Skip to main content

Effects and action mechanisms of individual cytokines contained in PRP on osteoarthritis

Abstract

Osteoarthritis (OA) is defined as a degenerative joint disease that can affect all tissues of the joint, including the articular cartilage, subchondral bone, ligaments capsule, and synovial membrane. The conventional nonoperative treatments are ineffective for cartilage repair and induce only symptomatic relief. Platelet-rich plasma (PRP) is a platelet concentrate derived from autologous whole blood with a high concentration of platelets, which can exert anti-inflammatory and regenerative effects by releasing multiple growth factors and cytokines. Recent studies have shown that PRP exhibits clinical benefits in patients with OA. However, high operational and equipment requirements greatly limit the application of PRP to OA treatment. Past studies have indicated that high-concentration PRP growth factors and cytokines may be applied as a commercial replacement for PRP. We reviewed the relevant articles to summarize the feasibility and mechanisms of PRP-based growth factors in OA. The available evidence suggests that transforming growth factor-α and β, platelet-derived growth factors, epidermal growth factor, insulin-like growth factor-1, and connective tissue growth factors might benefit OA, while vascular endothelial growth factor, tumor necrosis factor-α, angiopoietin-1, and stromal cell derived factor-1α might induce negative effects on OA. The effects of fibroblast growth factor, hepatocyte growth factor, platelet factor 4, and keratinocyte growth factor on OA remain uncertain. Thus, it can be concluded that not all cytokines released by PRP are beneficial, although the therapeutic action of PRP has a valuable potential to improve.

Background

Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage degradation, osteophyte formation, and synovial inflammation [1]. This disease can affect all tissues in the joint, including the articular cartilage, subchondral bone, the ligament capsule, and the synovial membrane, leading to joint failure [2]. The conventional nonsurgical treatments for OA, such as hyaluronan, corticosteroids, nonsteroidal anti-inflammatory drugs, IL-4 and IL-1 receptor antagonists, paracetamol, and chondroitin sulfate products, are mainly focused on relieving pain and improving joint functions [3,4,5,6]; these treatments are ineffective for cartilage repair and induce only symptomatic relief. An increasing number of novel treatments have been developed recently, including platelet-rich plasma (PRP), vitamin D, oral collagens, methylsulfonylmethane, and curcumin [7,8,9,10]. However, not all of these treatments have relatively credible guidelines.

PRP is a platelet concentrate derived from autologous whole blood, that has a high concentration of platelets, containing 3–6 fold platelets compared to that in whole blood [11,12,13]. PRP exerts anti-inflammatory and regenerative effects, which are mediated through the release of multiple growth factors and cytokines, including platelet-derived growth factors (PDGF), transforming growth factor (TGF), vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), connective tissue growth factor (CTGF), insulin-like growth factor (IGF), hepatocyte growth factor (HGF), keratinocyte growth factor (KGF), angiopoietin-1 (Ang-1), platelet factor 4 (PF4), stromal cell derived factor (SDF), and tumor necrosis factor (TNF) [14]. Currently, studies have demonstrated that PRP exhibits clinical benefits in patients with musculoskeletal disorders [15,16,17]. Moreover, systematic reviews and meta-analyses have concluded that OA patients can benefit from intra-articular administration of PRP [18,19,20]. Although PRP has been widely used in OA due to its regenerative potential, its efficacy remains debatable due to individual differences and different preparations, which lead to varied therapeutic effects. In particular, the different preparations of PRP may account for different effects on treating diseases in different studies.

As described previously, the high demand of operation and equipment limits the application of PRP [21]. If a patient receives PRP treatment, the patient’s blood is first drawn and centrifuged to collect the platelets. As a result, PRP is difficult to repeatedly administer and develop as a commercial drug for easy application. However, studies have indicated that high concentrations of growth factors and cytokines in PRP may be used as a commercial replacement for PRP [21, 22]. Many PRP-based growth factors and platelet cytokines have been examined in cells through in vitro and in vivo animal experiments to determine their effects and mechanisms in OA. We reviewed published articles and summarized the feasibility and mechanisms of PRP-based growth factors in OA.

Cytokines that are beneficial for OA treatment (Fig. 1)

Fig. 1
figure 1

Mechanisms of cytokines in PRP that have positive effects on OA treatment

TGF-β

TGF-β plays an important role in tissue repair and regeneration [23]. The TGF-β superfamily includes over 30 members, which can be divided into two ligand subfamilies [24]. These subfamilies, which are defined by sequence similarity and the activation of SMAD proteins, include the TGF-β/activin/nodal subfamily and the bone morphogenetic protein/growth and differentiation factor/Muellerian inhibiting substance subfamily [24]. TGF-β binds to TGF-β receptor type II and exerts its effects by bringing together pairs of type I and type II receptors on the cell surface [24]. TGF-β receptors are serine/threonine kinases, but type II receptors can induce type I receptor phosphorylation. TGF-β was significantly upregulated in osteophytes and cartilage in mice [25]. Moreover, TGF-β expression was increased in the joint synovium and subchondral bone in OA mouse models [26, 27]. Inhibiting endogenous TGF-β could decrease osteophyte formation and cartilage degradation in a mouse OA model [25, 27]. A previous review suggested that TGF-β could increase cartilage degradation and osteophyte formation and decrease cartilage maintenance through the TGF-β/SMAD2/3 and BMP/SMAD1/5/8 pathways, resulting in OA development [28]. However, these studies focused on endogenous TGF-β. Recent studies have gradually focused on the effects of exogenous TGF-β on OA, which is more suitable for elucidating the effect of TGF-β in PRP. Wang et al. treated OA with TGF-β1 (10 ng/mL)-induced mesenchymal stem cell-derived exosomes and found that it could decrease cartilage degeneration and enhance chondrocyte proliferation in rats [29]. Moreover, the researchers proved that the protective effects resulted from TGF-β1-mediated Sp1 downregulation and miR-135b upregulation [29]. Another study by these authors was published in 2021 and reported that TGF-β1 (10 ng/mL) was useful in the treatment of borrow mesenchymal stem cell exosomes, as it upregulated mir-135b, which suppressed the expression of MAPK6, resulting in M2 macrophage polarization in synovial tissue and reducing cartilage damage in OA rat models [30]. The results of Wang et al. seem to contradict to those of past studies. However, there are obvious differences in the methods that make a comparison unsuitable. The TGF-β1 concentration in PRP is approximately 200 ng/mL [31], which is much higher than that of endogenous TGF-β1 in the cartilage of OA patients. To the best of our knowledge, there is no direct evidence illustrating the relationship between endogenous and exogenous TGF-β and the effect of different concentrations of TGF-β in cartilage. However, these results showed that TGF-β had a positive effect on cartilage protection and that a high concentration of exogenous TGF-β released by mesenchymal stem cells could attenuate OA cartilage injury. Thus, the functions of TGF-β in PRP treatment of OA deserve to be further explored.

PDGF

PDGF was first identified in platelets and includes four monomers: PDGF-A, -B, -C, and -D [32]. These four monomers constitute five types of dimers: PDGF-AA, -AB, -BB, -CC, and -DD [32]. PDGF exerts its effects by binding to PDGFR. PDGF-AA, -AB, -BB, and -CC can bind to PDGFRα, while PDGF-BB and -DD can bind to PDGFRβ [32]. Moreover, PDGF-AB, -BB, and -CC can stimulate the heterodimeric PDGFRα/β complex [32]. The PDGF family plays an important role in wound healing through its positive effects on mitosis, chemotaxis, and angiogenesis [33]. The main types of PDGF in PRP are PDGF-AA, -AB, and -BB [14]. Regarding its effects on chondrocytes, PDGF-AA has been reported to increase proteoglycan production in chondrocytes and promote cartilage repair in rabbits [34]. Moreover, a study published in 2019 showed that suppressing the production of PDGF-AA in subchondral bone through strenuous running downregulated the PDGF/AKT signaling pathway and promoted cartilage degeneration in mice [35]. However, there have been relatively fewer studies on PDGF-AA and -AB than on PDGF-BB. Many studies have reported that PDGF-BB exerts a protective effect on OA by promoting cartilage repair, decreasing inflammation, and inhibiting cartilage hypertrophy and osteophyte formation [21, 22, 36,37,38]. Our study was published in 2021 and showed that PDGF-BB could suppress chondrocyte apoptosis by upregulating Erk phosphorylation and inhibiting the p38/Bax/caspase-3 pathway in rats [21]. In addition, we discovered that PDGF-BB binding to PDGFRβ could enhance chondrocyte proliferation and cartilage matrix synthesis in rat OA models [22]. However, a study reported that the serum levels PDGF-BB increased in OA patients during the early stage [39]. Sun et al. showed that an elevated serum level of endogenous PDGF-BB contributed to OA development by stimulating angiogenesis and that knocking out pdgfb led to less damage in articular cartilage after medial meniscus destabilization surgery in mice [40]. These results can be explained as follows: (1) the compensatory increase in PDGF-BB is an early event in OA; (2) the serum concentration of PDGF-BB is approximately 20 ng/mL in mice, which may be much less than that in cartilage [39, 40]; and (3) gene knockout affects the overall expression of PDGF, which may involve more confounding factors and thus influence the objectivity of the results. Although the PDGF-BB concentration in PRP is approximately 10 ng/mL in human [31], a relatively high local concentration of endogenous PDGF can exert different effects. Therefore, PDGF-BB still presents high potential in protecting cartilage and preventing OA progression. However, whether PDGF-AA and PDGF-AB exert similar effects is unclear and warrants further research.

EGF and TGF-α

The EGF family contains 11 structurally related proteins, including EGF, TGF-α, amphiregulin, epigen, heparin-binding EGF-like growth factor (HBEGF), epiregulin, betacellulin, and neuregulin [41]. These subfamilies have similar EGF-like motifs and exert effects via the Erb subclass of the receptor tyrosine kinase superfamily (EGFR) [42]. Previous studies have reported that EGFR activation can lead to cell proliferation, cell survival, and stem cell maintenance via the PI3K/AKT, MAPK, and JAK/STAT signaling pathways [43, 44]. The effects of EGF/TGF-α-EGFR activation have not been completely revealed. Long et al. reported that HBEGF expression was increased in a mouse OA model and human OA articular cartilage [45]. Moreover, 10–100 ng/mL HBEGF increased catabolic activity in normal chondrocytes and activation of the Erk and p38 signaling pathways in mice [45]. However, recent studies present different conclusions. Jia et al. showed that Egfr-deficient mice developed more severe OA than wild-type mice after surgical induction, as indicated by fewer superficial chondrocytes, less secretion of boundary lubrication, and weaker mechanical strength at the cartilage surface [46]. Moreover, previous studies suggested that activation of the EGFR signaling pathway under controlled conditions may promote the anabolic activity of articular cartilage and act as a viable strategy for articular cartilage repair and OA treatment in mice [47, 48]. A study conducted in 2021 on HBEGF-overexpressing mice investigated the effects of increasing EGFR activation during OA [49]. The researchers suggested that TGF-α and HBEGF were increased, which was associated with the formation of cell clusters after cartilage damage, but their endogenous expression was not sufficiently high for cartilage regeneration [49]. Therefore, whether increasing EGFR activity via TGF-α or HBEGF exert a protective effect on cartilage during OA needs further investigation.

IGF-1

IGF-1 is a cytokine that regulates skeletal growth and development [50]. Most IGF-1 in circulation is produced by the liver in response to stimulation with growth hormone (GH) [50]. Moreover, it has been proven that chondrocytes produce IGF-1 [50, 51]. A previous study demonstrated that IGF could upregulate the synthesis of proteoglycan and collagen II and downregulate the synthesis of MMP-13 via the phosphorylation of ERK1/2 and AKT in rat endplate chondrocytes [52]. A recent study published in 2021 demonstrated that IGF-1 downregulated the p38, PI3K/AKT, and NF-κB signaling pathways to decrease reactive oxygen species production, resulting in antiapoptotic effects in OA rabbit models [53]. However, studies focused on the whole body concluded that serum IGF-1 levels were positively related to morbidity due to OA in humans [54,55,56]. On the other hand, some studies showed that OA patients exhibited increased serum GH levels and decreased serum IGF-1 levels [57, 58]. Serum IGF-1 mainly comes from the liver and is regulated by the GH/IGF-1 axis [59]. As a result, serum levels of IGF-1 and its regulation are a complex process and are difficult to examine. However, to evaluate the local treatment efficacy of IGF-1 or PRP, it is not required to know whether IGF-1 expression decreases in OA cartilage and whether IGF-1 exerts positive effects when directly administered to chondrocytes.

Cytokines that have negative effects on OA treatment (Fig. 2)

Fig. 2
figure 2

Mechanisms of cytokines in PRP that have negative effects on OA treatment

VEGF

VEGF is the key survival factor for growth plate chondrocytes during embryonic development [60, 61] and plays an important role in endochondral ossification and skeletal development [62]. The VEGF family contains VEGF-A, -B, -C, -D, and placental growth factor. VEGF exerts its effects by binding to VEGF receptors (VEGFR)-1 (Flt-1), -2 (KDR), and -3 (Flt-4), which are tyrosine kinases that mediate subsequent signal transduction [63]. VEGFR-1 can bind with VEGF-A, -B, and placental growth factors; VEGFR-2 can bind with VEGF-A, -C, and -D; and VEGFR-3 can bind with VEGF-C and -D [64, 65]. During the progression of OA, the expression of VEGF increases in articular cartilage, the synovium, synovial fluid, subchondral bone, and serum, indicating that VEGF is a biomarker of OA, as indicated in the DisGeNET database [63]. The main effects of VEGF during OA progression are believed to be induced through angiogenesis. Previous studies have reported that VEGF promotes vascular invasion in not only cartilage [66,67,68] but also the synovium and meniscus [68,69,70,71,72]. As summarized in a published review, VEGF binding to VEGFR led to cartilage degeneration, osteophyte formation, subchondral bone cysts and sclerosis, synovitis, and pain [63]. Inhibiting the effects of VEGF and VEGFR in OA animal models could attenuate the progression of OA in rabbits [73]. Based on these studies, VEGF may exert negative effects on PRP treatment of OA due to the promotion of angiogenesis and cartilage degeneration. A study published in 2022 showed that VEGF-attenuated PRP-mediated improvements in OA in rats by preventing unwanted biological activity using growth factor-binding microspheres [74]. These studies suggested that VEGF-mediated attenuation of PRP exhibited better tissue repair effects than traditional PRP treatment [74]. This study suggests that if a synthetic cytokine mixture was developed to replace PRP, VEGF can be ignored.

TNF-α

TNF-α is a pleiotropic cytokine produced by a variety of cells, such as adipocytes, lymphocytes, endothelial cells, osteoblasts, and smooth muscle cells, that plays important roles in homeostasis and disease pathogenesis [75, 76]. TNF-α exerts its effects by binding to TNF receptors 1 and 2, and it has well-known antitumor functions [77]. In addition, it also plays a role in stimulating lymphocytes, monocytes, and neutrophils, modulating temperature, and combating infection [78,79,80]. Regarding its role in OA, TNF-α can increase the production of inflammatory cytokines, MMPs, and prostaglandin 9 and inhibit the synthesis of proteoglycan and collagen II in chondrocytes, leading to the degeneration of cartilage and promoting OA progression in rats [81, 82]. Moreover, TNF-α can decrease the expression of the transcription factor SOX-9 and reduce the efficiency of the respiratory chain in mice and normal human chondrocytes [83, 84]. Mechanistically, TNF-α can activate the NF-κB, JNK, and p38 signaling pathways in chondrocytes to promote the occurrence of OA in humans and rats [83, 85,86,87]. Recently, researchers have focused on the adipokine role of TNF-α, which has been proven to regulate the metabolic balance in joints by influencing the expression of cytokines, chemokines, matrix-degrading enzymes, and cell growth and differentiation factors in rats [88]. Moreover, an increase in serum levels of TNF-α was discovered in obese adults [89]. It has been discovered that obesity is positively related to the occurrence of musculoskeletal disorders, and adipocytokines secreted by adipose tissues can promote the occurrence of OA [90, 91]. The activation of the adipocytokine signaling pathway by TNF-α may explain the close relationship between obesity and OA [77, 92]. Considering its proinflammatory effect and the high probability of negative effects on OA, the levels of TNF-α in PRP should be monitored when PRP is used to treat OA.

Ang-1

Ang-1 is a cytokine that mainly regulates angiogenesis [93, 94]. The effects of Ang-1 are triggered by VEGF, followed by binding to its receptors Tie1 and Tie2 [93, 94]. When VEGF-mediated formation of immature vessels occurs, Ang-1 stabilizes the new blood vessels by recruiting neighboring mesenchymal cells and promoting mesenchymal cell differentiation into vascular smooth muscle cells in mice [93]. The level of Ang-1 is increased in the OA joint synovium, but there are no significant differences in OA osteoblasts in normal joints [95, 96]. The level of Ang-1 in OA chondrocytes has not yet been discovered. Although there is little evidence on the effects of Ang-1 on OA, previous studies have proven that it is a downstream factor of VEGF, indicating that its status in PRP treatment of OA may be similar to that of VEGF.

SDF-1α

SDF-1, which is also known as CXC motif chemokine ligand 12 (CXCL12), is a member of the CXC motif family [97]. It exerts effects by binding to its receptor CXC chemokine receptor types 4 and 7 [98]. SDF-1α is a subtype of SDF-1. SDF-1 is involved in the regulation of cell differentiation, cell distribution, cell proliferation, cell adhesion, gene expression, neovascularization in multiple cells and tissues, and embryonic development [99]. SDF-1α promotes chondrocyte proliferation via Erk- and NF-κB-mediated cyclin D1 upregulation in primary mouse chondrocytes [100]. Moreover, some studies suggest that the downregulation of SDF-1 could inhibit proliferation in human chondrocyte lines [101, 102]. The potential mechanisms of SDF-1 in OA cartilage are complicated. SDF-1 can be produced by the OA joint synovium and bind to CXCR4 and CXCR7, which are expressed on chondrocytes [103,104,105]. The SDF-1/CXCR4 axis can activate the PI3K/Akt, NF-κB, and Wnt/β-catenin pathways and suppress the TGF-β/Smad3 pathway, leading to an imbalance in aggrecan in rat OA cartilage [106, 107]. Compared to that in normal joints, the level of SDF-1 in subchondral bone is increased in OA, which results in the formation of subchondral osteosclerosis in humans [108, 109]. A study published in 2021 noted that inhibiting the SDF-1/CXCR4 axis could alleviate abnormal bone formation and angiogenesis, resulting in improvements in the subchondral bone microenvironment in OA mouse models [110]. Exogenous SDF-1α loaded on nanofibrous hyaluronic acid scaffolds has been applied to minipig joints [111]. This study reported that the articular cartilage repair capacity of SDF-1α was limited and could weaken the cartilage repair effects of TGF-β [111]. Although some studies have reported that SDF-1α could promote chondrocyte proliferation, its negative effects on the extracellular matrix and subchondral bone limit its potential use in the treatment of OA.

Cytokines with unknown effects on OA

CTGF

CTGF is composed of five domains: the secretory signal peptide, IGF binding protein (IGFBP), von Willebrand factor type C repeat (VWC), thrombospondin type 1 repeat (TSP1), and C-terminal cystine-knot (CT) modules [112]. IGFBP binds to IGF and enhances its affinity for IGFR [113]. The TSP1 domain binds to various molecules, including collagen V, fibronectin, TGF-β, and VEGF. Previous studies have reported that the TSP1 domain could upregulate the TGF-β signaling pathway and downregulate the VEGF pathway [114, 115]. Regarding its effects on OA, CTGF was overexpressed in OA cartilage and synovial fluid in mice and humans [116, 117]. CTGF could increase the production of inflammatory factors such as MMP3 and promote cartilage degeneration by activating NF-κB in mouse OA chondrocytes [117]. As mentioned earlier, PRP contains multiple biological factors, including CTGF, IGF, TGF-β, and VEGF. Whether exogenous CTGF in PRP exerts positive or negative effects on OA in the presence of IGF, TGF-β, or VEGF remains unclear. However, studies have shown that CTGF promotes the effects of TGF-β and inhibits the effects of VEGF, indicating that exogenous administration could benefit OA. These findings suggest that CTGF exerts complex effects due to its multiple biological targets in different contexts. Regarding the effect of PRP on OA, further studies need to be conducted to confirm this hypothesis.

FGF

FGF is a mitogen that regulates cellular migration, proliferation, differentiation, and survival. FGF exerts its effects by binding to the FGF receptor (FGFR), triggering conformational changes in FGFR and leading to the phosphorylation of tyrosine residues on the cytoplasmic side of FGFR [118]. The FGF/FGFR signaling pathway exerts important effects on the development and homeostasis of articular cartilage. One study revealed that the expression of FGFR1 was upregulated, and the expression of FGFR3 was downregulated in human OA cartilage [119]. The FGFR1 signaling pathway could accelerate matrix degradation by upregulating the transcription factors RUNX-2 and ELK-1 [120]. Previous studies showed that inhibiting FGR1 expression could decrease the synthesis of MMP13 and reduce the progression of cartilage degeneration [121]. Regarding the FGFR3 signaling pathway, previous studies have proven that it could promote anabolism in chondrocytes, decrease the production of inflammatory factors, and decrease hypertrophic differentiation by downregulating the IHH signaling pathway [122,123,124]. The results that FGFR1 and FGFR3 in OA were antipodals seemed contradictory because both factors were activated by FGF [125]. Moreover, FGFs are a large family associated with multiple reactions and effects, constituting a complex network [125, 126]. As a result, whether FGF exerts any effects on PRP treatment of OA remains unclear. Whether changing the mode of action of FGF by triggering FGFR3 can lead to a positive effect of PRP needs to be explored further.

HGF

HGF is a cytokine that is secreted by multiple cells, such as platelets, mesenchymal cells, and endothelial cells. It was first identified as a promoter of liver regeneration [127, 128]. HGF an exert its effects by being converted to an active heterodimeric form by HGF activator (HGFA) and then binding to its receptor c-Met, which is a transmembrane tyrosine kinase [129]. Previous studies have reported that HGF could promote the organization and reconstruction of tissues, indicating its potential for tissue repair [130]. Moreover, HGF levels were upregulated in chondrocytes and the plasma of OA patients [127, 131]. HGF has also been proven to modulate extracellular matrix components and increase the level of TGF-β in Leydig cells [132]. A study published in 2022 reported that HGF overexpression could activate the HGF/c-Met signaling pathway and induce the degradation of the extracellular cellular matrix, promoting the progression of OA in mice [133]. However, research focusing on the association between HGF and OA is limited, and the exact relationship between OA and HGF and exogenous HGF and whether it benefits OA patients remain unclear.

Conclusion

This is the first review to summarize the effects of cytokines in PRP on OA. However, because there are few available studies on PF4 and KGF, they have not been included in this review. The cytokines that are potentially beneficial or detrimental for OA treatment are summarized in Table 1. As described previously, PRP components are complex and could can various effects via complex mechanisms. The available evidence suggests that TGF-α and β, PDGF, EGF, and IGF-1 have potential for treating OA, while VEGF, TNF-α, Ang-1, and SDF-1α can induce negative effects on OA patients (Fig. 3). In addition, the effects of CTGF, FGF, HGF, PF4, and KGF on OA remain unclear (Fig. 3). Based on these studies, we concluded that not all cytokines in PRP can benefit OA patients and that the therapeutic effects of PRP have significant potential for improvement. Moreover, with consideration to the complicated preparation of PRP and the restrictions requiring repeated operations, a cytokine mixture containing these beneficial cytokines at appropriate concentrations and eliminating detrimental cytokines can be a strategy to replace PRP. This article mainly focused on OA. The same cytokines may exert different effects on other musculoskeletal disorders. For example, angiogenesis is generally considered a key process in OA and inhibits OA recovery, which explains why VEGF and Ang-1 are not beneficial for OA. However, angiogenesis is important for healing of rotator cuff tears [134, 135], indicating that VEGF and Ang-1 are beneficial cytokines for rotator cuff tears. Thus, the proposed replacement scheme can provide an opportunity to adjust the components for different diseases to induce the best benefits.

Table 1 Summary of the cytokines that are beneficial or detrimental for OA treatment
Fig. 3
figure 3

The effects of cytokines contained in PRP on OA. Cytokines can exert their effects by reacting with chondrocytes/cartilage, subchondral bone, and the synovium. In summary, TGF, PDGF, EGF and IGF-1 may exert positive effects on OA treatment; VEGF, TNF-α, and SDF-1α may exert negative effects on OA treatment; and the effects of CTGF, FGF, HGF, PF4 and KGF on OA are still unknown

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

OA:

Osteoarthritis

PRP:

Platelet-rich plasma

PDGF:

Platelet-derived growth factors

TGF:

Transforming growth factor

VEGF:

Vascular endothelial growth factor

EGF:

Epidermal growth factor

FGF:

Fibroblast growth factor

CTGF:

Connective tissue growth factor

IGF:

Insulin-like growth factor

HGF:

Hepatocyte growth factor

KGF:

Keratinocyte growth factor

Ang-1:

Angiopoietin-1

PF:

Platelet factor

SDF:

Stromal cell derived factor

TNF:

Tumor necrosis factor

References

  1. Hunter DJ, Bierma-Zeinstra S. Osteoarthritis. Lancet. 2019;393(10182):1745–59.

    CAS  PubMed  Google Scholar 

  2. Martel-Pelletier J, Barr AJ, Cicuttini FM, Conaghan PG, Cooper C, Goldring MB, Goldring SR, Jones G, Teichtahl AJ, Pelletier JP. Osteoarthritis. Nat Rev Dis Primers. 2016;2:16072.

    PubMed  Google Scholar 

  3. Zhang W, Nuki G, Moskowitz RW, Abramson S, Altman RD, Arden NK, Bierma-Zeinstra S, Brandt KD, Croft P, Doherty M, et al. OARSI recommendations for the management of hip and knee osteoarthritis: part III: changes in evidence following systematic cumulative update of research published through January 2009. Osteoarthr Cartil. 2010;18(4):476–99.

    CAS  Google Scholar 

  4. Bruyère O, Cooper C, Pelletier JP, Branco J, Luisa Brandi M, Guillemin F, Hochberg MC, Kanis JA, Kvien TK, Martel-Pelletier J, et al. An algorithm recommendation for the management of knee osteoarthritis in Europe and internationally: a report from a task force of the European society for clinical and economic aspects of osteoporosis and osteoarthritis (ESCEO). Semin Arthritis Rheum. 2014;44(3):253–63.

    PubMed  Google Scholar 

  5. Hochberg MC, Altman RD, April KT, Benkhalti M, Guyatt G, McGowan J, Towheed T, Welch V, Wells G, Tugwell P. American college of rheumatology 2012 recommendations for the use of nonpharmacologic and pharmacologic therapies in osteoarthritis of the hand, hip, and knee. Arthritis Care Res. 2012;64(4):465–74.

    CAS  Google Scholar 

  6. Hermann W, Lambova S, Muller-Ladner U. Current treatment options for osteoarthritis. Curr Rheumatol Rev. 2018;14(2):108–16.

    CAS  PubMed  Google Scholar 

  7. Fuggle NR, Cooper C, Oreffo ROC, Price AJ, Kaux JF, Maheu E, Cutolo M, Honvo G, Conaghan PG, Berenbaum F, et al. Alternative and complementary therapies in osteoarthritis and cartilage repair. Aging Clin Exp Res. 2020;32(4):547–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gupta A, Jeyaraman M, Maffulli N. Common medications which should be stopped prior to platelet-rich plasma injection. Biomedicines. 2022;10(9):2134.

    PubMed  PubMed Central  Google Scholar 

  9. Andia I, Maffulli N. Blood-derived products for tissue repair/regeneration. Int J Mol Sci. 2019;20(18):4581.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Andia I, Maffulli N. Some patients (and some of us) respond better to some biological therapies: the as yet unsolved conundrum. J Orthop Traumatol: Off J Italian Soc Orthop Traumatol. 2018;19(1):1.

    Google Scholar 

  11. Weibrich G, Hansen T, Kleis W, Buch R, Hitzler WE. Effect of platelet concentration in platelet-rich plasma on peri-implant bone regeneration. Bone. 2004;34(4):665–71.

    CAS  PubMed  Google Scholar 

  12. Graziani F, Ivanovski S, Cei S, Ducci F, Tonetti M, Gabriele M. The in vitro effect of different PRP concentrations on osteoblasts and fibroblasts. Clin Oral Implant Res. 2006;17(2):212–9.

    Google Scholar 

  13. Anitua E, Sánchez M, Nurden AT, Nurden P, Orive G, Andía I. New insights into and novel applications for platelet-rich fibrin therapies. Trends Biotechnol. 2006;24(5):227–34.

    CAS  PubMed  Google Scholar 

  14. Everts P, Onishi K, Jayaram P, Lana JF, Mautner K. Platelet-rich plasma: new performance understandings and therapeutic considerations in 2020. Int J Mol Sci. 2020;21(20):7794.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Filardo G, Di Matteo B, Kon E, Merli G, Marcacci M. Platelet-rich plasma in tendon-related disorders: results and indications. Knee Surg Sports Traumatol Arthroscop: Off J ESSKA. 2018;26(7):1984–99.

    Google Scholar 

  16. Xuan Z, Yu W, Dou Y, Wang T. Efficacy of platelet-rich plasma for low back pain: a systematic review and meta-analysis. J Neurol Surg Part A, Cent Eur Neurosurg. 2020;81(6):529–34.

    Google Scholar 

  17. Andia I, Maffulli N. A contemporary view of platelet-rich plasma therapies: moving toward refined clinical protocols and precise indications. Regen Med. 2018;13(6):717–28.

    CAS  PubMed  Google Scholar 

  18. Belk JW, Kraeutler MJ, Houck DA, Goodrich JA, Dragoo JL, McCarty EC. Platelet-rich plasma versus hyaluronic acid for knee osteoarthritis: a systematic review and meta-analysis of randomized controlled trials. Am J Sports Med. 2021;49(1):249–60.

    PubMed  Google Scholar 

  19. Zhao J, Huang H, Liang G, Zeng LF, Yang W, Liu J. Effects and safety of the combination of platelet-rich plasma (PRP) and hyaluronic acid (HA) in the treatment of knee osteoarthritis: a systematic review and meta-analysis. BMC Musculoskelet Disord. 2020;21(1):224.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Migliorini F, Driessen A, Quack V, Sippel N, Cooper B, Mansy YE, Tingart M, Eschweiler J. Comparison between intra-articular infiltrations of placebo, steroids, hyaluronic and PRP for knee osteoarthritis: a Bayesian network meta-analysis. Arch Orthop Trauma Surg. 2021;141(9):1473–90.

    PubMed  Google Scholar 

  21. Zhu P, Wang Z, Sun Z, Liao B, Cai Y. Recombinant platelet-derived growth factor-BB alleviates osteoarthritis in a rat model by decreasing chondrocyte apoptosis in vitro and in vivo. J Cell Mol Med. 2021;25(15):7472–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Cai Y, Wang Z, Liao B, Sun Z, Zhu P. Anti-inflammatory and chondroprotective effects of platelet-derived growth factor-BB on osteoarthritis rat models. J Gerontol: Ser A. 2023;78(1):51–9.

    CAS  Google Scholar 

  23. Hinck AP. Structural studies of the TGF-βs and their receptors - insights into evolution of the TGF-β superfamily. FEBS Lett. 2012;586(14):1860–70.

    CAS  PubMed  Google Scholar 

  24. Shi Y, Massagué J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell. 2003;113(6):685–700.

    CAS  PubMed  Google Scholar 

  25. Scharstuhl A, Glansbeek HL, van Beuningen HM, Vitters EL, van der Kraan PM, van den Berg WB. Inhibition of endogenous TGF-beta during experimental osteoarthritis prevents osteophyte formation and impairs cartilage repair. J Immunol. 2002;169(1):507–14.

    CAS  PubMed  Google Scholar 

  26. Bakker AC, van de Loo FA, van Beuningen HM, Sime P, van Lent PL, van der Kraan PM, Richards CD, van den Berg WB. Overexpression of active TGF-beta-1 in the murine knee joint: evidence for synovial-layer-dependent chondro-osteophyte formation. Osteoarth Cartil. 2001;9(2):128–36.

    CAS  Google Scholar 

  27. Zhen G, Wen C, Jia X, Li Y, Crane JL, Mears SC, Askin FB, Frassica FJ, Chang W, Yao J, et al. Inhibition of TGF-β signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis. Nat Med. 2013;19(6):704–12.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhai G, Doré J, Rahman P. TGF-β signal transduction pathways and osteoarthritis. Rheumatol Int. 2015;35(8):1283–92.

    CAS  PubMed  Google Scholar 

  29. Wang R, Xu B, Xu H. TGF-β1 promoted chondrocyte proliferation by regulating Sp1 through MSC-exosomes derived miR-135b. Cell Cycle. 2018;17(24):2756–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Wang R, Xu B. TGF-β1-modified MSC-derived exosomal miR-135b attenuates cartilage injury via promoting M2 synovial macrophage polarization by targeting MAPK6. Cell Tissue Res. 2021;384(1):113–27.

    CAS  PubMed  Google Scholar 

  31. Masuki H, Okudera T, Watanebe T, Suzuki M, Nishiyama K, Okudera H, Nakata K, Uematsu K, Su CY, Kawase T. Growth factor and pro-inflammatory cytokine contents in platelet-rich plasma (PRP), plasma rich in growth factors (PRGF), advanced platelet-rich fibrin (A-PRF), and concentrated growth factors (CGF). Int J Implant Dent. 2016;2(1):19.

    PubMed  PubMed Central  Google Scholar 

  32. Fredriksson L, Li H, Eriksson U. The PDGF family: four gene products form five dimeric isoforms. Cytokine Growth Factor Rev. 2004;15(4):197–204.

    CAS  PubMed  Google Scholar 

  33. Alvarez RH, Kantarjian HM, Cortes JE. Biology of platelet-derived growth factor and its involvement in disease. Mayo Clin Proc. 2006;81(9):1241–57.

    CAS  PubMed  Google Scholar 

  34. Peracchia F, Ferrari G, Poggi A, Rotilio D. IL-1 beta-induced expression of PDGF-AA isoform in rabbit articular chondrocytes is modulated by TGF-beta 1. Exp Cell Res. 1991;193(1):208–12.

    CAS  PubMed  Google Scholar 

  35. Yao Z, Chen P, Wang S, Deng G, Hu Y, Lin Q, Zhang X, Yu B. Reduced PDGF-AA in subchondral bone leads to articular cartilage degeneration after strenuous running. J Cell Physiol. 2019;234(10):17946–58.

    CAS  PubMed  Google Scholar 

  36. Razmara M, Eger G, Rorsman C, Heldin CH, Lennartsson J. MKP3 negatively modulates PDGF-induced Akt and Erk5 phosphorylation as well as chemotaxis. Cell Signal. 2012;24(3):635–40.

    CAS  PubMed  Google Scholar 

  37. Xiong LL, Xue LL, Jiang Y, Ma Z, Jin Y, Wang YC, Wang YY, Xia QJ, Zhang Y, Hu Q, et al. Suppression of PDGF induces neuronal apoptosis after neonatal cerebral hypoxia and ischemia by inhibiting P-PI3K and P-AKT signaling pathways. Brain Res. 2019;1719:77–88.

    CAS  PubMed  Google Scholar 

  38. Setiawan I, Suyasa IK, Astawa P, Dusak IWS, Kawiyana IKS, Aryana I. Recombinant platelet derived growth factor-BB and hyaluronic acid effect in rat osteoarthritis models. J Orthop. 2019;16(3):230–3.

    PubMed  PubMed Central  Google Scholar 

  39. Kisand K, Tamm AE, Lintrop M, Tamm AO. New insights into the natural course of knee osteoarthritis: early regulation of cytokines and growth factors, with emphasis on sex-dependent angiogenesis and tissue remodeling. A Pilot Study Osteoarth Cartil. 2018;26(8):1045–54.

    CAS  Google Scholar 

  40. Su W, Liu G, Liu X, Zhou Y, Sun Q, Zhen G, Wang X, Hu Y, Gao P, Demehri S, Cao X. Angiogenesis stimulated by elevated PDGF-BB in subchondral bone contributes to osteoarthritis development. JCI Insight 2020;5(8):e135446. https://doi.org/10.1172/jci.insight.135446.

  41. Abud HE, Chan WH, Jardé T. Source and impact of the EGF family of ligands on intestinal stem cells. Front Cell Develop Biol. 2021;9:685665.

    Google Scholar 

  42. Rayego-Mateos S, Rodrigues-Diez R, Morgado-Pascual JL, Valentijn F, Valdivielso JM, Goldschmeding R, Ruiz-Ortega M. Role of epidermal growth factor receptor (EGFR) and Its ligands in kidney inflammation and damage. Med Inflamm. 2018;2018:8739473.

    Google Scholar 

  43. Singh AB, Harris RC. Autocrine, paracrine and juxtacrine signaling by EGFR ligands. Cell Signal. 2005;17(10):1183–93.

    CAS  PubMed  Google Scholar 

  44. Iwakura Y, Nawa H. ErbB1-4-dependent EGF/neuregulin signals and their cross talk in the central nervous system: pathological implications in schizophrenia and Parkinson’s disease. Front Cell Neurosci. 2013;7:4.

    PubMed  PubMed Central  Google Scholar 

  45. Long DL, Ulici V, Chubinskaya S, Loeser RF. Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is increased in osteoarthritis and regulates chondrocyte catabolic and anabolic activities. Osteoarth Cartil. 2015;23(9):1523–31.

    CAS  Google Scholar 

  46. Jia H, Ma X, Tong W, Doyran B, Sun Z, Wang L, Zhang X, Zhou Y, Badar F, Chandra A, et al. EGFR signaling is critical for maintaining the superficial layer of articular cartilage and preventing osteoarthritis initiation. Proc Natl Acad Sci USA. 2016;113(50):14360–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Pest MA, Russell BA, Zhang YW, Jeong JW, Beier F. Disturbed cartilage and joint homeostasis resulting from a loss of mitogen-inducible gene 6 in a mouse model of joint dysfunction. Arthritis Rheumatol. 2014;66(10):2816–27.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Shepard JB, Jeong JW, Maihle NJ, O’Brien S, Dealy CN. Transient anabolic effects accompany epidermal growth factor receptor signal activation in articular cartilage in vivo. Arthritis Res Ther. 2013;15(3):R60.

    PubMed  PubMed Central  Google Scholar 

  49. Wei Y, Luo L, Gui T, Yu F, Yan L, Yao L, Zhong L, Yu W, Han B, Patel JM, et al. Targeting cartilage EGFR pathway for osteoarthritis treatment. Sci Trans Med. 2021;13(576):3946.

    Google Scholar 

  50. Laron Z. Insulin-like growth factor 1 (IGF-1): a growth hormone. Mol Pathol: MP. 2001;54(5):311–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Nilsson A, Isgaard J, Lindahl A, Dahlström A, Skottner A, Isaksson OG. Regulation by growth hormone of number of chondrocytes containing IGF-I in rat growth plate. Science. 1986;233(4763):571–4.

    CAS  PubMed  Google Scholar 

  52. Zhang M, Zhou Q, Liang QQ, Li CG, Holz JD, Tang D, Sheu TJ, Li TF, Shi Q, Wang YJ. IGF-1 regulation of type II collagen and MMP-13 expression in rat endplate chondrocytes via distinct signaling pathways. Osteoarth Cartil. 2009;17(1):100–6.

    CAS  Google Scholar 

  53. Hossain MA, Adithan A, Alam MJ, Kopalli SR, Kim B, Kang CW, Hwang KC, Kim JH. IGF-1 facilitates cartilage reconstruction by regulating PI3K/AKT, MAPK, and NF-kB signaling in rabbit osteoarthritis. J Inflamm Res. 2021;14:3555–68.

    PubMed  PubMed Central  Google Scholar 

  54. Claessen KM, Ramautar SR, Pereira AM, Smit JW, Biermasz NR, Kloppenburg M. Relationship between insulin-like growth factor-1 and radiographic disease in patients with primary osteoarthritis: a systematic review. Osteoarth Cartil. 2012;20(2):79–86.

    CAS  Google Scholar 

  55. Lloyd ME, Hart DJ, Nandra D, McAlindon TE, Wheeler M, Doyle DV, Spector TD. Relation between insulin-like growth factor-I concentrations, osteoarthritis, bone density, and fractures in the general population: the Chingford study. Ann Rheum Dis. 1996;55(12):870–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Hartley A, Sanderson E, Paternoster L, Teumer A, Kaplan RC, Tobias JH, Gregson CL. Mendelian randomization provides evidence for a causal effect of higher serum IGF-1 concentration on risk of hip and knee osteoarthritis. Rheumatology. 2021;60(4):1676–86.

    CAS  PubMed  Google Scholar 

  57. Denko CW, Boja B, Moskowitz RW. Growth promoting peptides in osteoarthritis: insulin, insulin-like growth factor-1, growth hormone. J Rheumatol. 1990;17(9):1217–21.

    CAS  PubMed  Google Scholar 

  58. Denko CW, Boja B, Moskowitz RW. Growth factors, insulin-like growth factor-1 and growth hormone, in synovial fluid and serum of patients with rheumatic disorders. Osteoarth Cartil. 1996;4(4):245–9.

    CAS  Google Scholar 

  59. Dixit M, Poudel SB, Yakar S. Effects of GH/IGF axis on bone and cartilage. Mol Cell Endocrinol. 2021;519:111052.

    CAS  PubMed  Google Scholar 

  60. Zelzer E, Mamluk R, Ferrara N, Johnson RS, Schipani E, Olsen BR. VEGFA is necessary for chondrocyte survival during bone development. Development. 2004;131(9):2161–71.

    CAS  PubMed  Google Scholar 

  61. Maes C, Araldi E, Haigh K, Khatri R, Van Looveren R, Giaccia AJ, Haigh JJ, Carmeliet G, Schipani E. VEGF-independent cell-autonomous functions of HIF-1α regulating oxygen consumption in fetal cartilage are critical for chondrocyte survival. J Bone Min Res: Off J Am Soc Bone Min Res. 2012;27(3):596–609.

    CAS  Google Scholar 

  62. Duan X, Murata Y, Liu Y, Nicolae C, Olsen BR, Berendsen AD. Vegfa regulates perichondrial vascularity and osteoblast differentiation in bone development. Development. 2015;142(11):1984–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Hamilton JL, Nagao M, Levine BR, Chen D, Olsen BR, Im HJ. Targeting VEGF and Its receptors for the treatment of osteoarthritis and associated pain. J Bone Min Res: Off J Am Soc Bone Min Res. 2016;31(5):911–24.

    Google Scholar 

  64. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9(6):669–76.

    CAS  PubMed  Google Scholar 

  65. Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling - in control of vascular function. Nat Rev Mol Cell Biol. 2006;7(5):359–71.

    CAS  PubMed  Google Scholar 

  66. Walsh DA, McWilliams DF, Turley MJ, Dixon MR, Fransès RE, Mapp PI, Wilson D. Angiogenesis and nerve growth factor at the osteochondral junction in rheumatoid arthritis and osteoarthritis. Rheumatology. 2010;49(10):1852–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Fransès RE, McWilliams DF, Mapp PI, Walsh DA. Osteochondral angiogenesis and increased protease inhibitor expression in OA. Osteoarth Cartil. 2010;18(4):563–71.

    Google Scholar 

  68. Walsh DA, Bonnet CS, Turner EL, Wilson D, Situ M, McWilliams DF. Angiogenesis in the synovium and at the osteochondral junction in osteoarthritis. Osteoarth Cartil. 2007;15(7):743–51.

    CAS  Google Scholar 

  69. Haywood L, McWilliams DF, Pearson CI, Gill SE, Ganesan A, Wilson D, Walsh DA. Inflammation and angiogenesis in osteoarthritis. Arthritis Rheum. 2003;48(8):2173–7.

    CAS  PubMed  Google Scholar 

  70. Giatromanolaki A, Sivridis E, Athanassou N, Zois E, Thorpe PE, Brekken RA, Gatter KC, Harris AL, Koukourakis IM, Koukourakis MI. The angiogenic pathway “vascular endothelial growth factor/flk-1(KDR)-receptor” in rheumatoid arthritis and osteoarthritis. J Pathol. 2001;194(1):101–8.

    CAS  PubMed  Google Scholar 

  71. Im HJ, Kim JS, Li X, Kotwal N, Sumner DR, van Wijnen AJ, Davis FJ, Yan D, Levine B, Henry JL, et al. Alteration of sensory neurons and spinal response to an experimental osteoarthritis pain model. Arthritis Rheum. 2010;62(10):2995–3005.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Ashraf S, Wibberley H, Mapp PI, Hill R, Wilson D, Walsh DA. Increased vascular penetration and nerve growth in the meniscus: a potential source of pain in osteoarthritis. Ann Rheum Dis. 2011;70(3):523–9.

    PubMed  Google Scholar 

  73. Nagai T, Sato M, Kobayashi M, Yokoyama M, Tani Y, Mochida J. Bevacizumab, an anti-vascular endothelial growth factor antibody, inhibits osteoarthritis. Arthritis Res Ther. 2014;16(5):427.

    PubMed  PubMed Central  Google Scholar 

  74. Lee JS, Guo P, Klett K, Hall M, Sinha K, Ravuri S, Huard J, Murphy WL. VEGF-attenuated platelet-rich plasma improves therapeutic effect on cartilage repair. Biomater Sci. 2022;10(9):2172–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Tracey KJ, Cerami A. Tumor necrosis factor, other cytokines and disease. Annu Rev Cell Biol. 1993;9:317–43.

    CAS  PubMed  Google Scholar 

  76. Tracey KJ, Cerami A. Tumor necrosis factor: a pleiotropic cytokine and therapeutic target. Annu Rev Med. 1994;45:491–503.

    CAS  PubMed  Google Scholar 

  77. Wang T, He C. Pro-inflammatory cytokines: the link between obesity and osteoarthritis. Cytokine Growth Factor Rev. 2018;44:38–50.

    PubMed  Google Scholar 

  78. Clohisy JC, Teitelbaum S, Chen S, Erdmann JM, Abu-Amer Y. Tumor necrosis factor-alpha mediates polymethylmethacrylate particle-induced NF-kappaB activation in osteoclast precursor cells. J Orthop Res: Off Publ Orthop Res Soc. 2002;20(2):174–81.

    CAS  Google Scholar 

  79. Merkel KD, Erdmann JM, McHugh KP, Abu-Amer Y, Ross FP, Teitelbaum SL. Tumor necrosis factor-alpha mediates orthopedic implant osteolysis. Am J Pathol. 1999;154(1):203–10.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Jiranek WA, Machado M, Jasty M, Jevsevar D, Wolfe HJ, Goldring SR, Goldberg MJ, Harris WH. Production of cytokines around loosened cemented acetabular components. Analysis with immunohistochemical techniques and in situ hybridization. J Bone Joint Surg Am. 1993;75(6):863–79.

    CAS  PubMed  Google Scholar 

  81. Klooster AR, Bernier SM. Tumor necrosis factor alpha and epidermal growth factor act additively to inhibit matrix gene expression by chondrocyte. Arthritis Res Ther. 2005;7(1):R127-138.

    CAS  PubMed  Google Scholar 

  82. Goldring SR, Goldring MB. The role of cytokines in cartilage matrix degeneration in osteoarthritis. Clin Orthop Related Res. 2004;1(427):S27-36.

    Google Scholar 

  83. Murakami S, Lefebvre V, de Crombrugghe B. Potent inhibition of the master chondrogenic factor Sox9 gene by interleukin-1 and tumor necrosis factor-alpha. J Biol Chem. 2000;275(5):3687–92.

    CAS  PubMed  Google Scholar 

  84. López-Armada MJ, Caramés B, Martín MA, Cillero-Pastor B, Lires-Dean M, Fuentes-Boquete I, Arenas J, Blanco FJ. Mitochondrial activity is modulated by TNFalpha and IL-1beta in normal human chondrocyte cells. Osteoarth Cartil. 2006;14(10):1011–22.

    Google Scholar 

  85. Ye Z, Chen Y, Zhang R, Dai H, Zeng C, Zeng H, Feng H, Du G, Fang H, Cai D. c-Jun N-terminal kinase - c-Jun pathway transactivates Bim to promote osteoarthritis. Can J Physiol Pharmacol. 2014;92(2):132–9.

    CAS  PubMed  Google Scholar 

  86. Séguin CA, Bernier SM. TNFalpha suppresses link protein and type II collagen expression in chondrocytes: role of MEK1/2 and NF-kappaB signaling pathways. J Cell Physiol. 2003;197(3):356–69.

    PubMed  Google Scholar 

  87. Lu YC, Jayakumar T, Duann YF, Chou YC, Hsieh CY, Yu SY, Sheu JR, Hsiao G. Chondroprotective role of sesamol by inhibiting MMPs expression via retaining NF-κB signaling in activated SW1353 cells. J Agric Food Chem. 2011;59(9):4969–78.

    CAS  PubMed  Google Scholar 

  88. Halade GV, El Jamali A, Williams PJ, Fajardo RJ, Fernandes G. Obesity-mediated inflammatory microenvironment stimulates osteoclastogenesis and bone loss in mice. Exp Gerontol. 2011;46(1):43–52.

    CAS  PubMed  Google Scholar 

  89. Park HS, Park JY, Yu R. Relationship of obesity and visceral adiposity with serum concentrations of CRP, TNF-alpha and IL-6. Diabetes Res Clin Pract. 2005;69(1):29–35.

    CAS  PubMed  Google Scholar 

  90. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Investig. 2005;115(5):1111–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Gnacińska M, Małgorzewicz S, Stojek M, Łysiak-Szydłowska W, Sworczak K. Role of adipokines in complications related to obesity: a review. Adv Med Sci. 2009;54(2):150–7.

    PubMed  Google Scholar 

  92. Xie C, Chen Q. Adipokines: new therapeutic target for osteoarthritis? Curr Rheumatol Rep. 2019;21(12):71.

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Hayes AJ, Huang WQ, Mallah J, Yang D, Lippman ME, Li LY. Angiopoietin-1 and its receptor Tie-2 participate in the regulation of capillary-like tubule formation and survival of endothelial cells. Microvasc Res. 1999;58(3):224–37.

    CAS  PubMed  Google Scholar 

  94. Polverini PJ. Angiogenesis in health and disease: insights into basic mechanisms and therapeutic opportunities. J Dent Educ. 2002;66(8):962–75.

    PubMed  Google Scholar 

  95. Lambert C, Mathy-Hartert M, Dubuc JE, Montell E, Vergés J, Munaut C, Noël A, Henrotin Y. Characterization of synovial angiogenesis in osteoarthritis patients and its modulation by chondroitin sulfate. Arthritis Res Ther. 2012;14(2):R58.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Kasama T, Isozaki T, Odai T, Matsunawa M, Wakabayashi K, Takeuchi HT, Matsukura S, Adachi M, Tezuka M, Kobayashi K. Expression of angiopoietin-1 in osteoblasts and its inhibition by tumor necrosis factor-alpha and interferon-gamma. Trans Res: J Lab Clin Med. 2007;149(5):265–73.

    CAS  Google Scholar 

  97. Bacon K, Baggiolini M, Broxmeyer H, Horuk R, Lindley I, Mantovani A, Matsushima K, Murphy P, Nomiyama H, Oppenheim J, Rot A. Chemokine/chemokine receptor nomenclature. J Leukoc Biol. 2001;70:465–6.

    CAS  Google Scholar 

  98. Balabanian K, Lagane B, Infantino S, Chow KY, Harriague J, Moepps B, Arenzana-Seisdedos F, Thelen M, Bachelerie F. The chemokine SDF-1/CXCL12 binds to and signals through the orphan receptor RDC1 in T lymphocytes. J Biol Chem. 2005;280(42):35760–6.

    CAS  PubMed  Google Scholar 

  99. Li J, Chen H, Zhang D, Xie J, Zhou X. The role of stromal cell-derived factor 1 on cartilage development and disease. Osteoarth Cartil. 2021;29(3):313–22.

    CAS  Google Scholar 

  100. Kim GW, Han MS, Park HR, Lee EJ, Jung YK, Usmani SE, Ulici V, Han SW, Beier F. CXC chemokine ligand 12a enhances chondrocyte proliferation and maturation during endochondral bone formation. Osteoarth Cartil. 2015;23(6):966–74.

    Google Scholar 

  101. Dai Y, Liu S, Xie X, Ding M, Zhou Q, Zhou X. MicroRNA-31 promotes chondrocyte proliferation by targeting C-X-C motif chemokine ligand 12. Mol Med Rep. 2019;19(3):2231–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Zheng X, Zhao FC, Pang Y, Li DY, Yao SC, Sun SS, Guo KJ. Downregulation of miR-221-3p contributes to IL-1β-induced cartilage degradation by directly targeting the SDF1/CXCR4 signaling pathway. J Mol Med. 2017;95(6):615–27.

    CAS  PubMed  Google Scholar 

  103. Xu Q, Sun XC, Shang XP, Jiang HS. Association of CXCL12 levels in synovial fluid with the radiographic severity of knee osteoarthritis. J Invest Med: Off Publ Am Federation for Clinical Research. 2012;60(6):898–901.

    CAS  Google Scholar 

  104. Kanbe K, Takagishi K, Chen Q. Stimulation of matrix metalloprotease 3 release from human chondrocytes by the interaction of stromal cell-derived factor 1 and CXC chemokine receptor 4. Arthritis Rheum. 2002;46(1):130–7.

    CAS  PubMed  Google Scholar 

  105. Jones SW, Brockbank SM, Mobbs ML, Le Good NJ, Soma-Haddrick S, Heuze AJ, Langham CJ, Timms D, Newham P, Needham MR. The orphan G-protein coupled receptor RDC1: evidence for a role in chondrocyte hypertrophy and articular cartilage matrix turnover. Osteoarth Cartil. 2006;14(6):597–608.

    CAS  Google Scholar 

  106. Lu W, He Z, Shi J, Wang Z, Wu W, Liu J, Kang H, Li F, Liang S. AMD3100 attenuates post-traumatic osteoarthritis by maintaining transforming growth factor-β1-induced expression of tissue inhibitor of metalloproteinase-3 via the phosphatidylinositol 3-kinase/akt pathway. Front Pharmacol. 2019;10:1554.

    CAS  PubMed  Google Scholar 

  107. Lu W, Shi J, Zhang J, Lv Z, Guo F, Huang H, Zhu W, Chen A. CXCL12/CXCR4 axis regulates aggrecanase activation and cartilage degradation in a post-traumatic osteoarthritis rat model. Int J Mol Sci. 2016;17(10):1522.

    PubMed  PubMed Central  Google Scholar 

  108. Lisignoli G, Toneguzzi S, Piacentini A, Cristino S, Grassi F, Cavallo C, Facchini A. CXCL12 (SDF-1) and CXCL13 (BCA-1) chemokines significantly induce proliferation and collagen type I expression in osteoblasts from osteoarthritis patients. J Cell Physiol. 2006;206(1):78–85.

    CAS  PubMed  Google Scholar 

  109. Chen Y, Lin S, Sun Y, Guo J, Lu Y, Suen CW, Zhang J, Zha Z, Ho KW, Pan X, et al. Attenuation of subchondral bone abnormal changes in osteoarthritis by inhibition of SDF-1 signaling. Osteoarth Cartil. 2017;25(6):986–94.

    CAS  Google Scholar 

  110. Qin H, Zhao X, Hu YJ, Wang S, Ma Y, He S, Shen K, Wan H, Cui Z, Yu B. Inhibition of SDF-1/CXCR4 axis to alleviate abnormal bone formation and angiogenesis could improve the subchondral bone microenvironment in osteoarthritis. Biomed Res Int. 2021;2021:8852574.

    PubMed  PubMed Central  Google Scholar 

  111. Martin AR, Patel JM, Locke RC, Eby MR, Saleh KS, Davidson MD, Sennett ML, Zlotnick HM, Chang AH, Carey JL, et al. Nanofibrous hyaluronic acid scaffolds delivering TGF-β3 and SDF-1α for articular cartilage repair in a large animal model. Acta Biomater. 2021;126:170–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Tu M, Yao Y, Qiao FH, Wang L. The pathogenic role of connective tissue growth factor in osteoarthritis. Biosci Rep. 2019;39(7):1374.

    Google Scholar 

  113. Baxter RC, Twigg SM. Actions of IGF binding proteins and related proteins in adipose tissue. Trends Endocrinol Metab. 2009;20(10):499–505.

    CAS  PubMed  Google Scholar 

  114. Bazzazi H, Zhang Y, Jafarnejad M, Isenberg JS, Annex BH, Popel AS. Computer simulation of TSP1 inhibition of VEGF-Akt-eNOS: an angiogenesis triple threat. Front Physiol. 2018;9:644.

    PubMed  PubMed Central  Google Scholar 

  115. Lu A, Miao M, Schoeb TR, Agarwal A, Murphy-Ullrich JE. Blockade of TSP1-dependent TGF-β activity reduces renal injury and proteinuria in a murine model of diabetic nephropathy. Am J Pathol. 2011;178(6):2573–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Omoto S, Nishida K, Yamaai Y, Shibahara M, Nishida T, Doi T, Asahara H, Nakanishi T, Inoue H, Takigawa M. Expression and localization of connective tissue growth factor (CTGF/Hcs24/CCN2) in osteoarthritic cartilage. Osteoarth Cartil. 2004;12(10):771–8.

    Google Scholar 

  117. Blaney Davidson EN, Vitters EL, Mooren FM, Oliver N, Berg WB, van der Kraan PM. Connective tissue growth factor/CCN2 overexpression in mouse synovial lining results in transient fibrosis and cartilage damage. Arthritis Rheum. 2006;54(5):1653–61.

    CAS  PubMed  Google Scholar 

  118. Farrell B, Breeze AL. Structure, activation and dysregulation of fibroblast growth factor receptor kinases: perspectives for clinical targeting. Biochem Soc Trans. 2018;46(6):1753–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Im HJ, Muddasani P, Natarajan V, Schmid TM, Block JA, Davis F, van Wijnen AJ, Loeser RF. Basic fibroblast growth factor stimulates matrix metalloproteinase-13 via the molecular cross-talk between the mitogen-activated protein kinases and protein kinase Cdelta pathways in human adult articular chondrocytes. J Biol Chem. 2007;282(15):11110–21.

    CAS  PubMed  Google Scholar 

  120. Yan D, Chen D, Cool SM, van Wijnen AJ, Mikecz K, Murphy G, Im HJ. Fibroblast growth factor receptor 1 is principally responsible for fibroblast growth factor 2-induced catabolic activities in human articular chondrocytes. Arthritis Res Ther. 2011;13(4):R130.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Weng T, Yi L, Huang J, Luo F, Wen X, Du X, Chen Q, Deng C, Chen D, Chen L. Genetic inhibition of fibroblast growth factor receptor 1 in knee cartilage attenuates the degeneration of articular cartilage in adult mice. Arthritis Rheum. 2012;64(12):3982–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Tang J, Su N, Zhou S, Xie Y, Huang J, Wen X, Wang Z, Wang Q, Xu W, Du X, et al. Fibroblast growth factor receptor 3 inhibits osteoarthritis progression in the knee joints of adult mice. Arthritis Rheumatol. 2016;68(10):2432–43.

    CAS  PubMed  Google Scholar 

  123. Zhou S, Xie Y, Li W, Huang J, Wang Z, Tang J, Xu W, Sun X, Tan Q, Huang S, et al. Conditional deletion of Fgfr3 in chondrocytes leads to osteoarthritis-like defects in temporomandibular joint of adult mice. Sci Rep. 2016;6:24039.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Kuang L, Wu J, Su N, Qi H, Chen H, Zhou S, Xiong Y, Du X, Tan Q, Yang J, et al. FGFR3 deficiency enhances CXCL12-dependent chemotaxis of macrophages via upregulating CXCR7 and aggravates joint destruction in mice. Ann Rheum Dis. 2020;79(1):112–22.

    CAS  PubMed  Google Scholar 

  125. Xie Y, Su N, Yang J, Tan Q, Huang S, Jin M, Ni Z, Zhang B, Zhang D, Luo F, et al. FGF/FGFR signaling in health and disease. Signal Transduct Target Ther. 2020;5(1):181.

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Xie Y, Zinkle A, Chen L, Mohammadi M. Fibroblast growth factor signalling in osteoarthritis and cartilage repair. Nat Rev Rheumatol. 2020;16(10):547–64.

    CAS  PubMed  Google Scholar 

  127. Nagashima M, Hasegawa J, Kato K, Yamazaki J, Nishigai K, Ishiwata T, Asano G, Yoshino S. Hepatocyte growth factor (HGF), HGF activator, and c-Met in synovial tissues in rheumatoid arthritis and osteoarthritis. J Rheumatol. 2001;28(8):1772–8.

    CAS  PubMed  Google Scholar 

  128. Nakamura T, Nawa K, Ichihara A. Partial purification and characterization of hepatocyte growth factor from serum of hepatectomized rats. Biochem Biophys Res Commun. 1984;122(3):1450–9.

    CAS  PubMed  Google Scholar 

  129. Miyazawa K, Shimomura T, Kitamura N. Activation of hepatocyte growth factor in the injured tissues is mediated by hepatocyte growth factor activator. J Biol Chem. 1996;271(7):3615–8.

    CAS  PubMed  Google Scholar 

  130. Gherardi E, Gray J, Stoker M, Perryman M, Furlong R. Purification of scatter factor, a fibroblast-derived basic protein that modulates epithelial interactions and movement. Proc Natl Acad Sci USA. 1989;86(15):5844–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Mabey T, Honsawek S, Saetan N, Poovorawan Y, Tanavalee A, Yuktanandana P. Angiogenic cytokine expression profiles in plasma and synovial fluid of primary knee osteoarthritis. Int Orthop. 2014;38(9):1885–92.

    PubMed  Google Scholar 

  132. Catizone A, Ricci G, Tufano MA, Perfetto B, Canipari R, Galdieri M. Hepatocyte growth factor (HGF) modulates Leydig cell extracellular matrix components. J Androl. 2010;31(3):306–13.

    CAS  PubMed  Google Scholar 

  133. Que W, Liu H, Yang Q. CircPRKCH modulates extracellular matrix formation and metabolism by regulating the miR-145/HGF axis in osteoarthritis. Arthritis Res Ther. 2022;24(1):216.

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Huang Y, He B, Wang L, Yuan B, Shu H, Zhang F, Sun L. Bone marrow mesenchymal stem cell-derived exosomes promote rotator cuff tendon-bone healing by promoting angiogenesis and regulating M1 macrophages in rats. Stem Cell Res Ther. 2020;11(1):496.

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Noh KC, Park SH, Yang CJ, Lee GW, Kim MK, Kang YH. Involvement of synovial matrix degradation and angiogenesis in oxidative stress-exposed degenerative rotator cuff tears with osteoarthritis. J Shoulder Elbow Surg. 2018;27(1):141–50.

    PubMed  Google Scholar 

Download references

Acknowledgements

None.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

YC and HY designed and directed the article. ZW and PZ wrote the manuscript. YC, HY, and BL revised the manuscript. All authors have read and verified the underlying data and approved the final version of the manuscript.

Corresponding authors

Correspondence to Hongbo You or Yu Cai.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publications

All authors agree to publish this article.

Competing interests

The authors declare that they have no conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Z., Zhu, P., Liao, B. et al. Effects and action mechanisms of individual cytokines contained in PRP on osteoarthritis. J Orthop Surg Res 18, 713 (2023). https://doi.org/10.1186/s13018-023-04119-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13018-023-04119-3

Keywords