJ (2015) Managing osteoarthritis. Aust Prescr 38(4):115–119

    Article  PubMed  PubMed Central  Google Scholar 

  10. Bennell KL, Hunter DJ, Hinman RS (2012) Management of osteoarthritis of the knee. BMJ 30(345):e4934

    Article  Google Scholar 

  11. Fransen M, McConnell S (2009) Land-based exercise for osteoarthritis of the knee: a metaanalysis of randomized controlled trials. J Rheumatol 36(6):1109–1117

    Article  PubMed  Google Scholar 

  12. Bosomworth NJ (2009) Exercise and knee osteoarthritis: benefit or hazard? Can Fam Physician 55(9):871–878

    PubMed  PubMed Central  Google Scholar 

  13. Henry SG (2016) Evaluating the risks of opioid use for chronic pain: moving beyond overdose. J Gen Intern Med 31(5):453–454

    Article  PubMed  Google Scholar 

  14. Wright EA, Katz JN, Abrams S, Solomon DH, Losina E (2014) Trends in prescription of opioids from 2003–2009 in persons with knee osteoarthritis. Arthritis Care Res 66(10):1489–1495

    Article  Google Scholar 

  15. Zhang W, Ouyang H, Dass CR, Xu J (2016) Current research on pharmacologic and regenerative therapies for osteoarthritis. Bone Res 1(4):15040

    Article  Google Scholar 

  16. Bannuru RR, Natov NS, Obadan IE, Price LL, Schmid CH, McAlindon TE (2009) Therapeutic trajectory of hyaluronic acid versus corticosteroids in the treatment of knee osteoarthritis: a systematic review and meta-analysis. Arthritis Rheum 61(12):1704–1711

    Article  CAS  PubMed  Google Scholar 

  17. Jevsevar DS, Brown GA, Jones DL, Matzkin EG, Manner PA, Mooar P, Schousboe JT, Stovitz S, Sanders JO, Bozic KJ, Goldberg MJ, Martin WR 3rd, Cummins DS, Donnelly P, Woznica A, Gross L (2013) The American Academy Of Orthopaedic Surgeons evidence-based guideline on: treatment of osteoarthritis of the knee, 2nd edition. J Bone Joint Surg Am 95(20):1885–1886

    PubMed  Google Scholar 

  18. Grayson CW, Decker RC (2012) Total joint arthroplasty for persons with osteoarthritis. PM R 4(5):S97–S103

    Article  PubMed  Google Scholar 

  19. Nazempour A, Van Wie BJ (2016) Chondrocytes, mesenchymal stem cells, and their combination in articular cartilage regenerative medicine. Ann Biomed Eng 44(5):1325–1354

    Article  CAS  PubMed  Google Scholar 

  20. Mobasheri A, Kalamegam G, Musumeci G, Batt ME (2014) Chondrocyte and mesenchymal stem cell-based therapies for cartilage repair in osteoarthritis and related orthopaedic conditions. Maturitas 78(3):188–198

    Article  CAS  PubMed  Google Scholar 

  21. Viste A, Piperno M, Desmarchelier R, Grosclaude S, Moyen B, Fessy MH (2012) Autologous chondrocyte implantation for traumatic full-thickness cartilage defects of the knee in 14 patients: 6-year functional outcomes. Orthop Traumatol Surg Res 98(7):737–743

    Article  CAS  PubMed  Google Scholar 

  22. Niemeyer P, Porichis S, Steinwachs M, Erggelet C, Kreuz PC, Schmal H, Uhl M, Ghanem N, Südkamp NP, Salzmann G (2014) Long-term outcomes after first-generation autologous chondrocyte implantation for cartilage defects of the knee. Am J Sports Med 42(1):150–157

    Article  PubMed  Google Scholar 

  23. Kreuz PC, Müller S, von Keudell A, Tischer T, Kaps C, Niemeyer P, Erggelet C (2013) Influence of sex on the outcome of autologous chondrocyte implantation in chondral defects of the knee. Am J Sports Med 41(7):1541–1548

    Article  PubMed  Google Scholar 

  24. Gomoll AH, Filardo G, deGirolamo L, Espregueira-Mendes J, Marcacci M, Rodkey WG, Steadman JR, Zaffagnini S, Kon E (2012) Surgical treatment for early osteoarthritis. Part I: cartilage repair procedures. Knee Surg Sports Traumatol Arthrosc 20(3):450–466

    Article  CAS  PubMed  Google Scholar 

  25. Wei Y, Zeng W, Wan R, Wang J, Zhou Q, Qiu S, Singh SR (2012) Chondrogenic differentiation of induced pluripotent stem cells from osteoarthritic chondrocytes in alginate matrix. Eur Cell Mater 12(23):1–12

    Google Scholar 

  26. Chang YH, Liu HW, Wu KC, Ding DC (2016) Mesenchymal stem cells and their clinical applications in osteoarthritis. Cell Transplant 25(5):937–950

    Article  PubMed  Google Scholar 

  27. Ding DC, Shyu WC, Lin SZ, Liu HW, Chiou SH, Chu TY (2012) Human umbilical cord mesenchymal stem cells support nontumorigenic expansion of human embryonic stem cells. Cell Transplant 21(7):1515–1527

    Article  PubMed  Google Scholar 

  28. Kobold S, Guhr A, Kurtz A, Löser P (2015) Human embryonic and induced pluripotent stem cell research trends: complementation and diversification of the field. Stem Cell Rep 4(5):914–925

    Article  CAS  Google Scholar 

  29. Suchorska WM, Lach MS, Richter M, Kaczmarczyk J, Trzeciak T (2016) Bioimaging: an useful tool to monitor differentiation of human embryonic stem cells into chondrocytes. Ann Biomed Eng 44(5):1845–1859

    Article  PubMed  Google Scholar 

  30. Son MY, Kim HJ, Kim MJ, Cho YS (2011) Physical passaging of embryoid bodies generated from human pluripotent stem cells. PLoS ONE 6(5):e19134

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Augustyniak E, Trzeciak T, Richter M, Kaczmarczyk J, Suchorska W (2015) The role of growth factors in stem cell-directed chondrogenesis: a real hope for damaged cartilage regeneration. Int Orthop 39(5):995–1003

    Article  PubMed  Google Scholar 

  32. Cherry AB, Daley GQ (2013) Reprogrammed cells for disease modeling and regenerative medicine. Annu Rev Med 64:277–290

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Giuliani N, Lisignoli G, Magnani M, Racano C, Bolzoni M, Dalla Palma B, Spolzino A, Manferdini C, Abati C, Toscani D, Facchini A, Aversa F. New insights into osteogenic and chondrogenic differentiation of human bone marrow mesenchymal stem cells and their potential clinical applications for bone regeneration in pediatric orthopaedics. Stem Cells Int. 2013;2013:312501

  34. Davatchi F, Abdollahi BS, Mohyeddin M, Shahram F, Nikbin B (2011) Mesenchymal stem cell therapy for knee osteoarthritis preliminary report of four patients. Int J Rheum Dis 14(2):211–215

    Article  PubMed  Google Scholar 

  35. Orozco L, Munar A, Soler R, Alberca M, Soler F, Huguet M, Sentís J, Sánchez A, García-Sancho J (2013) Treatment of knee osteoarthritis with autologous mesenchymal stem cells: a pilot study. Transplantation 95(12):1535–1541

    Article  CAS  PubMed  Google Scholar 

  36. Bianco P, Cao X, Frenette PS, Mao JJ, Robey PG, Simmons PJ, Wang CY (2013) The meaning, the sense and the significance: translating the science of mesenchymal stem cells into medicine. Nat Med 19(1):35–42

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Fulzele S, Chothe P, Sangani R, Chutkan N, Hamrick M, Bhattacharyya M, Prasad PD, Zakhary I, Bowser M, Isales C, Ganapathy V (2013) Sodium-dependent vitamin C transporter SVCT2: expression and function in bone marrow stromal cells and in osteogenesis. Stem Cell Res 10(1):36–47

    Article  CAS  PubMed  Google Scholar 

  38. Sangani R, Pandya CD, Bhattacharyya MH, Periyasamy-Thandavan S, Chutkan N, Markand S, Hill WD, Hamrick M, Isales C, Fulzele S (2014) Knockdown of SVCT2 impairs in vitro cell attachment, migration and wound healing in bone marrow stromal cells. Stem Cell Res 12(2):354–363

    Article  CAS  PubMed  Google Scholar 

  39. Sangani R, Periyasamy-Thandavan S, Kolhe R, Bhattacharyya MH, Chutkan N, Hunter M, Isales C, Hamrick M, Hill WD, Fulzele S (2015) MicroRNAs-141 and 200a regulate the SVCT2 transporter in bone marrow stromal cells. Mol Cell Endocrinol 15(410):19–26

    Article  Google Scholar 

  40. Freitag J, Bates D, Boyd R, Shah K, Barnard A, Huguenin L, Tenen A (2016) Mesenchymal stem cell therapy in the treatment of osteoarthritis: reparative pathways, safety and efficacy—a review. BMC Musculoskelet Disord 17(1):230

    Article  PubMed  PubMed Central  Google Scholar 

  41. Djouad F, Bouffi C, Ghannam S, Noël D, Jorgensen C (2009) Mesenchymal stem cells: innovative therapeutic tools for rheumatic diseases. Nat Rev Rheumatol 5(7):392–399

    Article  CAS  PubMed  Google Scholar 

  42. Im GI (2016) Regeneration of articular cartilage using adipose stem cells. J Biomed Mater Res A 104(7):1830–1844

    Article  CAS  PubMed  Google Scholar 

  43. Toghraie FS, Chenari N, Gholipour MA, Faghih Z, Torabinejad S, Dehghani S, Ghaderi A (2011) Treatment of osteoarthritis with infrapatellar fat pad derived mesenchymal stem cells in Rabbit. Knee 18(2):71–75

    Article  CAS  PubMed  Google Scholar 

  44. Desando G, Cavallo C, Sartoni F, Martini L, Parrilli A, Veronesi F, Fini M, Giardino R, Facchini A, Grigolo B (2013) Intra-articular delivery of adipose derived stromal cells attenuates osteoarthritis progression in an experimental rabbit model. Arthritis Res Ther 15(1):R22

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ude CC, Sulaiman SB, Min-Hwei N, Hui-Cheng C, Ahmad J, Yahaya NM, Saim AB, Idrus RB (2014) Cartilage regeneration by chondrogenic induced adult stem cells in osteoarthritic sheep model. PLoS ONE 9(6):e98770

    Article  PubMed  PubMed Central  Google Scholar 

  46. Koh YG, Choi YJ (2012) Infrapatellar fat pad-derived mesenchymal stem cell therapy for knee osteoarthritis. Knee 19(6):902–907

    Article  PubMed  Google Scholar 

  47. Koh YG, Jo SB, Kwon OR, Suh DS, Lee SW, Park SH, Choi YJ (2013) Mesenchymal stem cell injections improve symptoms of knee osteoarthritis. Arthroscopy 29(4):748–755

    Article  PubMed  Google Scholar 

  48. Jo CH, Lee YG, Shin WH, Kim H, Chai JW, Jeong EC, Kim JE, Shim H, Shin JS, Shin IS, Ra JC, Oh S, Yoon KS (2014) Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells 32(5):1254–1266

    Article  CAS  PubMed  Google Scholar 

  49. Pak J (2011) Regeneration of human bones in hip osteonecrosis and human cartilage in knee osteoarthritis with autologous adipose-tissue-derived stem cells: a case series. J Med Case Rep 7(5):296

    Article  Google Scholar 

  50. Afizah H, Yang Z, Hui JH, Ouyang HW, Lee EH (2007) A comparison between the chondrogenic potential of human bone marrow stem cells (BMSCs) and adipose-derived stem cells (ADSCs) taken from the same donors. Tissue Eng 13(4):659–666

    Article  CAS  PubMed  Google Scholar 

  51. Chiang ER, Ma HL, Wang JP, Liu CL, Chen TH, Hung SC (2016) Allogeneic mesenchymal stem cells in combination with hyaluronic acid for the treatment of osteoarthritis in rabbits. PLoS ONE 11(2):e0149835

    Article  PubMed  PubMed Central  Google Scholar 

  52. Caminal M, Fonseca C, Peris D, Moll X, Rabanal RM, Barrachina J, Codina D, García F, Cairó JJ, Gòdia F, Pla A, Vives J (2014) Use of a chronic model of articular cartilage and meniscal injury for the assessment of long-term effects after autologous mesenchymal stromal cell treatment in sheep. N Biotechnol 31(5):492–498

    Article  CAS  PubMed  Google Scholar 

  53. Orozco L, Munar A, Soler R, Alberca M, Soler F, Huguet M, Sentís J, Sánchez A, García-Sancho J (2013) Treatment of knee osteoarthritis with autologous mesenchymal stem cells: a pilot study. Transplantation 95(12):1535–1541

    Article  CAS  PubMed  Google Scholar 

  54. Emadedin M, Ghorbani Liastani M, Fazeli R, Mohseni F, Moghadasali R, Mardpour S, Hosseini SE, Niknejadi M, Moeininia F, Aghahossein Fanni A, Baghban Eslaminejhad R, Vosough Dizaji A, Labibzadeh N, Mirazimi Bafghi A, Baharvand H, Aghdami N (2015) Long-term follow-up of intra-articular injection of autologous mesenchymal stem cells in patients with knee, ankle, or hip osteoarthritis. Arch Iran Med 18(6):336–344

    PubMed  Google Scholar 

  55. Vega A, Martín-Ferrero MA, Del Canto F, Alberca M, García V, Munar A, Orozco L, Soler R, Fuertes JJ, Huguet M, Sánchez A, García-Sancho J (2015) Treatment of knee osteoarthritis with allogeneic bone marrow mesenchymal stem cells: a randomized controlled trial. Transplantation 99(8):1681–1690

    Article  CAS  PubMed  Google Scholar 

  56. Wankhade UD, Shen M, Kolhe R, Fulzele S (2016) Advances in adipose-derived stem cells isolation, characterization, and application in regenerative tissue engineering. Stem Cells Int 2016:3206807

    Article  PubMed  PubMed Central  Google Scholar 

  57. Kern S, Eichler H, Stoeve J, Klüter H, Bieback K (2006) Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells 24(5):1294–1301

    Article  CAS  PubMed  Google Scholar 

  58. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH (2002) Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 13(12):4279–4295

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. De Ugarte DA, Morizono K, Elbarbary A, Alfonso Z, Zuk PA, Zhu M, Dragoo JL, Ashjian P, Thomas B, Benhaim P, Chen I, Fraser J, Hedrick MH (2003) Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs 174(3):101–109

    Article  PubMed  Google Scholar 

  60. Ozeki N, Muneta T, Koga H, Nakagawa Y, Mizuno M, Tsuji K, Mabuchi Y, Akazawa C, Kobayashi E, Matsumoto K, Futamura K (2016) Not single but periodic injections of synovial mesenchymal stem cells maintain viable cells in knees and inhibit osteoarthritis progression in rats. Osteoarthritis Cartilage 24(6):1061–1070

    Article  CAS  PubMed  Google Scholar 

  61. Nakagawa Y, Muneta T, Kondo S, Mizuno M, Takakuda K, Ichinose S, Tabuchi T, Koga H, Tsuji K, Sekiya I (2015) Synovial mesenchymal stem cells promote healing after meniscal repair in microminipigs. Osteoarthritis Cartilage 23(6):1007–1017

    Article  CAS  PubMed  Google Scholar 

  62. Hatsushika D, Muneta T, Nakamura T, Horie M, Koga H, Nakagawa Y, Tsuji K, Hishikawa S, Kobayashi E, Sekiya I (2014) Repetitive allogeneic intraarticular injections of synovial mesenchymal stem cells promote meniscus regeneration in a porcine massive meniscus defect model. Osteoarthritis Cartilage 22(7):941–950

    Article  CAS  PubMed  Google Scholar 

  63. Théry C (2011) Exosomes: secreted vesicles and intercellular communications. Biol Rep 3(15):130

    Google Scholar 

  64. Burke J, Kolhe R, Hunter M, Isales C, Hamrick M, Fulzele S (2016) Stem cell-derived exosomes: a potential alternative therapeutic agent in orthopaedics. Stem Cells Int. 2016:5802529

    Article  PubMed  PubMed Central  Google Scholar 

  65. Hannafon BN, Carpenter KJ, Berry WL, Janknecht R, Dooley WC, Ding WQ (2015) Exosome-mediated microRNA signaling from breast cancer cells is altered by the anti-angiogenesis agent docosahexaenoic acid (DHA). Mol Cancer 16(14):133

    Article  Google Scholar 

  66. Raposo G, Stoorvogel W (2013) Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 200(4):373–383

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Dai L, Zhang X, Hu X, Liu Q, Man Z, Huang H, Meng Q, Zhou C, Ao Y (2015) Silencing of miR-101 prevents cartilage degradation by regulating extracellular matrix-related genes in a rat model of osteoarthritis. Mol Ther 23(8):1331–1340

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Min Z, Zhang R, Yao J, Jiang C, Guo Y, Cong F, Wang W, Tian J, Zhong N, Sun J, Ma J, Lu S (2015) MicroRNAs associated with osteoarthritis differently expressed in bone matrix gelatin (BMG) rat model. Int J Clin Exp Med 8(1):1009–1017

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Xu JF, Yang GH, Pan XH, Zhang SJ, Zhao C, Qiu BS, Gu HF, Hong JF, Cao L, Chen Y, Xia B, Bi Q, Wang YP (2014) Altered microRNA expression profile in exosomes during osteogenic differentiation of human bone marrow-derived mesenchymal stem cells. PLoS ONE 9(12):e114627

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Authors’ contributions

JB, MoH, RK, CI, MH and SF each made substantive intellectual contributions to the work described in this review paper. SF conceived the idea for the manuscript, and drafted the initial manuscript with JB. MoH, RK and CL provide clinical relevance and insight. MH made the final edits to the manuscript. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sadanand Fulzele.

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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Burke, J., Hunter, M., Kolhe, R. et al. Therapeutic potential of mesenchymal stem cell based therapy for osteoarthritis. Clin Trans Med 5, 27 (2016). https://doi.org/10.1186/s40169-016-0112-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40169-016-0112-7

Keywords