Skip to main content
Clinical Orthopaedics and Related Research logoLink to Clinical Orthopaedics and Related Research
. 2011 Mar 15;469(10):2706–2715. doi: 10.1007/s11999-011-1857-3

The Role of Growth Factors in Cartilage Repair

Lisa A Fortier 1,, Joseph U Barker 2, Eric J Strauss 2, Taralyn M McCarrel 1, Brian J Cole 2
PMCID: PMC3171543  PMID: 21403984

Abstract

Background

Full-thickness chondral defects and early osteoarthritis continue to present major challenges for the patient and the orthopaedic surgeon as a result of the limited healing potential of articular cartilage. The use of bioactive growth factors is under consideration as a potential therapy to enhance healing of chondral injuries and modify the arthritic disease process.

Questions/purposes

We reviewed the role of growth factors in articular cartilage repair and identified specific growth factors and combinations of growth factors that have the capacity to improve cartilage regeneration. Additionally, we discuss the potential use of platelet-rich plasma, autologous-conditioned serum, and bone marrow concentrate preparations as methods of combined growth factor delivery.

Methods

A PubMed search was performed using key words cartilage or chondrocyte alone and in combination with growth factor. The search was open for original manuscripts and review papers and open for all dates. From these searches we selected manuscripts investigating the effects of growth factors on extracellular matrix synthesis and excluded those investigating molecular mechanisms of action.

Results

By modulating the local microenvironment, the anabolic and anticatabolic effects of a variety of growth factors have demonstrated potential in both in vitro and animal studies of cartilage injury and repair. Members of the transforming growth factor-β superfamily, fibroblast growth factor family, insulin-like growth factor-I, and platelet-derived growth factor have all been investigated as possible treatment augments in the management of chondral injuries and early arthritis.

Conclusions

The application of growth factors in the treatment of local cartilage defects as well as osteoarthritis appears promising; however, further research is needed at both the basic science and clinical levels before routine application.

Introduction

Growth factors are a group of biologically active polypeptides produced by the body that can stimulate cellular division, growth, and differentiation. In articular cartilage, numerous growth factors work in concert to regulate development and homeostasis of articular cartilage throughout life [39]. Therefore, growth factors offer promising treatments for enhanced regeneration of cartilage in focal articular cartilage defects or in situations of more widespread cartilage loss such as those that occur in osteoarthritis (OA). When considering an injured or osteoarthritic joint, the effects of any treatment such as growth factors on the cartilage, synovial lining, ligaments, tendons, meniscus, any exposed subchondral bone as well as on mesenchymal stem cells (MSCs) that gain access to the articular environment should be collectively considered.

Numerous anabolic growth factors stimulate chondrocyte synthesis of proteoglycans, aggrecan and type II collagen, induce synoviocyte and MSC proliferation, drive chondrogenic differentiation of MSCs, and decrease the catabolic effects of cytokines such as interleukin-1 (IL-1) and the matrix metalloproteinases (MMP) (Table 1). In addition to being proanabolic and anticatabolic to restore cartilage in naturally occurring disease, an ideal growth factor for general cartilage tissue engineering or regeneration in OA would be effective regardless of the patient’s age or the presence of OA and would have no detrimental effects on either the cartilage or the synovial lining.

Table 1.

Summary of the effect of growth factors on chondrocytes/cartilage, synovium, and mesenchymal stem cells in vitro and in vivo

Growth factor Chondrocytes/cartilage Synovium Mesenchymal stem cell References
TGF-β1 Stimulates synthesis of ECM
Decreases catabolic activity of IL-1 and MMPs
Causes synovial proliferation and fibrosis
Induces chemotaxis of inflammatory leukocytes to synovium
Induction of osteophyte formation
Increases proliferation and ECM production
Downregulates collagen type 1 gene expression
7, 8, 27, 47, 89, 92
BMP-2 Stimulates synthesis of ECM
Partial reversal of dedifferentiated phenotype in OA
Increased ECM turnover (increased aggrecan degradation)
Presumed role in maturation of osteophytes
Multiple injections lead to synovial fibrosis
Stimulates synovial thickening in experimental OA
Increases proliferation and ECM production
Downregulates collagen type 1 gene expression
10, 27, 38, 47, 92
BMP-7 Stimulates ECM synthesis
Decreases cartilage degradation through decreasing activity/expression of numerous ILs and MMPs
Decreases expression of MMPs and aggrecanase
Does not appear to cause osteophyte formation or synovial fibrosis
Inhibits cell proliferation
Inconsistent ability to induce chondrogenesis alone
Potentiates chondrogenic differentiation with TGF-βs resulting in increased ECM synthesis and decreasing collagen type 1 compared with TGF-β alone
5, 40, 65, 81, 82
IGF-I Stimulates ECM synthesis
Decreases matrix catabolism except in aged and OA cartilage
Protective effect on synovium resulting in decreased thickening and decreased evidence of chronic inflammation Stimulates cell proliferation
Increases expression of ECM
Additive effect when combined with TGF-β
29, 52, 67, 96
FGF-2 Decreases aggrecanase activity
Antagonizes PG synthesis
Upregulates MMPs
Induces synovial proliferation
Inflammatory and induces osteophyte formation when used alone
Increases PG synthesis
Increases cell proliferation
24, 41, 64, 87
FGF-18 Increases chondrocyte proliferation and stimulates ECM in vitro and in injured joints but not in normal joints Induces synovial thickening
Enlargement of chondrophytes in experimental OA
24, 25, 66
PDGF No adverse effect in normal joints No adverse effect in normal joints Induces proliferation 43, 57

TGF-β1 = transforming growth factor-β1; BMP = bone morphogenetic protein; IGF-I = insulin growth factor I; FGF = fibroblast growth factor; PDGF = platelet-derived growth factor; ECM = extracellular matrix; IL = interleukin; MMP = matrix metalloproteinase; OA = osteoarthritis; PG = proteoglycan.

In addition to growth factors, the need for cells and a scaffold in a composite repair procedure is a widely accepted tenet. The relative magnitude, but not typically the direction, of the effect that a growth factor will have on cell proliferation or matrix synthesis could be altered by the type of cell or scaffold in the composite. In this review we focus on the growth factors that have promise in articular cartilage repair and not on the types of cells or biomaterials used for scaffolds as previously reviewed [19, 37, 52].

Historically, most growth factors have been evaluated on an independent basis, rather than in combination, to assess their effects on cartilage homeostasis in vitro or in vivo. Given the array and interactions of growth factors that are necessary for proper cartilage development and homeostasis, it is unlikely that any single growth factor will lead to complete cartilage repair or affect the arthritic milieu, but rather a combination approach will be required.

The purposes of our review were to first summarize the role of growth factors in articular cartilage repair and identify specific growth factors and combinations of growth factors that have the capacity to improve cartilage regeneration and second to explore the potential use of platelet-rich plasma, autologous-conditioned serum, and bone marrow concentrate preparations as methods of combined growth factor delivery in light of recent laboratory and clinical studies.

Search Strategies and Criteria

A PubMed search was performed using key words cartilage or chondrocyte alone and in combination with growth factor. The search was open for original manuscripts and review papers and open for all dates. From these searches we included papers that investigated the effects of growth factors on extracellular matrix synthesis and excluded those investigating molecular mechanisms of action.

Transforming Growth Factor-β Superfamily

Growth factors from the transforming growth factor-β (TGF-β) superfamily are structurally related and only active as homo- or heterodimers linked together with a single disulfide bond [96]. In cartilage repair, the three most thoroughly investigated members of the TGF-β superfamily include TGF-β1, bone morphogenetic protein-2 (BMP-2), and BMP-7 (also known as osteogenic protein-1 [OP-1]). More recently, in vitro studies of cartilage-derived morphogenetic protein (CDMP)-1 (also known as GDF-5; growth differentiation factor-5) and CDMP-2 have been performed and all show some capacity to stimulate cartilage matrix synthesis, but not all have been investigated in vivo and therefore will not be further reviewed as current potential treatments to enhance cartilage repair. In an elegant side-by-side comparison of the anabolic and anticatabolic activities of BMP-2, -4, -6, and -7 and CDMP-1 and -2, proteoglycan synthesis was stimulated to a greater extent by BMP-2, -4, and -7 with BMP-7 treatment resulting in maximal proteoglycan synthesis [18]. Furthermore, only BMP-7 showed consistent anticatabolic activity as indicated by restoration of proteoglycan synthesis after IL-1 treatment.

Transforming Growth Factor-β1 and -β3

TGF-β1 stimulates chondrocyte synthetic activity and decreases the catabolic activity of IL-1 [10]. In vitro, TGF-β1 stimulates chondrogenesis of synovial lining and bone marrow-derived MSCs [30, 50]. There have also been promising studies in rabbits in which TGF-β1 enhanced repair of cartilage defects [21]. However, in mouse and rabbit animal studies, numerous deleterious side effects of TGF-β1 supplementation have been reported, including stimulation of synovial proliferation and fibrosis, attraction of inflammatory leukocytes to the synovial lining, and induction of osteophyte formation [7, 10]. Given these serious safety concerns that are not components of other growth factor-based strategies, TGF-β1 therapy is not presently a viable option for use in the articular environment.

TGF-β3 also stimulated extracellular matrix (ECM) synthesis and has been evaluated in vitro in rabbit models of acute cartilage injury [29, 50, 51]. These studies were short term (8 weeks) and primarily looked at cartilage repair and not the synovial membrane and should be interpreted with caution given the potential for deleterious side effects of TGF-β1 in cartilage repair.

Bone Morphogenetic Protein-2

In vitro data indicate BMP-2 stimulates matrix synthesis and interestingly is capable of reversing chondrocyte dedifferentiation to some extent as indicated by an increase in synthesis of cartilage-specific collagen type IIB in dedifferentiated/OA chondrocytes [41]. The effect of BMP-2 on MSCs is similar to that of TGF-β1 with increased ECM production and decreased expression of collagen type 1 [30]. When injected into murine knees, chondrophytes induced by BMP-2 were found in the location where the growth plates meet the joint space [91]. In the rabbit trochlear groove, BMP-2-impregnated collagen sponges implanted into full-thickness cartilage defects enhance cartilage repair compared with empty defects or defects filled with collagen sponge alone. The enhanced repair remained evident 1 year after implantation [3]. In a mouse model of IL-1-induced cartilage degeneration, BMP-2 enhanced cartilage matrix turnover as evidenced by increased aggrecan degradation and increased collagen type II and aggrecan expression [11]. An increase in matrix turnover might indicate a reparative response after cartilage injury or OA, but it is unclear how an increase in a catabolic mechanism such as aggrecan degradation would be of benefit in a cartilage repair procedure.

Bone Morphogenetic Protein-7/Osteogenic Protein-1

BMP-7/OP-1 has been investigated for its capacity to regenerate articular cartilage and currently appears to be the gold standard growth factor for cartilage repair [16]. Like other anabolic growth factors, BMP-7 stimulates cartilage matrix synthesis and decreases catabolic activity of numerous catabolic cytokines, including IL-1, IL-6, IL-8, MMP-1, and MMP-13 [27]. However, unlike other chondrogenic growth factors, these effects of BMP-7 are not affected by age or OA [16]. BMP-7 is synthesized by chondrocytes and its gene and protein expression decrease with aging and cartilage degeneration, but degenerate cartilage is still able to respond to the anabolic cues of BMP-7 [17, 63]. How aged cartilage responds to the effects of BMP-7 is presently undefined. BMP-7 inhibits MSC proliferation but stimulates ECM synthesis in both synovial and bone marrow-derived MSCs [68, 84]. Chondrogenic differentiation of MSCs is enhanced when BMP-7 is in combination with TGF-β3.

BMP-7 acts synergistically with other anabolic growth factors such as insulin-like growth factor-I (IGF-I) [54]. In animal studies, BMP-7 appears effective in regeneration of osteochondral or focal chondral defects [16]. Administration of BMP-7 is also promising for the treatment of OA [43], but there are fewer animal model studies than there are for the treatment of focal cartilage lesions. In a rabbit ACL injury model, BMP-7 reduced cartilage degeneration [5] and decreased expression of MMPs in the cartilage and synovial membrane compared with untreated limbs [6]. Although BMP-7 is highly effective at stimulating bone repair, it does not appear to lead to osteophyte formation when administered into a joint nor does it stimulate uncontrolled fibroblast proliferation leading to joint fibrosis. In vitro studies indicate that the physiological range for BMP-7 anabolic activity is 50 to 200 ng/mL. Although the exact timing, frequency, or concentration of dosing for in vivo applications is not known, existing studies indicate that effective target concentrations can be reached in the synovial environment through direct injection or inclusion of BMP-7 in a composite scaffold. Collectively, the data suggest that BMP-7 therapy would lead to substantial clinical improvement in cartilage repair procedures; however, the addition of IGF-I might result in even greater healing potential.

Insulin-like Growth Factor-I

The role of IGF-I in articular cartilage metabolism has been extensively investigated in both health and disease [20, 61, 64, 73, 93]. When added exogenously to monolayer or explant cultures of normal articular cartilage from a variety of species, IGF-I induces a plethora of anabolic effects and decreases catabolic responses [78, 80, 90]. Chondrogenic differentiation of MSCs is induced by IGF-I but is enhanced when IGF-I and TGF-β1 are used in combination [56, 95].

The premise that IGF-I is required to maintain articular cartilage integrity is supported by an in vivo study in rats in which chronic IGF-I deficiency led to the development of articular cartilage lesions [24]. In animal models, IGF-I has led to enhanced repair of extensive cartilage defects and protection of the synovial membrane from chronic inflammation [32, 40]. However, there is a decreased capacity of chondrocytes to respond to IGF-I with age [4, 12, 31, 53, 55, 59] and in OA [23, 53, 55, 80]. Evidence suggests an uncoupling of IGF-I responsiveness in OA with IGF-I having the ability to robustly simulate matrix synthesis with the inability to decrease matrix catabolism [70].

Despite the diminished ability of IGF-I to decrease catabolism in aged and OA cartilage, studies suggest that a combination of IGF-I and BMP-7 results in greater repair potential than either growth factor alone [15, 54]. These studies demonstrated that in general, BMP-7 was more potent than IGF-I in stimulating matrix synthesis in aged and OA cells, but the greatest increase in matrix synthesis was observed after combination treatment with BMP-7 and IGF-I.

Fibroblast Growth Factor Family

Two members of the fibroblast growth factor (FGF) family have been investigated for their role in cartilage homeostasis, FGF-2 and FGF-18 [25]. In cartilage, FGF-2 (also known as basic FGF [bFGF]) is found in relative abundance in the pericellular matrix of cartilage [14]. On loading, FGF-2 becomes bound to cell surface receptors and activates anabolic pathways leading to decreased aggrecanase activity but no apparent change in proteoglycan content. In FGF-2 knockout mice, accelerated OA was noted in both unoperated mice and in those undergoing transection of the anterior horn of the medial meniscus. In the surgical OA group, subcutaneous administration of FGF-2 (1 μg every other day) suppressed the OA to a level not different from control mice. However, there is evidence suggesting that FGF-2 antagonizes proteoglycan synthesis mediated by IGF-I and/or OP-1, can upregulate MMPs, and results in synovial proliferation through protection from apoptosis [25]. Furthermore, animal models suggest intra-articular administration of FGF-2 results in inflammation and osteophyte formation [67] and does not aid in healing of cartilage defects [86]. A more recent rabbit study demonstrated improved healing of osteochondral lesions using a highly porous scaffold soaked in low-dose FGF-2 (10 μg/mL) compared with higher doses of FGF-2 (100 μg/mL) or scaffold without FGF-2, suggesting an inverse dose response [57]. FGF-2 treatment of bone marrow-derived MSC monolayer cultures results in enhanced proteoglycan synthesis and cell proliferation [88]. However, given the numerous potentially deleterious effects on the articular environment, the use of FGF-2 in cartilage repair is questionable.

Although there is less literature regarding the role of FGF-18 and cartilage homeostasis, it appears more promising than FGF-2 and elicits several anabolic effects on chondrocytes [25, 26]. In a rat model of rapid and severe OA, intra-articular administration of FGF-18 resulted in reduced cartilage degeneration scores with increased cartilage thickness of the tibial plateau, but also increased synovial thickness and chondrophyte formation [69]. The role of FGF-18 in repair of focal cartilage lesions is yet to be defined.

Platelet-derived Growth Factor

Platelet-derived growth factor (PDGF) exists as a homodimer (PDGF-AA or PDGF-BB) or a heterodimer (PDGF-AB). Evidence to support the use of PDGF in cartilage repair is extrapolated from the role of PDGF in wound healing or stimulation of matrix synthesis in growth plate chondrocytes [81]. In vivo, when injected into the knee of skeletally immature rats, no adverse effects were noted in the cartilage or synovial membrane [45]. Presently, the most commonly used form of PDGF is within the milieu of platelet-rich plasma (PRP) as discussed subsequently.

Growth Factors in Combination

In addition to those growth factors mentioned, there are several others that have been evaluated for their role in chondrogenesis and it could be concluded that these factors will therefore be important during cartilage regeneration or repair. These growth factors include vascular endothelial growth factor (VEGF), BMP-12, BMP-13, FGF-4, FGF-8, Wnt3a, and Wnt7a, but there are presently too little in vitro or in vivo data to fully discuss here for current applications in cartilage repair [36, 40, 75, 85]. Clearly numerous growth factors are needed to properly sequence chondrogenesis and it is likely that more than a single growth factor will be needed to achieve hyaline cartilage tissue in a reparative procedure. It is becoming increasingly clear that growth factors can work synergistically to enhance cartilage matrix synthesis as in the case of BMP-7 and IGF-I [54], and IGF-I, FGF-2, and TGF-β differentially regulate their own and each other’s gene expression and protein production in vitro [85]. Based on the concept that a combination of bioactive growth factors is likely necessary for cartilage repair, and the increasing application of autogenous biologics for tissue engineering, recent attention has been given to the use of PRP, autologous-conditioned serum (ACS), and bone marrow concentrate (BMC) in cartilage repair techniques. Another advantage of PRP and BMC is that on clotting, they form three-dimensional scaffolds to fill the cartilage defect and act as a guide for neochondrogenesis in situ.

Platelet-rich Plasma

PRP is defined by the American Red Cross as a sample of plasma with a twofold or more increase in platelet concentration above baseline levels or greater than 1.1 × 106 platelets/μL [65]. Presently, a number of different manufacturers have introduced systems for PRP preparation allowing for both intraoperative and outpatient use of PRP for a variety of orthopaedic pathologies [42]. There are classification schemes that categorize platelet concentrates based on relative concentrations of platelets, leucocytes, and fibrin, and although it is important to recognize and understand that there are obvious differences between types of platelet concentrates that are being used, the general term/abbreviation PRP is used herein [22, 42].

The concept that application of PRP would improve cartilage repair is based on the physiological role of platelets in wound healing [71]. In response to tissue injury, clots rich in platelets and fibrin form a scaffold for subsequent healing. There are over 1500 proteins within platelets and among them are growth factors stored in platelet α granules that are known to play important roles in the normal healing response, including PDGF, VEGF, TGF-β, FGF, and EGF [76, 83, 87]. Through modulation of the inflammatory response, promotion of local angiogenesis, attraction of fibroblasts and local stem cells to the site of injury, and an induction of autocrine growth factor production by uninjured adjacent cells, platelets and their products are instrumental in normal tissue repair and regeneration.

The application of PRP in cartilage repair is relatively new and therefore there are limited publications investigating its use. Chondrocytes and MSCs exposed to PRP both have increased cell proliferation and cartilage extracellular matrix synthesis of proteoglycans and collagen type II compared with controls [1, 66]. Synoviocytes from patients with OA cultured in PRP demonstrated increased hyaluronic acid production and secretion, suggesting that PRP could potentially serve as an endogenous source of chondroprotection and joint lubrication after intra-articular application [2]. In a rabbit model, osteochondral defects were treated with either autogenous PRP in a poly-lactic-glycolic acid (PLGA) carrier, PLGA alone, or left untreated [89]. The PRP group demonstrated a higher extent of cartilage regeneration as well as an increased production of the glycosaminoglycans in the ECM.

In a cohort of 30 patients comparing injections of PRP with hyaluronic acid (HA) in the management of OA, the success rate for the pain subscale reached 33.4% for the PRP group compared with 10% for the HA group (p = 0.004) [79]. Additionally, the percent reductions in the physical function subscale and overall WOMAC [9] at 5 weeks were also associated solely with treatment modality in favor of PRP with p = 0.043 and p = 0.010, respectively.

Kon et al. treated 115 knees of patients with four intra-articular PRP injections given every 21 days and followed the patients for 12 months [49]. Patients evaluated in this study included 58 with degenerative chondral lesions (Kellgren-Lawrence 0 [48]), 33 with early OA (Kellgren-Lawrence I–III), and 24 with advanced OA (Kellgren-Lawrence IV). A substantial improvement in International Knee Documentation Committee (IKDC) [47] and EuroQol (EQ-VAS) scores [28] was noted at the end of therapy and at both the 6- and 12-month time points. The IKDC subjective scores as well as the EQ-VAS score also demonstrated major improvements at the end of therapy. The authors concluded that treatment with PRP is safe and effective at improving pain, function, and quality of life in patients with degenerative articular pathology.

Autologous-conditioned Serum

Autologous-conditioned serum is generated by incubation of venous blood with glass beads, which results in increased concentration of growth factors such as PDGF and TGF and in some reports increased concentration of IL-1 receptor antagonist protein, which blocks the catabolic cytokine IL-1 [35]. In vivo studies in horses and humans support the use of ACS in the treatment of OA [8, 35]. However, a recent report assessing the in vitro effects of ACS on OA explant tissues did not support the use of ACS in the treatment of OA [77]. The authors found ACS contained increased levels of anti-inflammatory as well as proinflammatory cytokines, in particular TNF-alpha, but application of ACS to OA cartilage explant tissues did not have a net direct effect on cartilage metabolism. Improved appropriately powered clinical studies and increased length of followup should resolve the contradiction between current in vivo and in vitro data.

Bone Marrow Concentrate

BMC, like PRP, is generated through density gradient centrifugation of bone marrow aspirate. The advantage of BMC over PRP is that it contains MSCs, which have demonstrated benefits in the regeneration of cartilage and other tissues of the musculoskeletal system [13, 33, 92, 94]. MSCs represent only 0.001% to 0.01% of mononuclear cells of bone marrow aspirate [58, 72] and the concept of concentrating bone marrow aspirate to generate BMC is to increase the numbers of both platelets (and therefore growth factors) in addition to MSCs. Like PRP, BMC is a fully autogenous biologic that can be generated patient-side and forms a scaffold when clotted. Also, like PRP, BMC contains platelets and therefore is a rich source of growth factors, including PDGF and TGF-β [60, 82]. These growth factors are not only contained within the α granules of platelets, but they are also secreted by MSCs [62, 74] and can induce chondrogenesis of MSCs [13, 44, 46].

In an equine model of extensive (15-mm diameter) full-thickness cartilage defects, BMC resulted in improved cartilage repair compared with microfracture using both short-term arthroscopic inspection as well as in longer-term macroscopic, histologic, and quantitative MRI analyses (Fig. 1) [34]. Differences between BMC and microfracture observed arthroscopically at 12 weeks persisted at 8-month evaluation. In particular, repair tissue in BMC-treated defects was much better integrated into surrounding normal cartilage; the tissue was thicker and had a smoother surface. The results of this study indicate that BMC results in substantial improvements in both biochemical and structural components of neocartilage repair matrix. Clinically, BMC and PRP are simple to generate and easy to administer by needle injection to fill articular or osteochondral defects for tissue repair (Fig. 2). Clinical studies are needed to determine the ability of BMC grafting to relieve clinical symptoms and to determine the long-term durability of the repair tissue formed. Gobbi and Bathan have described a similar BMC procedure using a BMC paste in human cartilage defects, but full outcome analysis has not yet been performed on a large case series [38].

Fig. 1.

Fig. 1

Bone marrow concentrate (BMC) enhances articular cartilage regeneration. In an equine model of 15 mm diameter, full-thickness cartilage defects on the lateral trochlear ridge of the femur, BMC resulted in improved cartilage tissue formation at 8 months compared with microfracture treated defects.

Fig. 2A–C.

Fig. 2A–C

Clinical application of bone marrow concentrate (BMC) to treat a (A) large medial femoral condylar osteochondral cyst (arrows). Arthroscopic image (B) of the cyst after débridement and (C) transarthroscopic BMC administration under gas arthroscopy using an 18-gauge spinal needle.

Discussion

The appropriate treatment for cartilage injury remains a clinical challenge despite advances in surgical procedures and techniques. Bioactive growth factors are currently being considered as therapeutic possibilities to enhance healing of chondral injuries and modify early degenerative arthritis. The purposes of our review were therefore to first review the role of growth factors in articular cartilage repair and identify specific growth factors and combinations of growth factors that potentially improve cartilage regeneration and second to explore the potential use of PRP, ACS, and BMC preparations as methods of combined growth factor delivery in light of recent laboratory and clinical studies.

We acknowledge limitations of our review. First, we conducted a PubMed search using the broad search terms of cartilage or chondrocyte alone or in combination with growth factor. We did not use any other databases (eg, EMBASE, Google Scholar). This would necessarily limit the articles we identified. Second, we had no stringent criteria for selecting manuscripts among those identified. This might lead to potential bias in the articles selected. We emphasized inclusion of manuscripts related to preclinical or clinical applications of growth factors in articular cartilage with the aim to provide general recommendations to the clinician.

One potential method for improving clinical outcomes is to alter the local biologic environment where cartilage is damaged. Individual growth factors in animal models have demonstrated the ability to enhance cartilage production as well as decrease catabolic activity. To date, BMP-7 remains the gold standard growth factor with the ability to both decrease catabolic cytokines and stimulate cartilage matrix synthesis. IGF has also been studied in animal models but has demonstrated limited ability to affect chondrocytes in OA. The FGF and TGF-β families have demonstrated questionable ability to repair cartilage with additional deleterious effects on the articular environment. Currently, the most commonly used forms of combination growth factor therapy are PRP, ACS, and BMC.

There are currently limited publications on any single or combination of growth factors (PRP, ACS, or BMC) in the preclinical or clinical setting. Although the use of growth factors in treating both local cartilage defects as well as OA appears promising, clearly further research will be needed to help guide clinicians in this regard. The biggest obstacles are entrenched in intellectual property limiting the combination of recombinant growth factors, the arduous and resource-intense regulatory pathway, and the lack of any near-term solution for the reimbursement environment. Study design will inevitably require a truly randomized prospective trial in which patients and evaluators are blinded as to the treatment options in addition to a formal characterization of the therapeutic being used. Although these issues are not insurmountable, the most likely agents to succeed in this regard are likely to include the polygrowth factor environment provided by the PRP products.

Acknowledgments

We thank Ms Paula Sharp for her technical assistance in generating the manuscript.

Footnotes

Fortier, Cole are consultants for Arthrex, Inc.

Each author certifies that he or she has no commercial associations (eg, consultancies, stock ownership, equity interest, patent/licensing arrangements, etc) that might pose a conflict of interest in connection with the submitted article.

References

  • 1.Akeda K, An HS, Okuma M, Attawia M, Miyamoto K, Thonar EJ, Lenz ME, Sah RL, Masuda K. Platelet-rich plasma stimulates porcine articular chondrocyte proliferation and matrix biosynthesis. Osteoarthritis Cartilage. 2006;14:1272–1280. doi: 10.1016/j.joca.2006.05.008. [DOI] [PubMed] [Google Scholar]
  • 2.Anitua E, Sanchez M, Nurden AT, Zalduendo MM, la Fuente M, Azofra J, Andia I. Platelet-released growth factors enhance the secretion of hyaluronic acid and induce hepatocyte growth factor production by synovial fibroblasts from arthritic patients. Rheumatology (Oxford) 2007;46:1769–1772. doi: 10.1093/rheumatology/kem234. [DOI] [PubMed] [Google Scholar]
  • 3.Arai Y, Kubo T, Kobayashi K, Takeshita K, Takahashi K, Ikeda T, Imanishi J, Takigawa M, Hirasawa Y. Adenovirus vector-mediated gene transduction to chondrocytes: in vitro evaluation of therapeutic efficacy of transforming growth factor-B1 and heat-shock protein 70 gene transduction. J Rheumatol. 1997;24:1787–1795. [PubMed] [Google Scholar]
  • 4.Ashton IK, Matheson JA. Change in response with age of human articular cartilage to plasma somatomedin activity. Calcif Tiss Int. 1979;29:89–94. doi: 10.1007/BF02408062. [DOI] [PubMed] [Google Scholar]
  • 5.Badlani N, Inoue A, Healey R, Coutts R, Amiel D. The protective effect of OP-1 on articular cartilage in the development of osteoarthritis. Osteoarthritis Cartilage. 2008;16:600–606. doi: 10.1016/j.joca.2007.09.009. [DOI] [PubMed] [Google Scholar]
  • 6.Badlani N, Oshima Y, Healey R, Coutts R, Amiel D. Use of bone morphogenic protein-7 as a treatment for osteoarthritis. Clin Orthop Relat Res. 2009;467:3221–3229. doi: 10.1007/s11999-008-0569-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bakker AC, Loo FA, Beuningen HM, Sime P, Lent PL, Kraan PM, Richards CD, Berg WB. Overexpression of active TGF-beta-1 in the murine knee joint: evidence for synovial-layer-dependent chondro-osteophyte formation. Osteoarthritis Cartilage. 2001;9:128–136. doi: 10.1053/joca.2000.0368. [DOI] [PubMed] [Google Scholar]
  • 8.Baltzer AW, Moser C, Jansen SA, Krauspe R. Autologous conditioned serum (Orthokine) is an effective treatment for knee osteoarthritis. Osteoarthritis Cartilage. 2009;17:152–160. doi: 10.1016/j.joca.2008.06.014. [DOI] [PubMed] [Google Scholar]
  • 9.Bellamy N, Buchanan WW, Goldsmith CH, Campbell J, Stitt L. Validation Study of WOMAC: a health status instrument for measuring clinically-important-patient-relevant outcomes following total hip or knee arthroplasty in osteoarthritis. J Orthop Rheumatol. 1988;1:95–108. [PubMed] [Google Scholar]
  • 10.Blaney Davidson EN, Kraan PM, Berg WB. TGF-beta and osteoarthritis. Osteoarthritis Cartilage. 2007;15:597–604. doi: 10.1016/j.joca.2007.02.005. [DOI] [PubMed] [Google Scholar]
  • 11.Blaney Davidson EN, Vitters EL, Lent PL, Loo FA, Berg WB, Kraan PM. Elevated extracellular matrix production and degradation upon bone morphogenetic protein-2 (BMP-2) stimulation point toward a role for BMP-2 in cartilage repair and remodeling. Arthritis Res Ther. 2007;9:R102. doi: 10.1186/ar2305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Boehm AK, Seth M, Mayr KG, Fortier LA. Hsp90 mediates insulin-like growth factor 1 and interleukin-1beta signaling in an age-dependent manner in equine articular chondrocytes. Arthritis Rheum. 2007;56:2335–2343. doi: 10.1002/art.22664. [DOI] [PubMed] [Google Scholar]
  • 13.Chen FH, Tuan RS. Mesenchymal stem cells in arthritic diseases. Arthritis Res Ther. 2008;10:223–232. doi: 10.1186/ar2514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Chia SL, Sawaji Y, Burleigh A, McLean C, Inglis J, Saklatvala J, Vincent T. Fibroblast growth factor 2 is an intrinsic chondroprotective agent that suppresses ADAMTS-5 and delays cartilage degradation in murine osteoarthritis. Arthritis Rheum. 2009;60:2019–2027. doi: 10.1002/art.24654. [DOI] [PubMed] [Google Scholar]
  • 15.Chubinskaya S, Hakimiyan A, Pacione C, Yanke A, Rappoport L, Aigner T, Rueger DC, Loeser RF. Synergistic effect of IGF-1 and OP-1 on matrix formation by normal and OA chondrocytes cultured in alginate beads. Osteoarthritis Cartilage. 2007;15:421–430. doi: 10.1016/j.joca.2006.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chubinskaya S, Hurtig M, Rueger DC. OP-1/BMP-7 in cartilage repair. Int Orthop. 2007;31:773–781. doi: 10.1007/s00264-007-0423-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chubinskaya S, Kumar B, Merrihew C, Heretis K, Rueger DC, Kuettner KE. Age-related changes in cartilage endogenous osteogenic protein-1 (OP-1) Biochim Biophys Acta. 2002;1588:126–134. doi: 10.1016/s0925-4439(02)00158-8. [DOI] [PubMed] [Google Scholar]
  • 18.Chubinskaya S, Segalite D, Pikovsky D, Hakimiyan AA, Rueger DC. Effects induced by BMPS in cultures of human articular chondrocytes: comparative studies. Growth Factors. 2008;26:275–283. doi: 10.1080/08977190802291733. [DOI] [PubMed] [Google Scholar]
  • 19.Chung C, Burdick JA. Engineering cartilage tissue. Adv Drug Deliv Rev. 2008;60:243–262. doi: 10.1016/j.addr.2007.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Denko CW, Boja B, Moskowitz RW. Growth promoting peptides in osteoarthritis and diffuse idiopathic skeletal hyperostosis—insulin, insulin-like growth factor-I, growth hormone. J Rheumatol. 1994;21:1725–1730. [PubMed] [Google Scholar]
  • 21.Diao H, Wang J, Shen C, Xia S, Guo T, Dong L, Zhang C, Chen J, Zhao J, Zhang J. Improved cartilage regeneration utilizing mesenchymal stem cells in TGF-beta1 gene-activated scaffolds. Tissue Eng Part A. 2009;15:2687–2698. doi: 10.1089/ten.tea.2008.0621. [DOI] [PubMed] [Google Scholar]
  • 22.Dohan Ehrenfest DM, Rasmusson L, Albrektsson T. Classification of platelet concentrates: from pure platelet-rich plasma (P-PRP) to leucocyte- and platelet-rich fibrin (L-PRF) Trends Biotechnol. 2009;27:158–167. doi: 10.1016/j.tibtech.2008.11.009. [DOI] [PubMed] [Google Scholar]
  • 23.Dore S, Pelletier J, DiBattista JA, Tardif G, Brazeau P, Martel-Pelletier J. Human osteoarthritic chondrocytes possess an increased number of insulin-like growth factor 1 binding sites but are unresponsive to its stimulation: possible role of IGF-1 binding proteins. Arthritis Rheum. 1994;37:253–263. doi: 10.1002/art.1780370215. [DOI] [PubMed] [Google Scholar]
  • 24.Ekenstedt KJ, Sonntag WE, Loeser RF, Lindgren BR, Carlson CS. Effects of chronic growth hormone and insulin-like growth factor 1 deficiency on osteoarthritis severity in rat knee joints. Arthritis Rheum. 2006;54:3850–3858. doi: 10.1002/art.22254. [DOI] [PubMed] [Google Scholar]
  • 25.Ellman MB, An HS, Muddasani P, Im HJ. Biological impact of the fibroblast growth factor family on articular cartilage and intervertebral disc homeostasis. Gene. 2008;420:82–89. doi: 10.1016/j.gene.2008.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ellsworth JL, Berry J, Bukowski T, Claus J, Feldhaus A, Holderman S, Holdren MS, Lum KD, Moore EE, Raymond F, Ren H, Shea P, Sprecher C, Storey H, Thompson DL, Waggie K, Yao L, Fernandez RJ, Eyre DR, Hughes SD. Fibroblast growth factor-18 is a trophic factor for mature chondrocytes and their progenitors. Osteoarthritis Cartilage. 2002;10:308–320. doi: 10.1053/joca.2002.0514. [DOI] [PubMed] [Google Scholar]
  • 27.Elshaier AM, Hakimiyan AA, Rappoport L, Rueger DC, Chubinskaya S. Effect of interleukin-1beta on osteogenic protein 1-induced signaling in adult human articular chondrocytes. Arthritis Rheum. 2009;60:143–154. doi: 10.1002/art.24151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.EuroQol Group. EuroQol—A New Facility for the Measurement of Health-related Quality of Life. Health Policy. The EuroQol Group. York, UK: Center for Health Economics, University of York; 1990;16:199–208. [DOI] [PubMed]
  • 29.Fan H, Tao H, Wu Y, Hu Y, Yan Y, Luo Z. TGF-β3 immobilized PLGA-gelatin/chondroitin sulfate/hyaluronic acid hybrid scaffold for cartilage regeneration. J Biomed Mater Res A. 2010;95:982–992. doi: 10.1002/jbm.a.32899. [DOI] [PubMed] [Google Scholar]
  • 30.Fan J, Gong Y, Ren L, Varshney RR, Cai D, Wang DA. In vitro engineered cartilage using synovium-derived mesenchymal stem cells with injectable gellan hydrogels. Acta Biomater. 2010;6:1178–1185. doi: 10.1016/j.actbio.2009.08.042. [DOI] [PubMed] [Google Scholar]
  • 31.Fortier LA, Miller BJ. Signaling through the small G-protein Cdc42 is involved in insulin-like growth factor-I resistance in aging articular chondrocytes. J Orthop Res. 2006;24:1765–1772. doi: 10.1002/jor.20185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Fortier LA, Mohammed HO, Lust G, Nixon AJ. Insulin-like growth factor-I enhances cell-based repair of articular cartilage. J Bone Joint Surg Br. 2002;84:276–288. doi: 10.1302/0301-620X.84B2.11167. [DOI] [PubMed] [Google Scholar]
  • 33.Fortier LA, Nixon AJ, Williams J, Cable CS. Isolation and chondrocytic differentiation of equine bone marrow-derived mesenchymal stem cells. Am J Vet Res. 1998;59:1182–1187. [PubMed] [Google Scholar]
  • 34.Fortier LA, Potter HG, Rickey EJ, Schnabel LV, Foo LF, Chong LR, Stokol T, Cheetham J, Nixon AJ. Concentrated bone marrow aspirate improves full-thickness cartilage repair compared to microfracture in an equine model of extensive cartilage loss. J Bone Joint Surg Am. 2010;92:1927–1937. doi: 10.2106/JBJS.I.01284. [DOI] [PubMed] [Google Scholar]
  • 35.Frisbie DD, Kawcak CE, Werpy NM, Park RD, McIlwraith CW. Clinical, biochemical, and histologic effects of intra-articular administration of autologous conditioned serum in horses with experimentally induced osteoarthritis. Am J Vet Res. 2007;68:290–296. doi: 10.2460/ajvr.68.3.290. [DOI] [PubMed] [Google Scholar]
  • 36.Gaissmaier C, Koh JL, Weise K. Growth and differentiation factors for cartilage healing and repair. Injury. 2008;39:S88–S96. doi: 10.1016/j.injury.2008.01.035. [DOI] [PubMed] [Google Scholar]
  • 37.Getgood A, Brooks R, Fortier L, Rushton N. Articular cartilage tissue engineering: today’s research, tomorrow’s practice? J Bone Joint Surg Br. 2009;91:565–576. doi: 10.1302/0301-620X.91B5.21832. [DOI] [PubMed] [Google Scholar]
  • 38.Gobbi A, Bathan L. Biological approaches for cartilage repair. J Knee Surg. 2009;22:36–44. doi: 10.1055/s-0030-1247726. [DOI] [PubMed] [Google Scholar]
  • 39.Goldring MB, Tsuchimochi K, Ijiri K. The control of chondrogenesis. J Cell Biochem. 2006;97:33–44. doi: 10.1002/jcb.20652. [DOI] [PubMed] [Google Scholar]
  • 40.Goodrich LR, Hidaka C, Robbins PD, Evans CH, Nixon AJ. Genetic modification of chondrocytes with insulin-like growth factor-1 enhances cartilage healing in an equine model. J Bone Joint Surg Br. 2007;89:672–685. doi: 10.1302/0301-620X.89B5.18343. [DOI] [PubMed] [Google Scholar]
  • 41.Gouttenoire J, Valcourt U, Ronziere MC, Aubert-Foucher E, Mallein-Gerin F, Herbage D. Modulation of collagen synthesis in normal and osteoarthritic cartilage. Biorheology. 2004;41:535–542. [PubMed] [Google Scholar]
  • 42.Hall MP, Band PA, Meislin RJ, Jazrawi LM, Cardone DA. Platelet-rich plasma: current concepts and application in sports medicine. J Am Acad Orthop Surg. 2009;17:602–608. doi: 10.5435/00124635-200910000-00002. [DOI] [PubMed] [Google Scholar]
  • 43.Hayashi M, Muneta T, Ju YJ, Mochizuki T, Sekiya I. Weekly intra-articular injections of bone morphogenetic protein-7 inhibits osteoarthritis progression. Arthritis Res Ther. 2008;10:R118. doi: 10.1186/ar2521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Huang AH, Motlekar NA, Stein A, Diamond SL, Shore EM, Mauck RL. High-throughput screening for modulators of mesenchymal stem cell chondrogenesis. Ann Biomed Eng. 2008;36:1909–1921. doi: 10.1007/s10439-008-9562-4. [DOI] [PubMed] [Google Scholar]
  • 45.Hulth A, Johnell O, Miyazono K, Lindberg L, Heinegard D, Heldin C-H. Effect of transforming growth factor-B and platelet-derived growth factor-BB on articular cartilage in rats. J Orthop Res. 1996;14:547–553. doi: 10.1002/jor.1100140408. [DOI] [PubMed] [Google Scholar]
  • 46.Indrawattana N, Chen G, Tadokoro M, Shann LH, Ohgushi H, Tateishi T, Tanaka J, Bunyaratvej A. Growth factor combination for chondrogenic induction from human mesenchymal stem cell. Biochem Biophys Res Commun. 2004;320:914–919. doi: 10.1016/j.bbrc.2004.06.029. [DOI] [PubMed] [Google Scholar]
  • 47.Irrgang JJ, Anderson AF, Boland AL, Harner CD, Kurosaka M, Neyret P, Richmond JC, Shelborne KD. Development and validation of the International Knee Documentation Committee subjective knee form. Am J Sports Med. 2001;29:600–613. doi: 10.1177/03635465010290051301. [DOI] [PubMed] [Google Scholar]
  • 48.Kellgren JH, Lawrence JS. Radiological assessment of osteo-arthrosis. Ann Rheum Dis. 1957;16:494–502. doi: 10.1136/ard.16.4.494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kon E, Buda R, Filardo G, Di Martino A, Timoncini A, Cenacchi A, Fornasari PM, Giannini S, Marcacci M. Platelet-rich plasma: intra-articular knee injections produced favorable results on degenerative cartilage lesions. Knee Surg Sports Traumatol Arthrosc. 2010;18:472–479. doi: 10.1007/s00167-009-0940-8. [DOI] [PubMed] [Google Scholar]
  • 50.Kurth T, Hedbom E, Shintani N, Sugimoto M, Chen FH, Haspl M, Martinovic S, Hunziker EB. Chondrogenic potential of human synovial mesenchymal stem cells in alginate. Osteoarthritis Cartilage. 2007;15:1178–1189. doi: 10.1016/j.joca.2007.03.015. [DOI] [PubMed] [Google Scholar]
  • 51.Lee CH, Cook JL, Mendelson A, Moioli EK, Yao H, Mao JJ. Regeneration of the articular surface of the rabbit synovial joint by cell homing: a proof of concept study. Lancet. 2010;376:440–448. doi: 10.1016/S0140-6736(10)60668-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lee SH, Shin H. Matrices and scaffolds for delivery of bioactive molecules in bone and cartilage tissue engineering. Adv Drug Deliv Rev. 2007;59:339–359. doi: 10.1016/j.addr.2007.03.016. [DOI] [PubMed] [Google Scholar]
  • 53.Loeser RF, Carlson CS, Del Carlo M, Cole A. Detection of nitrotyrosine in aging and osteoarthritic cartilage: correlation of oxidative damage with the presence of interleukin-1beta and with chondrocyte resistance to insulin-like growth factor 1. Arthritis Rheum. 2002;46:2349–2357. doi: 10.1002/art.10496. [DOI] [PubMed] [Google Scholar]
  • 54.Loeser RF, Pacione CA, Chubinskaya S. The combination of insulin-like growth factor 1 and osteogenic protein 1 promotes increased survival of and matrix synthesis by normal and osteoarthritic human articular chondrocytes. Arthritis Rheum. 2003;48:2188–2196. doi: 10.1002/art.11209. [DOI] [PubMed] [Google Scholar]
  • 55.Loeser RF, Shanker G, Carlson CS, Gardin JF, Shelton BJ, Sonntag WE. Reduction in the chondrocyte response to insulin-like growth factor 1 in aging and osteoarthritis: studies in a non-human primate model of naturally occurring disease. Arthritis Rheum. 2000;43:2110–2120. doi: 10.1002/1529-0131(200009)43:9<2110::AID-ANR23>3.0.CO;2-U. [DOI] [PubMed] [Google Scholar]
  • 56.Longobardi L, O’Rear L, Aakula S, Johnstone B, Shimer K, Chytil A, Horton WA, Moses HL, Spagnoli A. Effect of IGF-I in the chondrogenesis of bone marrow mesenchymal stem cells in the presence or absence of TGF-beta signaling. J Bone Miner Res. 2006;21:626–636. doi: 10.1359/jbmr.051213. [DOI] [PubMed] [Google Scholar]
  • 57.Maehara H, Sotome S, Yoshii T, Torigoe I, Kawasaki Y, Sugata Y, Yuasa M, Hirano M, Mochizuki N, Kikuchi M, Shinomiya K, Okawa A. Repair of large osteochondral defects in rabbits using porous hydroxyapatite/collagen (HAp/Col) and fibroblast growth factor-2 (FGF-2) J Orthop Res. 2010;28:677–686. doi: 10.1002/jor.21032. [DOI] [PubMed] [Google Scholar]
  • 58.Martin DR, Cox NR, Hathcock TL, Niemeyer GP, Baker HJ. Isolation and characterization of multipotential mesenchymal stem cells from feline bone marrow. Exp Hematol. 2002;30:879–886. doi: 10.1016/S0301-472X(02)00864-0. [DOI] [PubMed] [Google Scholar]
  • 59.Martin JS, Ellerbrock SM, Buckwalter JA. Age-related decline in chondrocyte response to insulin-like growth factor-I: the role of growth factor binding proteins. J Orthop Res. 1997;15:491–498. doi: 10.1002/jor.1100150403. [DOI] [PubMed] [Google Scholar]
  • 60.McCarrel T, Fortier L. Temporal growth factor release from platelet-rich plasma, trehalose lyophilized platelets, and bone marrow aspirate and their effect on tendon and ligament gene expression. J Orthop Res. 2009;27:1033–1042. doi: 10.1002/jor.20853. [DOI] [PubMed] [Google Scholar]
  • 61.McQuillan DJ, Handley CJ, Campbell MA, Bolis S, Milway VE, Herington AC. Stimulation of proteoglycan biosynthesis by serum and insulin-like growth factor-1 in cultured bovine articular cartilage. Biochem J. 1986;240:423–430. doi: 10.1042/bj2400423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Mehta S, Watson JT. Platelet rich concentrate: basic science and current clinical applications. J Orthop Trauma. 2008;22:432–438. doi: 10.1097/BOT.0b013e31817e793f. [DOI] [PubMed] [Google Scholar]
  • 63.Merrihew C, Kumar B, Heretis K, Rueger DC, Kuettner KE, Chubinskaya S. Alterations in endogenous osteogenic protein-1 with degeneration of human articular cartilage. J Orthop Res. 2003;21:899–907. doi: 10.1016/S0736-0266(03)00055-X. [DOI] [PubMed] [Google Scholar]
  • 64.Middleton J, Manthey A, Tyler J. Insulin-like growth factor (IGF) receptor, IGF-I, interleukin-1b (IL-1b), and IL-6 mRNA expression in osteoarthritic and normal human cartilage. J Histochem Cytochem. 1996;44:133–141. doi: 10.1177/44.2.8609369. [DOI] [PubMed] [Google Scholar]
  • 65.Miller Y, Bachowski G, Benjamin R, Eklund DK, Hibbard AJ, Lightfoot T. Practice Guidelines for Blood Transfusion: A Compilation From Recent Peer-reviewed Literature. 2nd ed. Washington, DC: American Red Cross; 2007. p. 56. [Google Scholar]
  • 66.Mishra A, Tummala P, King A, Lee B, Kraus M, Tse V, Jacobs CR. Buffered platelet-rich plasma enhances mesenchymal stem cell proliferation and chondrogenic differentiation. Tissue Eng Part C Methods. 2009;15:431–435. doi: 10.1089/ten.tec.2008.0534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Miyakoshi N, Kobayashi M, Nozaka K, Okada K, Shimada Y, Itoi E. Effects of intraarticular administration of basic fibroblast growth factor with hyaluronic acid on osteochondral defects of the knee in rabbits. Arch Orthop Trauma Surg. 2005;125:683–692. doi: 10.1007/s00402-005-0052-y. [DOI] [PubMed] [Google Scholar]
  • 68.Miyamoto C, Matsumoto T, Sakimura K, Shindo H. Osteogenic protein-1 with transforming growth factor-beta1: potent inducer of chondrogenesis of synovial mesenchymal stem cells in vitro. J Orthop Sci. 2007;12:555–561. doi: 10.1007/s00776-007-1176-4. [DOI] [PubMed] [Google Scholar]
  • 69.Moore EE, Bendele AM, Thompson DL, Littau A, Waggie KS, Reardon B, Ellsworth JL. Fibroblast growth factor-18 stimulates chondrogenesis and cartilage repair in a rat model of injury-induced osteoarthritis. Osteoarthritis Cartilage. 2005;13:623–631. doi: 10.1016/j.joca.2005.03.003. [DOI] [PubMed] [Google Scholar]
  • 70.Morales TI. The quantitative and functional relation between insulin-like growth factor-I (IGF) and IGF-binding proteins during human osteoarthritis. J Orthop Res. 2008;26:465–474. doi: 10.1002/jor.20549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Nurden AT, Nurden P, Sanchez M, Andia I, Anitua E. Platelets and wound healing. Front Biosci. 2008;13:3532–3548. doi: 10.2741/2947. [DOI] [PubMed] [Google Scholar]
  • 72.Pittenger MF, Mackay AM, Beck SC, Jaiswal RK. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–147. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
  • 73.Posever J, Phillips FM, Pottenger LA. Effects of basic fibroblast growth factor, transforming growth factor-B1, insulin-like growth factor-1, and insulin on human osteoarthritic articular cartilage explants. J Orthop Res. 1995;13:832–837. doi: 10.1002/jor.1100130605. [DOI] [PubMed] [Google Scholar]
  • 74.Potier E, Ferreira E, Dennler S, Mauviel A, Oudina K, Logeart-Avramoglou D, Petite H. Desferrioxamine-driven upregulation of angiogenic factor expression by human bone marrow stromal cells. J Tissue Eng Regen Med. 2008;2:272–278. doi: 10.1002/term.92. [DOI] [PubMed] [Google Scholar]
  • 75.Quintana L, zur Nieden NI, Semino CE. Morphogenetic and regulatory mechanisms during developmental chondrogenesis: new paradigms for cartilage tissue engineering. Tissue Eng Part B Rev. 2009;15:29–41. doi: 10.1089/ten.teb.2008.0329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Qureshi AH, Chaoji V, Maiguel D, Faridi MH, Barth CJ, Salem SM, Singhal M, Stoub D, Krastins B, Ogihara M, Zaki MJ, Gupta V. Proteomic and phospho-proteomic profile of human platelets in basal, resting state: insights into integrin signaling. PLoS One. 2009;4:e7627. doi: 10.1371/journal.pone.0007627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rutgers M, Saris DB, Dhert WJ, Creemers LB. Cytokine profile of autologous conditioned serum for treatment of osteoarthritis, in vitro effects on cartilage metabolism and intra-articular levels after injection. Arthritis Res Ther. 2010;12:R114. doi: 10.1186/ar3050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Sah RL, Chen AC, Grodzinsky AJ, Trippel SB. Differential effects of bFGF and IGF-I on matrix metabolism in calf and adult bovine cartilage explants. Archives Biochem Biophys. 1994;308:137–147. doi: 10.1006/abbi.1994.1020. [DOI] [PubMed] [Google Scholar]
  • 79.Sanchez M, Anitua E, Azofra J, Aguirre JJ, Andia I. Intra-articular injection of an autologous preparation rich in growth factors for the treatment of knee OA: a retrospective cohort study. Clin Exp Rheumatol. 2008;26:910–913. [PubMed] [Google Scholar]
  • 80.Schalkwijk J, Joosten LAB, Berg WB, Putte LBA. Chondrocyte nonresponsiveness to insulin-like growth factor 1 in experimental arthritis. Arthritis Rheum. 1989;32:894–900. doi: 10.1002/anr.1780320111. [DOI] [PubMed] [Google Scholar]
  • 81.Schmidt MB, Chen EH, Lynch SE. A review of the effects of insulin-like growth factor and platelet derived growth factor on in vivo cartilage healing and repair. Osteoarthritis Cartilage. 2006;14:403–412. doi: 10.1016/j.joca.2005.10.011. [DOI] [PubMed] [Google Scholar]
  • 82.Schnabel LV, Mohammed HO, Miller BJ, McDermott WG, Jacobson MS, Santangelo KS, Fortier LA. Platelet rich plasma (PRP) enhances anabolic gene expression patterns in flexor digitorum superficialis tendons. J Orthop Res. 2007;25:230–240. doi: 10.1002/jor.20278. [DOI] [PubMed] [Google Scholar]
  • 83.Senzel L, Gnatenko DV, Bahou WF. The platelet proteome. Curr Opin Hematol. 2009;16:329–333. doi: 10.1097/MOH.0b013e32832e9dc6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Shen B, Wei A, Whittaker S, Williams LA, Tao H, Ma DD, Diwan AD. The role of BMP-7 in chondrogenic and osteogenic differentiation of human bone marrow multipotent mesenchymal stromal cells in vitro. J Cell Biochem. 2010;109:406–416. doi: 10.1002/jcb.22412. [DOI] [PubMed] [Google Scholar]
  • 85.Shi S, Mercer S, Eckert GJ, Trippel SB. Growth factor regulation of growth factors in articular chondrocytes. J Biol Chem. 2009;284:6697–6704. doi: 10.1074/jbc.M807859200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Siebert CH, Miltner O, Weber M, Sopka S, Koch S, Niedhart C. Healing of osteochondral grafts in an ovine model under the influence of bFGF. Arthroscopy. 2003;19:182–187. doi: 10.1053/jars.2003.50000. [DOI] [PubMed] [Google Scholar]
  • 87.Smyth SS, McEver RP, Weyrich AS, Morrell CN, Hoffman MR, Arepally GM, French PA, Dauerman HL. Becker RC; 2009 Platelet Colloquium Participants. Platelet functions beyond hemostasis. J Thromb Haemost. 2009;7:1759–1766. doi: 10.1111/j.1538-7836.2009.03586.x. [DOI] [PubMed] [Google Scholar]
  • 88.Stewart AA, Byron CR, Pondenis H, Stewart MC. Effect of fibroblast growth factor-2 on equine mesenchymal stem cell monolayer expansion and chondrogenesis. Am J Vet Res. 2007;68:941–945. doi: 10.2460/ajvr.68.9.941. [DOI] [PubMed] [Google Scholar]
  • 89.Sun Y, Feng Y, Zhang CQ, Chen SB, Cheng XG. The regenerative effect of platelet-rich plasma on healing in large osteochondral defects. Int Orthop. 2010;34:589–597. doi: 10.1007/s00264-009-0793-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Tyler JA. Insulin-like growth factor 1 can decrease degradation and promote synthesis of proteoglycan in cartilage exposed to cytokines. Biochem J. 1989;260:543–548. doi: 10.1042/bj2600543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Beuningen HM, Glansbeek HL, Kraan PM, Berg WB. Differential effects of local application of BMP-2 or TGF-beta 1 on both articular cartilage composition and osteophyte formation. Osteoarthritis Cartilage. 1998;6:306–317. doi: 10.1053/joca.1998.0129. [DOI] [PubMed] [Google Scholar]
  • 92.Wakitani S, Saito T, Caplan AI. Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve. 1995;18:1417–1426. doi: 10.1002/mus.880181212. [DOI] [PubMed] [Google Scholar]
  • 93.Wang E, Wang J, Chin E, Zhou J, Bondy CA. Cellular patterns of insulin-like growth factor system gene expression in murine chondrogenesis and osteogenesis. Endocrinology. 1995;136:2741–2751. doi: 10.1210/en.136.6.2741. [DOI] [PubMed] [Google Scholar]
  • 94.Wilke MM, Nydam DV, Nixon AJ. Enhanced early chondrogenesis in articular defects following arthroscopic mesenchymal stem cell implantation in an equine model. J Orthop Res. 2007;25:913–925. doi: 10.1002/jor.20382. [DOI] [PubMed] [Google Scholar]
  • 95.Worster AA, Brower-Toland BD, Fortier LA, Bent SJ, Williams J, Nixon AJ. Chondrocytic differentiation of mesenchymal stem cells sequentially exposed to transforming growth factor-beta1 in monolayer and insulin-like growth factor-I in a three-dimensional matrix. J Orthop Res. 2001;19:738–749. doi: 10.1016/S0736-0266(00)00054-1. [DOI] [PubMed] [Google Scholar]
  • 96.Wu MY, Hill CS. TGF-beta superfamily signaling in embryonic development and homeostasis. Dev Cell. 2009;16:329–343. doi: 10.1016/j.devcel.2009.02.012. [DOI] [PubMed] [Google Scholar]

Articles from Clinical Orthopaedics and Related Research are provided here courtesy of The Association of Bone and Joint Surgeons

RESOURCES