Next Article in Journal
Designing Personalized Antigen-Specific Immunotherapies for Autoimmune Diseases—The Case for Using Ignored Target Cell Antigen Determinants
Next Article in Special Issue
Platelet Aggregation, Mitochondrial Function and Morphology in Cold Storage: Impact of Resveratrol and Cytochrome c Supplementation
Previous Article in Journal
Leaky Gum: The Revisited Origin of Systemic Diseases
Previous Article in Special Issue
Platelets, a Key Cell in Inflammation and Atherosclerosis Progression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Platelets in Osteoarthritis—Updated Systematic Review and Meta-Analysis on the Role of Platelet-Rich Plasma in Osteoarthritis

Centre of Postgraduate Medical Education, Department of Orthopaedics and Trauma Surgery, Professor A. Gruca Teaching Hospital, Konarskiego 13, 05-400 Otwock, Poland
*
Author to whom correspondence should be addressed.
Cells 2022, 11(7), 1080; https://doi.org/10.3390/cells11071080
Submission received: 30 January 2022 / Revised: 13 March 2022 / Accepted: 17 March 2022 / Published: 23 March 2022
(This article belongs to the Special Issue Role of Platelets in Inflammatory Diseases)

Abstract

:
Platelets are an essential component of hemostasis, with an increasing role in host inflammatory processes in injured tissues. The reaction between receptors and vascular endothelial cells results in the recruitment of platelets in the immune response pathway. The aim of the present review is to describe the role of platelets in osteoarthritis. Platelets induce secretion of biological substances, many of which are key players in the inflammatory response in osteoarthritis. Molecules involved in cartilage degeneration, or being markers of inflammation in osteoarthritis, are cytokines, such as tumor necrosis factor α (TNFα), interleukins (IL), type II collagen, aggrecan, and metalloproteinases. Surprisingly, platelets may also be used as a treatment modality for osteoarthritis. Multiple randomized controlled trials included in our systematic review and meta-analyses prove the effectiveness of platelet-rich plasma (PRP) as a minimally invasive method of pain alleviation in osteoarthritis treatment.

Graphical Abstract

1. Osteoarthritis

Osteoarthritis (OA) is a degenerative joint disease that affects between 10% and 18% of middle-aged persons, and up to 44% of older populations [1,2,3]. Although OA was previously deemed to be mechanically activated wear of the cartilage, it is now well established as a low-grade inflammatory disease of the entire joint. OA may be characterized as primary, if there is absence of an antecedent and underlying cause, or secondary, if there is an underlying cause or significant triggering event such as prior trauma [3,4,5].
The main symptoms of OA include pain, stiffness, and loss of function and its sequelae that result in muscle weakness, poor balance, and joint deformity and instability [3,6]. Factors influencing a higher risk and severity of OA are significantly related to older age, female gender, joint injury, anatomic factors, heavy work activities, some professional sport activities, obesity, and genetics [3,4,7]. Additionally, smoking, bone density, and diet remain a focus of OA research [8].
The diagnosis of osteoarthritis is undertaken in symptomatic patients. The radiographic findings of OA can be seen in different modalities, including plain radiographs, ultrasound (US), computed tomography (CT), or magnetic resonance imaging (MRI). Although plain radiograph is the most commonly used modality, MRI has many advantages, including the possibility of assessing joint structures [3,9].
Beneficial treatment of OA may be divided into non-surgical (non-pharmacological and pharmacological) or surgical means [1,10,11]. Non-pharmacological interventions, which include lifestyle modifications such as exercise or weight loss, are the backbone of knee OA management. Irrespective of OA severity, all of these interventions can be used as add-ons to pharmacological treatment. Conventional pharmacological interventions include topical or oral non-steroidal anti-inflammatory drugs (NSAID) and other analgesic drugs, in addition to intra-articular injections with corticosteroids, hyaluronic acid or platelet-rich plasma (PRP). These interventions have proven to be successful, albeit with short-term benefits [12,13].
Surgical treatment plays a significant role when joint-related symptoms persist, despite the use of non-surgical interventions. A growing focus concerns the potential application of experimental methods, which are currently being extensively developed [14,15,16].

2. Inflammation—Role in the Pathogenesis of Osteoarthritis

Osteoarthritis is the most common cause of joint degenerative disease, and is a leading cause of disability worldwide; it remains difficult to treat [4]. It has been regarded primarily as a degenerative disease of the cartilage, focusing mainly on acknowledged pathological changes such as cartilage breakdown, the formation of osteophytes, and subchondral bone sclerosis [17]. Although there have been previous studies which indicate that inflammation has a critical role in its pathogenesis, comparatively little attention has been given to the concept of OA pathogenesis involving not only malfunction of cartilage, but also inflammation of the synovial lining [18]. Currently, we no longer view OA as a quintessential degenerative disease resulting from exposure to average bodily wear and tear, but rather as a multifactorial disorder in which low-grade, chronic inflammation has a central role. The involvement of inflammatory components such as cytokines, chemokines, and other inflammatory mediators that are produced by the synovium and chondrocytes, can be easily measured in the synovial fluid of OA patients, and is now well recognized as means of diagnosis [19]. Inflammation is a typical immunological response to pathogens and cellular aberrations that triggers the immune system. Undesired material is primarily recognized by neutrophils, then by specialized phagocytic, megakaryocyte, macrophage cells, and by other cellular mechanisms that are engaged to resolve the pathology and restore homeostasis. Notwithstanding, unresolved chronic inflammation can result in harmful effects. It may result in tissue breakdown and degradation, causing the development of multiple chronic conditions including arthrosclerosis, neurodegenerative diseases, arthritis, inflammatory bowel disease, and cancer. Antecedently, inflammatory arthritis was defined as cellular inflammation described by increased leukocyte numbers in the affected joint tissues and synovial fluid. It was proved that OA is characterized by the presence of a host of proinflammatory mediators, including cytokines and chemokines, which are part of an immune response to joint injury [20]. It remains unclear whether morphological changes that occur in the OA synovium are primary to a systemic immune response or secondary to cartilage degradation and lesions of the subchondral bone [17].

3. Osteoarthritis—The Role of Cytokines and Other Inflammatory Mediators

There is a long list of cytokines and chemokines that are detected in OA synovial fluid, such as the following: interleukin (IL) IL-1β, IL-6, IL-7, IL-8, IL-15, IL-17, IL-18, and IL-36; tumor necrosis factor α (TNFα), oncostatin M (OSM), and growth-related oncogene (GRO)-α; chemokine (C-C-motif), ligand 19 (CCL19), macrophage inflammatory protein (MIP)-1β, and transforming growth factor (TGF) α; interferon-induced protein (IP) 10 and monokine induced by interferon (MIG) [20,21,22,23].
Surprisingly, some research has provided evidence to support the hypothesis that inhibition of complement activation in OA by gene deletion or pharmacologic modulation was found to protect mice from surgically induced OA [24,25]. Fragments of matrix proteins including fibronectin, cartilage oligomeric protein (COMP), fibromodulin, proteoglycans, and collagen are released from the damaged matrix. These proteins are able to stimulate the innate immune response and promote the upregulation of degradative pathways via activation of toll-like receptors (TLR) and integrins [20,26,27].
Many of the proinflammatory and catabolic pathways in the OA joint involve activation of the nuclear factor kappa-light-chain-enhancer of activated B cell (NF-kB) family of transcriptional regulators. Other inflammatory mediators include the alarmins (S100 proteins) and the damage-associated molecular patterns (DAMPs). These mediators attract macrophages and control synovitis by enhancing the expression of proteases into the joint. The destruction of joint tissues in OA is mediated by a large number of proteases [28]. Our understanding of the metalloproteinases (MMPs), cysteine proteinases (including cathepsin K), and serine proteinases that are involved in OA is mainly concentrated on possible causes of degradation of cartilage extracellular matrix proteins. Aggrecan is a large proteoglycan that provides much of the resiliency of cartilage. It is degraded in the early stages of OA by members of the ADAMTS (adisintegrin and metalloproteinase with thrombospondin motifs) family, typically referred to as aggrecanases (ADAMTS-4 and -5) [14,28].
Type II collagen, the most abundant collagen in cartilage, is responsible for the tensile strength of cartilage, and is degraded by collagenases. Given the importance of ADAMTS-5 and MMP-13 in OA, development of specific inhibitors to these proteases has been proven as a potentially beneficial, disease-modifying therapy [14]. Thus, we cannot disregard the role of tissue inhibitors of metalloproteinases (TIMPs) that are endogenous inhibitors found in joint tissues and synovial fluid; these may serve as a substitute to synthetic small molecule inhibitors.
MMPs are produced as pro-forms that require proteolytic cleavage in order to be activated. Serin proteases, including HtrA1 and activated protein C, can serve this role and therefore also serve as therapeutic targets in OA research. Cathepsin K is a cysteine proteinase that is expressed by osteoclasts; it can degrade type I collagen in bone but also may degrade type II collagen in cartilage [14,21,28].
Several studies have indicated that there are a number of potential mediators of OA that are not considered proinflammatory mediators, but appear to promote OA by either activating pathways that promote joint tissue destruction, or inhibit the ability of cells to repair the damaged matrix; these are bone morphogenetic proteins (BMPs), fibroblast growth factors (FGF), and Wnts [29,30,31,32].
One further area explored in the novel investigation of OA pathogenesis is epigenetics. There is a growing number of processes, including DNA methylation, histone modifications (acetylation and methylation), microRNAs (miRNA), and long noncoding RNAs, in the epigenetic control of gene transcription. It is not yet known whether epigenetic changes play a key role in the development of OA in humans [33,34,35].
The importance of innate immunity has been associated with the development of synovitis, activation of downstream inflammatory, and catabolic events in articular cartilage that result in progression of OA [19,20].
Therefore, forasmuch as OA might not adhere to the classical presence of cardinal symptoms of inflammation (rubor, calor, dolor, and tumor), there is no strong evidence for the presence of robust adaptive immune reactions; moreover, there is still significant evidence to support a pathogenic role for both local and systemic inflammation in OA development.

4. Platelets

Although platelets were traditionally considered essential components of hemostasis, there is growing accedence to the fact that they also play a critical role in the inflammatory and immune responses [36]. The normal range of anucleate discoid platelets is 150,000–400,000/µL, and their main function is to assure hemostasis by preventing loss of blood. Platelets usually circulate for 7 to 10 days; then, they are either used up in hemostasis or undergo apoptosis [37]. Platelets are produced in vast numbers via a multistep process in the bone marrow by megakaryocytes and, as anucleated cells, are incapable of nucleic acid transcription; however, they maintain a meaningful amount of messenger RNA (mRNA), microRNAs and noncoding RNAs (ncRNA). With regard to these features, platelets are able to actively control mRNA translation, perform protein synthesis, and secrete peptides around them which are fundamental features of the complex hemostatic response.
Recent studies have shown increasing evidence that platelets play a significant role in host inflammatory and immune responses [38]. Consequently, antiplatelet medications influence immunity and modify platelet response to inflammation, which result in a significant reduction in mortality rate from either infections or sepsis [39]. In physiological conditions, the role of platelets is related to their reactivity, formed as a result of the reaction between their receptors and vascular endothelial cells in the damaged blood vessel. It is well established that an increase in platelet count is related to the reactivity of megakaryocytes, and to the activity of released pro-platelet cytokines [40,41]. When blood vessels are disturbed by inflammation, platelets are the first cells to be recruited by the vascular endothelium via markers such as selectin and integrin receptors; TLR, complement receptor (CR), cytokines, and chemokines; adenosine diphosphate receptor (ADP), purinoreceptor (PCY); and protease-activated receptors 1 and 4 (PAR-1 and 4). They may be activated and forced to merge with the blood vessel, including polymorphonuclear cells (PMNs), mononuclear cells (MNs), dendritic cells (DC), and T and B cells, usually leading to local inflammation [42,43]. Moreover, the activation of platelets is frequently intensified by P-selectin, which is secreted by the cells of the injured endothelium. P-selectin also stimulates platelets, and activates neutrophils and monocytes towards the activation of endothelial cells [44]. The characteristic features of activated platelets are based on the presence of intracellular adhesion molecules ICAM-1 and monocyte chemoattractant protein-1 (MCP-1), which cause increased adhesion of neutrophils and monocytes. There are many well-known proinflammatory factors that are designated as either recruiters or activators of leukocytes. Their main role is to coordinate the release of clusters of differentiation (CD) 154 (CD154), or CD40 ligands; CXC chemokine ligands (CXCL) CXCL1, CXCL4, CXCL5, CXCL7, and CXCL12; and IL-8 and TGF-β. Platelets induce the secretion of biologically active substances (growth factors and mitogens, coagulation system proteins, cytokines, chemokines, proteolytic enzyme, microRNA, mRNAs, and others), including Weibel–Palade bodies and extracellular vesicles (EV) [45,46,47]; these cause not only a change in the activity of endothelial cells, but also a change in the activity of immune system cells [48,49,50].
The increased exposure of collagen fragments in the damaged endothelium causes inflammation [40,51]. The inflammatory effect of activated platelets is associated with the secretion of platelet-activating factor (PAF) and vascular endothelial growth factor (VEGF); these cause relaxation of the endothelium of blood vessels and increase the inflow of leukocytes and platelets. In addition to this effect, activated platelets can stimulate neutrophils in the neutrophil extracellular traps (NET) network, with the participation of platelets and the process of PMN cell apoptosis. As a result, this affects the lifetime of these cells and the duration of inflammation. We know that various platelet functions may activate an inflammatory response that may result in worsened pathogenesis of many diseases, such as rheumatoid arthritis or cardiovascular diseases. Platelets usually affect neutrophils, and by increasing the production of leukotrienes (LT) such as LTC4, LTD4, and LTE4, this may increase vascular permeability, causing exudate and oedema. The activation state of platelets includes change of their shape from discoid to circular, with numerous pseudopodia [41,52,53] (Figure 1).
Platelet involvement in different medical conditions is described in the context of bacterial and viral infection, sepsis, cardiological conditions, oncologic process, rheumatoid arthritis, and related autoimmune diseases; diabetes and obesity, spleen or kidney damage, neurological diseases of the central nervous system, lung diseases, especially asthma and pneumonia; atherosclerosis and vascular diseases; and also inflammation of the mouth, large intestine, and kidneys. Currently, it is widely recognized that platelets represent a hallmark of immunity and may be considered key players in the inflammatory response as a consequence of their capacity to interact with almost all immune cells. Current understanding of platelet activation pathways and their role as biomarkers could provide new realms of both diagnostic and therapeutic possibilities in evaluation of disease activities and responses to treatment [52,53].

5. Platelets in OA

In the past, OA has been described as a local disorder limited to articular cartilage. Recently, OA has been treated as a low-grade inflammatory systemic disease affecting the synovium, synovial fluid, cartilage, subchondral bone, and muscles [54,55,56,57]. Platelets engaged in the inflammatory process, vascular integrity, and hemostasis may play a significant role in OA. Nevertheless, the connection between OA and platelets is still indistinct [54]. Biochemical markers associated with the development of OA are products of cartilage and bone degeneration, such as type II collagen, aggrecan, matrix MMPs, and procollagen type I. Additionally, proinflammatory and anti-inflammatory (IL-6, IL-1B, TNF-a, IL-10, IL-13, and IL-4) molecules are related to the progression of OA by angiogenesis and chemotaxis. Most of them are produced, stimulated, connected, or released by platelets [58,59].
Platelets contain three types of granules: α-granules, dense granules, and lysosomes. α-granules play the main role in inflammation. They interact with leukocytes and vascular cells directly, as well as indirectly, by the secretion of mediators such as cytokines and chemokines. The predominant role of cytokines is in the regulation of the inflammatory response, such as the proliferation and differentiation of lymphocytes [60]. Proinflammatory cytokines secreted by platelets are IL-1, TGF-β, platelet-derived growth factor (PDGF), macrophage inflammatory protein-1a (MIP-1a), and TNF-α [60,61,62]. Chemokines called chemotactic cytokines are signaling proteins released from granules after platelet activation, and have a major role in the activation of the immune response. α-granules secrete many chemokines, such as CXCL1 (β-tromboglobulin), CXCL4 (PF4, platelet factor 4), CCL5 (RANTES, regulated upon activation and normal T cell expressed and secreted), and CXCL12 (SDF-1, stromal cell-derived factor-1) [60,63]. They recruit and differentiate lymphocyte T, and activate neutrophils and phagocytosis of macrophages [60].
Another important molecule produced by α-granules is P-selectin (CD62P), which is also a marker of platelet activation. P-selectin glycoprotein ligand-1 (PSGL-1) connects to P-selectin, and this complex mediates platelet adhesion to leukocytes and monocytes. As an effect, it leads to the recruitment of leukocytes during inflammation [60,64,65]. P-selectin also plays a role in the tethering and rolling of leukocytes [65].
Microparticles (MPs) are platelet-derived vesicles present in arthritic joint fluid. They contain signaling molecules and membrane receptors, alongside activating synovial fibroblasts. MPs induce the production of inflammatory mediators which play an integral role in the development of osteoarthritis [62,63]. They have been recognized as markers of platelet activation, since fragmentation of MPs spontaneously occurs during platelet aggregation and during interaction with monocytes and leucocytes [64,66]. MPs, by releasing IL-1α and IL-1β, stimulate fibroblast-like synoviocytes (FLS) to secrete IL-6 and IL-8 [52,62,67]. MPs are released by the signaling receptor glycoprotein VI (GPVI), which triggers MP production by stimulation of IL-1. GPVI is the predominant collagen receptor expressed by megakaryocytes and platelets [66]. Platelets are activated by collagens I, III, IV, V, VI, and XIII. Among these, collagens I, III, and IV are more reactive with platelets, and induce strong adhesion [68]. The soft tissue surrounding the joint consists mostly of collagens I and III, and it is known that platelets react with them deep below the subendothelium [63]. Platelets and chondrocytes generate reactive oxygen species (ROS). The basal level of intercellular ROS is enlarged by collagen GPVI stimulation. An increased amount of ROS leads to the activation of platelets and thrombus formation, and then to cartilage destruction as well as inhibition of cartilage synthesis [54,69].
Platelet number is associated with plasma IL-1β concentration. IL-1β stimulates platelets to form an autocrine stimulatory loop by platelets expressing the IL1R1 receptor [67]. IL-1β is a proinflammatory cytokine and a suppressor of type II collagen and aggrecan synthesis [58]. In addition, IL-1β increases the expression of MMPs and induces production of a cartilage catabolic mediator, nitric oxide [70,71], which inhibits platelet aggregation [72]. Moreover, some studies showed mild to moderate correlation of IL-1β concentration and joint space width or joint alignment [73], in addition to increased risk of familial OA [71]. A negative correlation between a decrease in pain severity and a change of IL-1β in synovial fluid was observed [74]. IL-1β also stimulates the production of IL-6 and IL-8, which are also proinflammatory cytokines entailed in platelet hyperactivation [70,75].
As a multifunctional cytokine, IL-6 regulates the acute phase of the inflammatory and immune responses. Biological activities of IL-6 are promoted by trans-signaling, which results in conversion from acute to chronic inflammation [75,76]. Signal transduction is mediated by two molecules: membrane-bound β-receptor glycoprotein 130 present on platelets (gp130), and IL-6 receptor (IL-6R) [75]. IL-6 builds a complex with soluble forms of IL-6R; then, this complex binds to gp130 [76]. Gp130 is expressed, inter alia, on platelets. The presence of IL-6 leads to platelet-derived IL-6 trans-signaling [75], which causes platelet hyperactivation [77]. Higher platelet activation leads to survival and leukocyte activation [57]. IL-6 is also a mediator of platelet hyperactivity, and by acting directly on megakaryocytes, increases platelets in circulation and moreover initiates the coagulation cascade [54,77,78]. In animal models, IL-6 decreased the production of collagen type II [79]. Livshits et al. demonstrated a relationship between IL-6 and the increased prevalence of OA diagnoses in long term studies (15-year follow-up) [80]. Stannus et al. found associations between the circulating levels of IL-6 and TNF-α and cartilage loss in knee OA in addition to joint space narrowing in the 3-year follow-up [59].
Mean platelet volume (MPV) is suggested to be a marker of low-grade inflammation in OA [81]. MPV reflects platelet size and platelet activity, and additionally regulates thrombopoesis in conjunction with other inflammatory cytokines (IL-1, Il-6, and TNF-a) which are found in OA serum [57,81]. Balbaloglu et al. [81] investigated MPV levels in reference to patients with synovitis in knee OA. They compared MPV results in three groups—patients with synovitis associated to knee osteoarthritis, patients with knee osteoarthritis, and a control group (patients who did not have joint complaints). MPV blood levels significantly differed between the patients with synovitis associated with osteoarthritis and the patients with knee osteoarthritis; the levels also differed with the control group. However, there was no difference between OA and control. This suggests that MPV may be useful as a marker for inflammation in osteoarthritis in synovitis patients [81]. Another study analyzed MPV as a predictor of hip OA. Retrospectively, radiographic images were classified according to the Kellgren–Lawrence (KL) grading score. In statistical analyses, MPV was significantly higher in KL grades 3–4 compared with KL 1–2 [57]. Kwon et al. [54] investigated the correlation between platelet count and knee or hip osteoarthritis in Korean women over 50 years of age. The KL grading system was used for evaluating the OA grade. This study showed significantly higher platelet and WBC counts in patients with symptomatic OA, confirmed by radiography. The researchers also found a linear relationship between OA and platelet count [54].
TNF-α, a proinflammatory cytokine, activates platelets by interaction with tumor necrosis factor receptor superfamily member 1A (TNFRSF1A), and additionally stimulates the arachidonic acid pathway. Other cells witch release TNF-α may exert agonist activity for platelets [59]. Breda et al. found that a higher level of TNF receptors was associated with increased pain, stiffness, and moreover worse radiographic scores in osteoarthritis [82].
Extended platelet interaction with cells involved in the inflammatory process may lead to an adverse effect as a result of overstimulation of the immunological system [60]. Prolonged inflammation leads to destruction of cartilage, bone, and ligaments.

6. Platelets as a Treatment for OA

Platelet-rich plasma as a minimally invasive therapy for OA is currently well known and widely used. It is a safe, low-cost, and effective means as an alternative non-surgical treatment for knee osteoarthritis. PRP consists of autologous plasma with platelets, which induce secretion of growth factors such as PDGF, TGF-β1, TGF-β2, insulin-like growth factor (IGF), epidermal growth factor (EGF), VEGF, and FGF. Platelets also contain cytokines and immunomodulators. As a result of their anti-inflammatory nature and potential role in tissue healing, they are a research target for the potential treatment for osteoarthritis [83,84,85]. Several authors demonstrated that platelets promote healing and modulate inflammation, both in vitro and in vivo [86,87,88]. TGF-β is important for chondrogenesis by inducing cell proliferation in cartilage, osteochondrogenic differentiation, and by increasing gene expression of aggrecan and collagen II [89]. PDGF stimulates cell proliferation and production of proteoglycan [90], and FGF induces chondrocyte and MSC proliferation [91]. Several GFs have a synergistic effect in the OA joint.
PRP is activated by the addition of calcium chloride or natural collagen from damaged tissue, which becomes prepared to secrete granules and form a clot. Platelet-rich fibrin (PRF) does not require activators, and the clotting process occurs naturally. In addition, both PRP and PRF may contain leukocytes. This process yields four main products (pure PRP, leukocyte-rich PRP, pure PRF, and leukocyte-rich PRF), each of which has a different mechanism of action, contains different molecules, and should be used in different indications [84,92].
An in vitro study performed by Osterman et al. created a coculture system with synovia and cartilage harvested from patients during total knee arthroplasties. Two preparations were used—leukocyte-poor and leukocyte-rich PRP. Both preparations decreased the expression of genes correlated with the inflammatory process in OA (TIMP-1, ADAMTS-5) cartilage and synovia, and increased aggrecan expression compared to non-OA cartilage. Moreover, nitric oxide production, which is an indicator of inflammation, was significantly lower in both PRP groups [87].
A similar study compared cartilage and synovia from patients undergoing TKA and cocultured with HA or PRP. An increaseaggrecan gene expression and decreased COL1A1 in the PRP group were observed in comparison to HA. A decreased volume of TNF-α in both PRP and HA was observed in comparison to the control, and IL-6 concentration was significantly decreased in the HA group. IL-1B was decreased in all treatment groups. There was no difference in cartilage MMP-13 expression in all samples of cartilage, but MMP-13 expression in synoviocytes decreased in the PRP group compared to the control and HA groups. Likewise, an increase in synoviocyte gene expression of HAS-2 was observed in the PRP group, but there was no difference in TNF-α in all three groups [93].
Lacko et al. investigated the effect of three PRP injections in patients with unilateral knee OA. This study showed an improvement in the WOMAC score and a reduction in the VAS score for pain at the 3-month follow-up. The most important finding was a significant decrease in proinflammatory markers (eotaxin, MCP-1, MMP-1, IL-1o, EGF, PDGF, and TGF-B) compared to baseline level, and an increase in pro-anabolic markers (BMP-2, COMP, collagen type II, and GRO) of the cartilage and anti-inflammatory markers [94].
Tang et al. compared PRP injection versus hyaluronic acid (HA) for patients with OA. In the 20 randomized controlled trials (RCTs) enrolled in their meta-analysis in 2020, it was shown that PRP has a better analgesic effect than HA in long-term recovery (at the 6-month and 12-month follow-ups) and also leads to better functional recovery in 12 months. The risk of adverse events was not increased compared with HA [95]. Similar observations were noted by Trams et al. in their meta-analysis [96].
Kon et al. performed a systematic review of PRP injections in OA patients. Twelve meta-analyses were included, comparing PRP injections to HA, corticosteroids (CS), and placebo. The main finding showed an advantage of PRP over other injections in the 12-month follow-up. The main limitation of the study was in the variability of the applied PRP, including harvest blood volume, preparation method, and number of injections [85].

7. Platelets in OA—An Updated Meta-Analysis

In 2020 we published an article, “The Clinical Use of Platelet-Rich Plasma in Knee Disorders and Surgery—A Systematic Review and Meta-Analysis” [96]. In this article we have updated the chapter titled “osteoarthritis”.
A literature search of electronic databases identified a total of 386 records according to the selected search algorithm. There were 355 citations excluded as irrelevant, according to title and/or abstract. The abstracts of 31 remaining articles were assessed for eligibility. From these, 21 were excluded. The remaining 10 clinical studies published between 2020 and 2022, with a total of 974 patients, were included in this review. The literature search flowchart is shown in Figure 2.
Forty-eight studies (published between 2005 and 2022), including 3936 patients, evaluated the use of PRP in osteoarthritis treatment. Follow-up ranged from 6 months up to 2 years.
Thirty-seven studies compared PRP versus control groups [88,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132], six studies compared PRP with the addition of another substance (MSC or HA) versus control groups [122,123,133,134,135], six studies compared multiple injections of PRP [100,118,119,136,137,138], five studies compared PRGF-Endoret versus control groups [125,128,139,140,141], two studies compared autologous conditioned plasma (ACP) versus control groups [142,143], and one study compared intraosseous injection versus intra-articular injection versus the control group [108]. In twenty-nine studies, HA [101,103,104,106,107,108,109,110,112,113,114,115,116,117,118,120,122,123,125,126,127,128,132,136,139,141,143] was used as a control; in eleven studies placebo was used as a control (saline, no injection, physical therapy) [98,99,111,118,120,122,124,133,136,137,138]; in five studies corticosteroids [88,100,113,129,134] were used as the control; and in two studies acetaminophen [97,105] was used as the control. One study used peptides [132], ozone [128], dextrose prolotherapy [130], or bone marrow aspirate concentrate [127].
Thirty-two studies reported pain via the VAS comparing PRP versus placebo [97,98,99,117,123,130,135], dextrose [129] corticosteroids [88,100,128], or HA [101,102,103,104,106,107,108,109,110,113,116,118,122,124,125,126,127,131,132,138,143] (Figure 3). Placebo and HA subgroups showed significant differences in favour of PRP (p = 0.02; p < 0.00001); moreover, the steroid group showed significant differences in favour of PRP (p = 0.04), despite the dextrose subgroup showing significant differences in favour of the control group (p < 0.00001). The pooled estimates for these studies also showed significant differences in favour of PRP (p = 0.002).
Functional outcome was measured in thirty-five studies via the WOMAC scale. One study was excluded from the meta-analysis as a consequence of only reporting WOMAC pain scores [104]. Thirty-four studies compared PRP versus control groups: placebo [97,98,99,105,111,117,119,120,123,130,141], corticosteroids [100], dextrose [129], or HA [103,106,108,109,110,113,115,116,118,121,122,124,125,126,127,131,132,138,139,140,142] (Figure 4). The pooled estimates for these studies showed significant differences in favour of PRP (p < 0.00001); furthermore, each subgroup showed significant differences in favour of PRP (p < 0.00001).
Ten studies evaluated functional outcomes in IKDC rating scores, and showed significant differences in favour of PRP (p < 0.0001) (Figure 5). Seven studies showed significant differences in favour of PRP compared to HA as a control group (p = 0.0004) [101,104,107,115,125,126,143]; one study compared PRP with corticosteroids and [128] showed significant differences in favour of the experimental group (p < 0.0001); and two studies showed non-significant differences in favour of PRP when compared with the placebo (p = 0.24) [120,135].
Eight studies evaluated osteoarthritis outcomes via KOOS scores [88,101,102,114,126,130,133,134] (Figure 6). We excluded from the meta-analysis two of these studies due to a lack of measurements in one [98], and division of the results according to the physician in another [112]. The pooled estimates for these studies showed non-significant differences in KOOS for sport (p = 0.93), quality of life (p = 0.76), and ADL (p = 0.91); KOOS symptoms (p = 0.21) and pain (0.91) sub-scales were in favour of the control group (p > 0.05).
Functional outcomes were also measured with KSS scores in two studies [109,129]; Lysholm scores in three studies [104,109,123]; Tegner scores in four studies [101,109,133,143]; Outcome Measures in Arthritis Clinical Trials–Osteoarthritis Research Society International (OMERACT–OARSI) pain measure in four studies [98,131,140,141]; and Lequesne scores in nine studies [100,109,123,125,128,132,139,140,141]. Quality of life was measured with SF-36 scores in five studies [98,99,105,115], SF-12 in two studies [97,112], and European Quality of Life (EQoL) in two studies [102,112].
Thirty-two studies reported adverse events (Figure 7). Nine studies [98,99,111,118,120,121,124,131,142] comparing PRP versus placebo reported significant differences in favour of the control groups (p = 0.03); twenty studies [101,102,106,108,109,110,112,113,117,119,122,123,125,126,127,133,139,140,141,143] comparing PRP versus HA also reported significant differences in favour of the control groups (p = 0.003); furthermore, two studies [88,129] comparing PRP versus steroids (p = 0.002), and one study comparing PRP versus MSC [135] showed non-significant differences in favour of PRP (p = 0.60). The pooled estimates for these studies showed significant differences in favour of the control groups (p = 0.03).
Two studies were at high risk of bias for three domains [126,142], seven studies were at high risk of bias for two domains [105,113,116,118,129,130,137], and sixteen studies were at high risk of bias for one domain [100,103,107,108,109,110,112,114,117,121,123,124,128,131,135,138]. One study was at high risk of performance bias for three domains, with the risk of reporting bias for two domains [115], and seven studies had a lack of possibilities to define bias [99,105,113,115,119,123,127] (Figure 8).

8. Materials and Methods

8.1. Search Strategy

In this review we concentrated on PRP applications to knee osteoarthritis compared with placebo or other treatment control groups. This study was completed in compatibility with the 2009 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. A systematic review of the use of platelet-rich plasma in knee lesions was completed with a comprehensive published literature search in PubMed, Embase, Cochrane Database of Systematic Reviews, and Clinicaltrials.gov. The references of the investigations found in this search were cross-referenced to identify additional pertinent studies not identified in the original searches. All searches were performed in January 2022. The searches were performed combining the following keywords: (1) “PRP”, or “platelet-rich plasma”, or “plasma rich in growth factors”, or “platelet derived growth factor”, or “platelet derived”, or “platelet gel”, or “platelet concentrate”, or “PRF”, or “platelet rich fibrin”, or “ACP”, or ”autologous conditioned plasma”, or ”PRGF”, or “platelet lysate”; and (2) “knee osteoarthritis” or “osteoarthritis”. This was an update on a systematic review registered with PROSPERO (International Prospective Register of Systematic Reviews, ID 167715).

8.2. Inclusion and Exclusion Criteria

This review included all clinical studies meeting the following inclusion criteria: PRP utilization as conservative treatment in knee osteoarthritis, English language, human subjects, paper published in a peer-reviewed journal, and full text available. Only randomized controlled trials were included. Exclusion criteria included all animal studies, basic scientific investigations, case reports, review articles, expert opinions, letters to editor, studies without control groups, studies not using PRP, papers not peer reviewed, papers not in English, trials evaluating platelet-poor plasma, and investigations on other diseases unrelated to the knee joint. The investigations included in this study were independently reviewed by two orthopedic surgeons/authors for inclusion and exclusion criteria.

8.3. Types of Interventions

We compared intralesional, injected PRP preparation with the following:
-
Placebo injection (low-volume saline injection, matching the PRP volume);
-
High-volume saline image-guided injection;
-
Local steroidal injection;
-
Hyaluronic acid injection;
-
Exercise and other physical therapies (e.g., low-dose radiation therapy, eccentric loading program, dry needling);
-
Any other medications administered locally or systemically aimed at treating pain; and
-
Combinations of the active interventions listed above.

8.4. Outcomes

Primary outcomes included the following:
-
Pain as measured by standard validated pain scale, such as visual analogue score (VAS), EQ-VAS, or numerical rating scale (NRS);
-
Functional measurement by any standard validated scale, such as the International Knee Documentation Committee (IKDC), Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), Knee Society Score (KSS), Victorian Institute of Sport Assessment (VISA), 36-Item Short Form Survey (SF-36), Knee injury and Osteoarthritis Outcome Score (KOOS), Lysholm Knee Scoring Scale, Tegner Activity Score, and Ikeuchi grade knee rating scale.

8.5. Data Collection and Analysis

For each study included in the analysis, the following data were extracted by two independent reviewers: authors, year of publication, type of knee lesions, details of interventions in the study, sample size (randomized and analysed), outcome measurements, follow-up period, main results, and percentage and type of adverse events included in the publication. Each study’s level of evidence was examined and evaluated based on criteria established by the Oxford Centre for Evidence-Based Medicine Levels of Evidence Working Group [109]. Measures of treatment effect at the final point used were the means and standard deviations for continuous outcome measures. When studies reported other measures (e.g., median) and other dispersion measures, such as standard error (SE) of the mean or 95% CI of the mean, range or interquartile range (IQR), we calculated the SD in order to perform the relevant meta-analytical pooling according to previous studies [144,145].
The study weight was calculated using the Mantel–Haenszel method. We assessed statistical heterogeneity using Tau2 or Chi2, df, and I2 statistics. The I2 statistic describes the percentage of total variation across trials that is a result of heterogeneity. In the case of low heterogeneity (I2 < 40%), studies were pooled using a fixed-effects model; otherwise, a random-effects analysis was performed.
Subgroup analysis was undertaken for the type of clinical trial and for the type of control intervention.

8.6. Risk of Bias Assessment

Revised Cochrane risk-of-bias tool was used to evaluate risk. Disagreement in the risk of bias assessment was resolved by consensus and, if necessary, by the opinion of a third reviewer. A study was deemed to be:
-
“Low risk” if all items were scored as “low risk”;
-
“Moderate risk” if up to two items were classified as “high risk” or “unclear risk”;
-
“High risk” if more than two items were scored as “high risk”.
We presented our assessment of risk of bias using two “Risk of Bias” summary figures for every subsection of the manuscript.

8.7. Statistical Analysis

Qualitative statistical analysis and meta-analysis were performed using R software and REVMAN 5.4 [145,146], with p-values of <0.05.

9. Conclusions

OA is a degenerative joint disease primarily associated with aging. Since affected patients are mainly characterized by cartilage malfunction, inflammation plays an essential role in the pathogenesis of OA. Although there are many well-described molecular mechanisms and mediators involved in inflammation, it still remains controversial whether inflammatory mediators are primary or secondary regulators of the cartilage damage and disturbed repair processes in OA. The latest knowledge about the activation of inflammatory mediators could play a significant role in the early diagnosis of inflammation in OA and become potentially useful in therapeutic protocols.
Given the pivotal role of platelets in hemostasis, inflammation, and the immune response, it can be assumed that platelets also contribute to inflammation in OA; however, their exact role in this process requires further investigation.
Joint diffusion as a result of inflammation is commonly observed in other inflammatory arthropathies, including RA; in contrast to OA, the role of platelets in RA has been established. In RA, the platelet count is related to disease activity, and thrombocytosis is commonly detected in the active phase of the disease.
Of special interest are the multiple mechanisms involved in platelet activation in osteoarthritis, which offer numerous options for potential arthritis therapies.
As stated in this review, the role of platelet-rich plasma is a well described, commonly used, minimally invasive method for knee osteoarthritis treatment. Multiple randomized controlled trials described PRP as a safe, easily accessible, and low-cost procedure as an alternative to other non-surgical treatments for osteoarthritis used as various control groups. There is still a need for a standardized PRP injection to be established from the multitude of products available for medical usage. Moreover, there is still a lack of studies for using PRP as an alternative to surgical treatment, since the majority of trials only compare PRP to other intra-articular injections. Platelets may have a potential healing role as a result of their proinflammatory and anti-inflammatory functions.

Author Contributions

Conceptualization, R.K.; data curation, K.M., E.T., and R.K.; formal analysis, R.K.; funding acquisition, R.K. and S.P.; investigation E.T. and K.M.; methodology, R.K., and S.P.; project administration, E.T.; resources, R.K., software, R.K.; supervision, R.K. and S.P.; writing—review and editing, R.K., E.T., K.M., and S.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Centre of Postgraduate Medical Education Grant, grant number 501-1-007-18-21.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nelson, A.E. Osteoarthritis Year in Review 2017: Clinical. Osteoarthr. Cartil. 2018, 26, 319–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Vina, E.R.; Kwoh, C.K. Epidemiology of Osteoarthritis: Literature Update. Curr. Opin. Rheumatol. 2018, 30, 160–167. [Google Scholar] [CrossRef]
  3. Abramoff, B.; Caldera, F.E. Osteoarthritis. Med. Clin. N. Am. 2020, 104, 293–311. [Google Scholar] [CrossRef] [PubMed]
  4. Hunter, D.J.; Bierma-Zeinstra, S. Osteoarthritis. Lancet 2019, 393, 1745–1759. [Google Scholar] [CrossRef]
  5. O’Neill, T.W.; McCabe, P.S.; McBeth, J. Update on the Epidemiology, Risk Factors and Disease Outcomes of Osteoarthritis. Best Pract. Res. Clin. Rheumatol. 2018, 32, 312–326. [Google Scholar] [CrossRef]
  6. Hurley, M.V.; Scott, D.L.; Rees, J.; Newham, D.J. Sensorimotor Changes and Functional Performance in Patients with Knee Osteoarthritis. Ann. Rheum. Dis. 1997, 56, 641–648. [Google Scholar] [CrossRef]
  7. He, Y.; Li, Z.; Alexander, P.G.; Ocasio-Nieves, B.D.; Yocum, L.; Lin, H.; Tuan, R.S. Pathogenesis of Osteoarthritis: Risk Factors, Regulatory Pathways in Chondrocytes, and Experimental Models. Biology 2020, 9, 194. [Google Scholar] [CrossRef]
  8. Bortoluzzi, A.; Furini, F.; Scirè, C.A. Osteoarthritis and Its Management—Epidemiology, Nutritional Aspects and Environmental Factors. Autoimmun. Rev. 2018, 17, 1097–1104. [Google Scholar] [CrossRef]
  9. Pigeolet, M.; Jayaram, A.; Park, K.B.; Meara, J.G. Osteoarthritis in 2020 and Beyond. Lancet 2021, 397, 1059–1060. [Google Scholar] [CrossRef]
  10. Glyn-Jones, S.; Palmer, A.J.R.; Agricola, R.; Price, A.J.; Vincent, T.L.; Weinans, H.; Carr, A.J. Osteoarthritis. Lancet 2015, 386, 376–387. [Google Scholar] [CrossRef]
  11. Taruc-Uy, R.L.; Lynch, S.A. Diagnosis and Treatment of Osteoarthritis. Prim. Care Clin. Off. Pract. 2013, 40, 821–836. [Google Scholar] [CrossRef] [PubMed]
  12. van den Bosch, M.H.J. Osteoarthritis Year in Review 2020: Biology. Osteoarthr. Cartil. 2021, 29, 143–150. [Google Scholar] [CrossRef]
  13. Primorac, D.; Molnar, V.; Rod, E.; Jeleč, Ž.; Čukelj, F.; Matišić, V.; Vrdoljak, T.; Hudetz, D.; Hajsok, H.; Borić, I. Knee Osteoarthritis: A Review of Pathogenesis and State-Of-The-Art Non-Operative Therapeutic Considerations. Genes 2020, 11, 854. [Google Scholar] [CrossRef] [PubMed]
  14. Tonge, D.P.; Pearson, M.J.; Jones, S.W. The Hallmarks of Osteoarthritis and the Potential to Develop Personalised Disease-Modifying Pharmacological Therapeutics. Osteoarthr. Cartil. 2014, 22, 609–621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Schulze-Tanzil, G. Experimental Therapeutics for the Treatment of Osteoarthritis. JEP 2021, 13, 101–125. [Google Scholar] [CrossRef] [PubMed]
  16. Kim, J.-R.; Yoo, J.; Kim, H. Therapeutics in Osteoarthritis Based on an Understanding of Its Molecular Pathogenesis. Int. J. Mol. Sci. 2018, 19, 30674. [Google Scholar] [CrossRef] [Green Version]
  17. Sellam, J.; Berenbaum, F. The Role of Synovitis in Pathophysiology and Clinical Symptoms of Osteoarthritis. Nat. Rev. Rheumatol. 2010, 6, 625–635. [Google Scholar] [CrossRef]
  18. Robinson, W.H.; Lepus, C.M.; Wang, Q.; Raghu, H.; Mao, R.; Lindstrom, T.M.; Sokolove, J. Low-Grade Inflammation as a Key Mediator of the Pathogenesis of Osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 580–592. [Google Scholar] [CrossRef]
  19. Daghestani, H.N.; Kraus, V.B. Inflammatory Biomarkers in Osteoarthritis. Osteoarthr. Cartil. 2015, 23, 1890–1896. [Google Scholar] [CrossRef] [Green Version]
  20. Van den Bosch, M.H.J.; van Lent, P.L.E.M.; van der Kraan, P.M. Identifying Effector Molecules, Cells, and Cytokines of Innate Immunity in OA. Osteoarthr. Cartil. 2020, 28, 532–543. [Google Scholar] [CrossRef]
  21. Liu-Bryan, R. Synovium and the Innate Inflammatory Network in Osteoarthritis Progression. Curr. Rheumatol. Rep. 2013, 15, 323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Goldring, M.B.; Otero, M. Inflammation in Osteoarthritis. Curr. Opin. Rheumatol. 2011, 23, 471–478. [Google Scholar] [CrossRef] [PubMed]
  23. Sohn, D.; Sokolove, J.; Sharpe, O.; Erhart, J.C.; Chandra, P.E.; Lahey, L.J.; Lindstrom, T.M.; Hwang, I.; Boyer, K.A.; Andriacchi, T.P.; et al. Plasma Proteins Present in Osteoarthritic Synovial Fluid Can Stimulate Cytokine Production via Toll-like Receptor 4. Arthritis Res. Ther. 2012, 14, R7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Wang, Q.; Rozelle, A.L.; Lepus, C.M.; Scanzello, C.R.; Song, J.J.; Larsen, D.M.; Crish, J.F.; Bebek, G.; Ritter, S.Y.; Lindstrom, T.M.; et al. Identification of a Central Role for Complement in Osteoarthritis. Nat. Med. 2011, 17, 1674–1679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Silawal, S.; Triebel, J.; Bertsch, T.; Schulze-Tanzil, G. Osteoarthritis and the Complement Cascade. Clin. Med. Insights Arthritis Musculoskelet. Disord. 2018, 11, 117954411775143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Sofat, N. Analysing the Role of Endogenous Matrix Molecules in the Development of Osteoarthritis. Int. J. Exp. Pathol. 2009, 90, 463–479. [Google Scholar] [CrossRef]
  27. Loeser, R.F.; Goldring, S.R.; Scanzello, C.R.; Goldring, M.B. Osteoarthritis: A Disease of the Joint as an Organ. Arthritis Rheum. 2012, 64, 1697–1707. [Google Scholar] [CrossRef] [Green Version]
  28. Troeberg, L.; Nagase, H. Proteases Involved in Cartilage Matrix Degradation in Osteoarthritis. Biochim. Biophys. Acta (BBA) Proteins Proteom. 2012, 1824, 133–145. [Google Scholar] [CrossRef] [Green Version]
  29. Loeser, R.F. Osteoarthritis Year in Review 2013: Biology. Osteoarthr. Cartil. 2013, 21, 1436–1442. [Google Scholar] [CrossRef] [Green Version]
  30. Luyten, F.P.; Tylzanowski, P.; Lories, R.J. Wnt Signaling and Osteoarthritis. Bone 2009, 44, 522–527. [Google Scholar] [CrossRef]
  31. Blaney Davidson, E.N.; Vitters, E.L.; Bennink, M.B.; van Lent, P.L.E.M.; van Caam, A.P.M.; Blom, A.B.; van den Berg, W.B.; van de Loo, F.A.J.; van der Kraan, P.M. Inducible Chondrocyte-Specific Overexpression of BMP2 in Young Mice Results in Severe Aggravation of Osteophyte Formation in Experimental OA without Altering Cartilage Damage. Ann. Rheum. Dis. 2015, 74, 1257–1264. [Google Scholar] [CrossRef] [PubMed]
  32. Blom, A.B.; Brockbank, S.M.; van Lent, P.L.; van Beuningen, H.M.; Geurts, J.; Takahashi, N.; van der Kraan, P.M.; van de Loo, F.A.; Schreurs, B.W.; Clements, K.; et al. Involvement of the Wnt Signaling Pathway in Experimental and Human Osteoarthritis: Prominent Role of Wnt-Induced Signaling Protein 1. Arthritis Rheum. 2009, 60, 501–512. [Google Scholar] [CrossRef] [PubMed]
  33. Gabay, O.; Sanchez, C. Epigenetics, Sirtuins and Osteoarthritis. Jt. Bone Spine 2012, 79, 570–573. [Google Scholar] [CrossRef] [PubMed]
  34. Tsezou, A. Osteoarthritis Year in Review 2014: Genetics and Genomics. Osteoarthr. Cartil. 2014, 22, 2017–2024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Shen, J.; Abu-Amer, Y.; O’Keefe, R.J.; McAlinden, A. Inflammation and Epigenetic Regulation in Osteoarthritis. Connect. Tissue Res. 2017, 58, 49–63. [Google Scholar] [CrossRef] [PubMed]
  36. Thomas, M.; Storey, R. The Role of Platelets in Inflammation. Thromb. Haemost. 2015, 114, 449–458. [Google Scholar] [CrossRef]
  37. Nurden, A.T. The Biology of the Platelet with Special Reference to Inflammation Wound Healing and Immunity. Front. Biosci. 2018, 23, 726–751. [Google Scholar] [CrossRef] [Green Version]
  38. Repsold, L.; Joubert, A.M. Platelet Function, Role in Thrombosis, Inflammation, and Consequences in Chronic Myeloproliferative Disorders. Cells 2021, 10, 3034. [Google Scholar] [CrossRef]
  39. Yun, S.-H.; Sim, E.-H.; Goh, R.-Y.; Park, J.-I.; Han, J.-Y. Platelet Activation: The Mechanisms and Potential Biomarkers. Biomed. Res. Int. 2016, 2016, 9060143. [Google Scholar] [CrossRef] [Green Version]
  40. Guo, L.; Rondina, M.T. The Era of Thromboinflammation: Platelets Are Dynamic Sensors and Effector Cells During Infectious Diseases. Front. Immunol. 2019, 10, 2204. [Google Scholar] [CrossRef] [Green Version]
  41. McDonald, B.; Dunbar, M. Platelets and Intravascular Immunity: Guardians of the Vascular Space During Bloodstream Infections and Sepsis. Front. Immunol. 2019, 10, 2400. [Google Scholar] [CrossRef] [PubMed]
  42. Jenne, C.N.; Kubes, P. Platelets in Inflammation and Infection. Platelets 2015, 26, 286–292. [Google Scholar] [CrossRef] [PubMed]
  43. Manne, B.K.; Xiang, S.C.; Rondina, M.T. Platelet Secretion in Inflammatory and Infectious Diseases. Platelets 2017, 28, 155–164. [Google Scholar] [CrossRef] [PubMed]
  44. Sonmez, O.; Sonmez, M. Role of Platelets in Immune System and Inflammation. Porto Biomed. J. 2017, 2, 311–314. [Google Scholar] [CrossRef] [PubMed]
  45. Duerschmied, D.; Bode, C.; Ahrens, I. Immune Functions of Platelets. Thromb. Haemost. 2014, 112, 678–691. [Google Scholar] [CrossRef] [Green Version]
  46. Morrell, C.N.; Aggrey, A.A.; Chapman, L.M.; Modjeski, K.L. Emerging Roles for Platelets as Immune and Inflammatory Cells. Blood 2014, 123, 2759–2767. [Google Scholar] [CrossRef] [Green Version]
  47. Herter, J.M.; Rossaint, J.; Zarbock, A. Platelets in Inflammation and Immunity. J. Thromb. Haemost. 2014, 12, 1764–1775. [Google Scholar] [CrossRef]
  48. Nurden, A. Platelets, Inflammation and Tissue Regeneration. Thromb. Haemost. 2011, 105, S13–S33. [Google Scholar] [CrossRef]
  49. Andia, I.; Sánchez, M.; Maffulli, N. Joint Pathology and Platelet-Rich Plasma Therapies. Expert Opin. Biol. Ther. 2012, 12, 7–22. [Google Scholar] [CrossRef]
  50. Alunno, A.; Falcinelli, E.; Luccioli, F.; Petito, E.; Bartoloni, E.; Momi, S.; Mirabelli, G.; Mancini, G.; Gerli, R.; Gresele, P. Platelets Contribute to the Accumulation of Matrix Metalloproteinase Type 2 in Synovial Fluid in Osteoarthritis. Thromb. Haemost. 2017, 117, 2116–2124. [Google Scholar] [CrossRef] [Green Version]
  51. Gaertner, F.; Massberg, S. Patrolling the Vascular Borders: Platelets in Immunity to Infection and Cancer. Nat. Rev. Immunol. 2019, 19, 747–760. [Google Scholar] [CrossRef] [PubMed]
  52. Cafaro, G.; Bartoloni, E.; Alunno, A.; Bistoni, O.; Cipriani, S.; Topini, F.; Gerli, R. A Platelet’s Guide to Synovitis. Isr. Med. Assoc. J. 2019, 21, 454–459. [Google Scholar] [PubMed]
  53. Tokarz-Deptuła, B.; Palma, J.; Baraniecki, Ł.; Stosik, M.; Kołacz, R.; Deptuła, W. What Function Do Platelets Play in Inflammation and Bacterial and Viral Infections? Front. Immunol. 2021, 12, 770436. [Google Scholar] [CrossRef] [PubMed]
  54. Kwon, Y.-J.; Koh, I.-H.; Chung, K.; Lee, Y.-J.; Kim, H.-S. Association between Platelet Count and Osteoarthritis in Women Older than 50 Years. Ther. Adv. Musculoskelet. 2020, 12, 1759720X2091286. [Google Scholar] [CrossRef]
  55. Hall, A.J.; Stubbs, B.; Mamas, M.A.; Myint, P.K.; Smith, T.O. Association between Osteoarthritis and Cardiovascular Disease: Systematic Review and Meta-Analysis. Eur. J. Prev. Cardiol. 2016, 23, 938–946. [Google Scholar] [CrossRef]
  56. Wojdasiewicz, P.; Poniatowski, Ł.A.; Szukiewicz, D. The Role of Inflammatory and Anti-Inflammatory Cytokines in the Pathogenesis of Osteoarthritis. Mediat. Inflamm. 2014, 2014, 561459. [Google Scholar] [CrossRef] [Green Version]
  57. Taşoğlu, Ö.; Şahin, A.; Karataş, G.; Koyuncu, E.; Taşoğlu, İ.; Tecimel, O.; Özgirgin, N. Blood Mean Platelet Volume and Platelet Lymphocyte Ratio as New Predictors of Hip Osteoarthritis Severity. Medicine 2017, 96, e6073. [Google Scholar] [CrossRef]
  58. Mabey, T. Cytokines as Biochemical Markers for Knee Osteoarthritis. WJO 2015, 6, 95. [Google Scholar] [CrossRef]
  59. Stannus, O.; Jones, G.; Cicuttini, F.; Parameswaran, V.; Quinn, S.; Burgess, J.; Ding, C. Circulating Levels of IL-6 and TNF-α Are Associated with Knee Radiographic Osteoarthritis and Knee Cartilage Loss in Older Adults. Osteoarthr. Cartil. 2010, 18, 1441–1447. [Google Scholar] [CrossRef] [Green Version]
  60. Maślanka, K. Udział Płytek Krwi w Procesach Zapalnych. J. Transfus. Med. 2014, 7, 102–109. [Google Scholar]
  61. Boilard, E.; Blanco, P.; Nigrovic, P.A. Platelets: Active Players in the Pathogenesis of Arthritis and SLE. Nat. Rev. Rheumatol. 2012, 8, 534–542. [Google Scholar] [CrossRef] [PubMed]
  62. Harifi, G.; Sibilia, J. Pathogenic Role of Platelets in Rheumatoid Arthritis and Systemic Autoimmune Diseases: Perspectives and Therapeutic Aspects. SMJ 2016, 37, 354–360. [Google Scholar] [CrossRef] [PubMed]
  63. Olumuyiwa-Akeredolu, O.; Page, M.J.; Soma, P.; Pretorius, E. Platelets: Emerging Facilitators of Cellular Crosstalk in Rheumatoid Arthritis. Nat. Rev. Rheumatol. 2019, 15, 237–248. [Google Scholar] [CrossRef] [PubMed]
  64. Gasparyan, A.Y.; Stavropoulos-Kalinoglou, A.; Mikhailidis, D.P.; Douglas, K.M.J.; Kitas, G.D. Platelet Function in Rheumatoid Arthritis: Arthritic and Cardiovascular Implications. Rheumatol. Int. 2011, 31, 153–164. [Google Scholar] [CrossRef]
  65. Jin, K.; Luo, Z.; Zhang, B.; Pang, Z. Biomimetic Nanoparticles for Inflammation Targeting. Acta Pharm. Sin. B 2018, 8, 23–33. [Google Scholar] [CrossRef] [PubMed]
  66. Boilard, E.; Nigrovic, P.A.; Larabee, K.; Watts, G.F.M.; Coblyn, J.S.; Weinblatt, M.E.; Massarotti, E.M.; Remold-O’Donnell, E.; Farndale, R.W.; Ware, J.; et al. Platelets Amplify Inflammation in Arthritis via Collagen-Dependent Microparticle Production. Science 2010, 327, 580–583. [Google Scholar] [CrossRef] [Green Version]
  67. Tunjungputri, R.; Li, Y.; de Groot, P.; Dinarello, C.; Smeekens, S.; Jaeger, M.; Doppenberg-Oosting, M.; Cruijsen, M.; Lemmers, H.; Toenhake-Dijkstra, H.; et al. The Inter-Relationship of Platelets with Interleukin-1β-Mediated Inflammation in Humans. Thromb. Haemost. 2018, 118, 2112–2125. [Google Scholar] [CrossRef]
  68. Kehrel, B. Platelet-Collagen Interactions. Semin. Thromb. Hemost. 1995, 21, 123–129. [Google Scholar] [CrossRef]
  69. Qiao, J.; Arthur, J.F.; Gardiner, E.E.; Andrews, R.K.; Zeng, L.; Xu, K. Regulation of Platelet Activation and Thrombus Formation by Reactive Oxygen Species. Redox Biol. 2018, 14, 126–130. [Google Scholar] [CrossRef]
  70. Riyazi, N.; Slagboom, E.; de Craen, A.J.M.; Meulenbelt, I.; Houwing-Duistermaat, J.J.; Kroon, H.M.; van Schaardenburg, D.; Rosendaal, F.R.; Breedveld, F.C.; Huizinga, T.W.J.; et al. Association of the Risk of Osteoarthritis with High Innate Production of Interleukin-1β and Low Innate Production of Interleukin-10 Ex Vivo, upon Lipopolysaccharide Stimulation. Arthritis Rheum. 2005, 52, 1443–1450. [Google Scholar] [CrossRef]
  71. Wang, J.; Sun, H.; Fu, Z.; Liu, M. Chondroprotective Effects of Alpha-Lipoic Acid in a Rat Model of Osteoarthritis. Free Radic. Res. 2016, 50, 767–780. [Google Scholar] [CrossRef] [PubMed]
  72. Radomski, M.W.; Palmer, R.M.J.; Moncada, S. Characterization of the L-Arginine: Nitric Oxide Pathway in Human Platelets. Br. J. Pharmacol. 1990, 101, 325–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ning, L.; Ishijima, M.; Kaneko, H.; Kurihara, H.; Arikawa-Hirasawa, E.; Kubota, M.; Liu, L.; Xu, Z.; Futami, I.; Yusup, A.; et al. Correlations between Both the Expression Levels of Inflammatory Mediators and Growth Factor in Medial Perimeniscal Synovial Tissue and the Severity of Medial Knee Osteoarthritis. Int. Orthop. 2011, 35, 831–838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Vincent, H.K.; Percival, S.S.; Conrad, B.P.; Seay, A.N.; Montero, C.; Vincent, K.R. Hyaluronic Acid (HA) Viscosupplementation on Synovial Fluid Inflammation in Knee Osteoarthritis: A Pilot Study. TOORTHJ 2013, 7, 378–384. [Google Scholar] [CrossRef] [PubMed]
  75. Bester, J.; Pretorius, E. Effects of IL-1β, IL-6 and IL-8 on Erythrocytes, Platelets and Clot Viscoelasticity. Sci. Rep. 2016, 6, 32188. [Google Scholar] [CrossRef] [PubMed]
  76. Reeh, H.; Rudolph, N.; Billing, U.; Christen, H.; Streif, S.; Bullinger, E.; Schliemann-Bullinger, M.; Findeisen, R.; Schaper, F.; Huber, H.J.; et al. Response to IL-6 Trans- and IL-6 Classic Signalling Is Determined by the Ratio of the IL-6 Receptor α to Gp130 Expression: Fusing Experimental Insights and Dynamic Modelling. Cell Commun. Signal. 2019, 17, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Senchenkova, E.Y.; Komoto, S.; Russell, J.; Almeida-Paula, L.D.; Yan, L.-S.; Zhang, S.; Granger, D.N. Interleukin-6 Mediates the Platelet Abnormalities and Thrombogenesis Associated with Experimental Colitis. Am. J. Pathol. 2013, 183, 173–181. [Google Scholar] [CrossRef] [Green Version]
  78. Williams, N.; Bertoncello, I.; Jackson, H.; Arnold, J.; Kavnoudias, H. The Role of Interleukin 6 in Megakaryocyte Formation, Megakaryocyte Development and Platelet Production. In Novartis Foundation Symposia; Bock, G.R., Marsh, J., Widdows, K., Eds.; John Wiley & Sons, Ltd.: Chichester, UK, 2007; pp. 160–173. ISBN 978-0-470-51426-9. [Google Scholar]
  79. Porée, B.; Kypriotou, M.; Chadjichristos, C.; Beauchef, G.; Renard, E.; Legendre, F.; Melin, M.; Gueret, S.; Hartmann, D.-J.; Malléin-Gerin, F.; et al. Interleukin-6 (IL-6) and/or Soluble IL-6 Receptor Down-Regulation of Human Type II Collagen Gene Expression in Articular Chondrocytes Requires a Decrease of Sp1·Sp3 Ratio and of the Binding Activity of Both Factors to the COL2A1 Promoter. J. Biol. Chem. 2008, 283, 4850–4865. [Google Scholar] [CrossRef] [Green Version]
  80. Livshits, G.; Zhai, G.; Hart, D.J.; Kato, B.S.; Wang, H.; Williams, F.M.K.; Spector, T.D. Interleukin-6 Is a Significant Predictor of Radiographic Knee Osteoarthritis: The Chingford Study. Arthritis Rheum. 2009, 60, 2037–2045. [Google Scholar] [CrossRef] [Green Version]
  81. Balbaloglu, O.; Korkmaz, M.; Yolcu, S.; Karaaslan, F.; Beceren, N.G.Ç. Evaluation of Mean Platelet Volume (MPV) Levels in Patients with Synovitis Associated with Knee Osteoarthritis. Platelets 2014, 25, 81–85. [Google Scholar] [CrossRef]
  82. Penninx, B.W.J.H.; Abbas, H.; Ambrosius, W.; Nicklas, B.J.; Davis, C.; Messier, S.P.; Pahor, M. Inflammatory Markers and Physical Function among Older Adults with Knee Osteoarthritis. J. Rheumatol. 2004, 31, 2027–2031. [Google Scholar] [PubMed]
  83. Eppley, B.L.; Woodell, J.E.; Higgins, J. Platelet Quantification and Growth Factor Analysis from Platelet-Rich Plasma: Implications for Wound Healing. Plast. Reconstr. Surg. 2004, 1502–1508. [Google Scholar] [CrossRef] [PubMed]
  84. Dohan Ehrenfest, D.M.; Bielecki, T.; Mishra, A.; Borzini, P.; Inchingolo, F.; Sammartino, G.; Rasmusson, L.; Evert, P.A. In Search of a Consensus Terminology in the Field of Platelet Concentrates for Surgical Use: Platelet-Rich Plasma (PRP), Platelet-Rich Fibrin (PRF), Fibrin Gel Polymerization and Leukocytes. CPB 2012, 13, 1131–1137. [Google Scholar] [CrossRef] [Green Version]
  85. Kon, E.; Di Matteo, B.; Delgado, D.; Cole, B.J.; Dorotei, A.; Dragoo, J.L.; Filardo, G.; Fortier, L.A.; Giuffrida, A.; Jo, C.H.; et al. Platelet-Rich Plasma for the Treatment of Knee Osteoarthritis: An Expert Opinion and Proposal for a Novel Classification and Coding System. Expert Opin. Biol. Ther. 2020, 20, 1447–1460. [Google Scholar] [CrossRef] [PubMed]
  86. Wanstrath, A.W.; Hettlich, B.F.; Su, L.; Smith, A.; Zekas, L.J.; Allen, M.J.; Bertone, A.L. Evaluation of a Single Intra-Articular Injection of Autologous Protein Solution for Treatment of Osteoarthritis in a Canine Population: Autologous Protein Solution for Treatment of Canine Osteoarthritis. Vet. Surg. 2016, 45, 764–774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Osterman, C.; McCarthy, M.B.R.; Cote, M.P.; Beitzel, K.; Bradley, J.; Polkowski, G.; Mazzocca, A.D. Platelet-Rich Plasma Increases Anti-Inflammatory Markers in a Human Coculture Model for Osteoarthritis. Am. J. Sports Med. 2015, 43, 1474–1484. [Google Scholar] [CrossRef] [PubMed]
  88. Joshi Jubert, N.; Rodríguez, L.; Reverté-Vinaixa, M.M.; Navarro, A. Platelet-Rich Plasma Injections for Advanced Knee Osteoarthritis: A Prospective, Randomized, Double-Blinded Clinical Trial. Orthop. J. Sports Med. 2017, 5, 232596711668938. [Google Scholar] [CrossRef]
  89. Re’em, T.; Kaminer-Israeli, Y.; Ruvinov, E.; Cohen, S. Chondrogenesis of HMSC in Affinity-Bound TGF-Beta Scaffolds. Biomaterials 2012, 33, 751–761. [Google Scholar] [CrossRef]
  90. Schmidt, M.B.; Chen, E.H.; Lynch, S.E. A Review of the Effects of Insulin-like Growth Factor and Platelet Derived Growth Factor on in Vivo Cartilage Healing and Repair. Osteoarthr. Cartil. 2006, 14, 403–412. [Google Scholar] [CrossRef] [Green Version]
  91. Solchaga, L.A.; Penick, K.; Porter, J.D.; Goldberg, V.M.; Caplan, A.I.; Welter, J.F. FGF-2 Enhances the Mitotic and Chondrogenic Potentials of Human Adult Bone Marrow-Derived Mesenchymal Stem Cells. J. Cell. Physiol. 2005, 203, 398–409. [Google Scholar] [CrossRef]
  92. Garbin, L.C.; Olver, C.S. Platelet-Rich Products and Their Application to Osteoarthritis. J. Equine Vet. Sci. 2020, 86, 102820. [Google Scholar] [CrossRef] [PubMed]
  93. Sundman, E.A.; Cole, B.J.; Karas, V.; Della Valle, C.; Tetreault, M.W.; Mohammed, H.O.; Fortier, L.A. The Anti-Inflammatory and Matrix Restorative Mechanisms of Platelet-Rich Plasma in Osteoarthritis. Am. J. Sports Med. 2014, 42, 35–41. [Google Scholar] [CrossRef] [PubMed]
  94. Lacko, M.; Harvanová, D.; Slovinská, L.; Matuška, M.; Balog, M.; Lacková, A.; Špaková, T.; Rosocha, J. Effect of Intra-Articular Injection of Platelet-Rich Plasma on the Serum Levels of Osteoarthritic Biomarkers in Patients with Unilateral Knee Osteoarthritis. JCM 2021, 10, 5801. [Google Scholar] [CrossRef]
  95. Tang, J.Z.; Nie, M.J.; Zhao, J.Z.; Zhang, G.C.; Zhang, Q.; Wang, B. Platelet-Rich Plasma versus Hyaluronic Acid in the Treatment of Knee Osteoarthritis: A Meta-Analysis. J. Orthop. Surg. Res. 2020, 15, 403. [Google Scholar] [CrossRef]
  96. Trams, E.; Kulinski, K.; Kozar-Kaminska, K.; Pomianowski, S.; Kaminski, R. The Clinical Use of Platelet-Rich Plasma in Knee Disorders and Surgery—A Systematic Review and Meta-Analysis. Life 2020, 10, 94. [Google Scholar] [CrossRef] [PubMed]
  97. Simental-Mendía, M.; Vílchez-Cavazos, J.F.; Peña-Martínez, V.M.; Said-Fernández, S.; Lara-Arias, J.; Martínez-Rodríguez, H.G. Leukocyte-Poor Platelet-Rich Plasma Is More Effective than the Conventional Therapy with Acetaminophen for the Treatment of Early Knee Osteoarthritis. Arch. Orthop. Trauma Surg. 2016, 136, 1723–1732. [Google Scholar] [CrossRef]
  98. Kon, E.; Engebretsen, L.; Verdonk, P.; Nehrer, S.; Filardo, G. Clinical Outcomes of Knee Osteoarthritis Treated With an Autologous Protein Solution Injection: A 1-Year Pilot Double-Blinded Randomized Controlled Trial. Am. J. Sports Med. 2018, 46, 171–180. [Google Scholar] [CrossRef]
  99. Elik, H.; Doğu, B.; Yılmaz, F.; Begoğlu, F.A.; Kuran, B. The Efficiency of Platelet-Rich Plasma Treatment in Patients with Knee Osteoarthritis. J. Back Musculoskelet. Rehabil. 2020, 33, 127–138. [Google Scholar] [CrossRef]
  100. Uslu Güvendi, E.; Aşkin, A.; Güvendi, G.; Koçyiğit, H. Comparison of Efficiency Between Corticosteroid and Platelet Rich Plasma Injection Therapies in Patients with Knee Osteoarthritis. Arch. Rheumatol. 2018, 33, 273–281. [Google Scholar] [CrossRef]
  101. Filardo, G.; Di Matteo, B.; Di Martino, A.; Merli, M.L.; Cenacchi, A.; Fornasari, P.; Marcacci, M.; Kon, E. Platelet-Rich Plasma Intra-Articular Knee Injections Show No Superiority Versus Viscosupplementation: A Randomized Controlled Trial. Am. J. Sports Med. 2015, 43, 1575–1582. [Google Scholar] [CrossRef]
  102. Paterson, K.L.; Nicholls, M.; Bennell, K.L.; Bates, D. Intra-Articular Injection of Photo-Activated Platelet-Rich Plasma in Patients with Knee Osteoarthritis: A Double-Blind, Randomized Controlled Pilot Study. BMC Musculoskelet. Disord. 2016, 17, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Duymus, T.M.; Mutlu, S.; Dernek, B.; Komur, B.; Aydogmus, S.; Kesiktas, F.N. Choice of Intra-Articular Injection in Treatment of Knee Osteoarthritis: Platelet-Rich Plasma, Hyaluronic Acid or Ozone Options. Knee Surg. Sports Traumatol. Arthrosc. 2017, 25, 485–492. [Google Scholar] [CrossRef] [PubMed]
  104. Cole, B.J.; Karas, V.; Hussey, K.; Merkow, D.B.; Pilz, K.; Fortier, L.A. Hyaluronic Acid Versus Platelet-Rich Plasma: A Prospective, Double-Blind Randomized Controlled Trial Comparing Clinical Outcomes and Effects on Intra-Articular Biology for the Treatment of Knee Osteoarthritis. Am. J. Sports Med. 2017, 45, 339–346. [Google Scholar] [CrossRef] [PubMed]
  105. Rayegani, S.M.; Raeissadat, S.A.; Sanei Taheri, M.; Babaee, M.; Bahrami, M.H.; Eliaspour, D.; Ghorbani, E. Does Intra Articular Platelet Rich Plasma Injection Improve Function, Pain and Quality of Life in Patients with Osteoarthritis of the Knee? A Randomized Clinical Trial. Orthop. Rev. 2014, 6, 5405. [Google Scholar] [CrossRef] [Green Version]
  106. Louis, M.L.; Magalon, J.; Jouve, E.; Bornet, C.E.; Mattei, J.C.; Chagnaud, C.; Rochwerger, A.; Veran, J.; Sabatier, F. Growth Factors Levels Determine Efficacy of Platelets Rich Plasma Injection in Knee Osteoarthritis: A Randomized Double Blind Noninferiority Trial Compared With Viscosupplementation. Arthrosc. J. Arthrosc. Relat. Surg. 2018, 34, 1530–1540.e2. [Google Scholar] [CrossRef]
  107. Ahmad, H.S.; Farrag, S.E.; Okasha, A.E.; Kadry, A.O.; Ata, T.B.; Monir, A.A.; Shady, I. Clinical Outcomes Are Associated with Changes in Ultrasonographic Structural Appearance after Platelet-Rich Plasma Treatment for Knee Osteoarthritis. Int. J. Rheum. Dis. 2018, 21, 960–966. [Google Scholar] [CrossRef]
  108. Su, K.; Bai, Y.; Wang, J.; Zhang, H.; Liu, H.; Ma, S. Comparison of Hyaluronic Acid and PRP Intra-Articular Injection with Combined Intra-Articular and Intraosseous PRP Injections to Treat Patients with Knee Osteoarthritis. Clin. Rheumatol. 2018, 37, 1341–1350. [Google Scholar] [CrossRef]
  109. Lisi, C.; Perotti, C.; Scudeller, L.; Sammarchi, L.; Dametti, F.; Musella, V.; Di Natali, G. Treatment of Knee Osteoarthritis: Platelet-Derived Growth Factors vs. Hyaluronic Acid. A Randomized Controlled Trial. Clin. Rehabil. 2018, 32, 330–339. [Google Scholar] [CrossRef]
  110. Buendía-López, D.; Medina-Quirós, M.; Fernández-Villacañas Marín, M.Á. Clinical and Radiographic Comparison of a Single LP-PRP Injection, a Single Hyaluronic Acid Injection and Daily NSAID Administration with a 52-Week Follow-up: A Randomized Controlled Trial. J. Orthop. Traumatol. 2018, 19, 3. [Google Scholar] [CrossRef]
  111. Rahimzadeh, P.; Imani, F.; Faiz, S.H.R.; Entezary, S.R.; Narimani Zamanabadi, M.; Alebouyeh, M.R. The Effects of Injecting Intra-Articular Platelet-Rich Plasma or Prolotherapy on Pain Score and Function in Knee Osteoarthritis. CIA 2018, 13, 73–79. [Google Scholar] [CrossRef] [Green Version]
  112. Montañez-Heredia, E.; Irízar, S.; Huertas, P.; Otero, E.; del Valle, M.; Prat, I.; Díaz-Gallardo, M.; Perán, M.; Marchal, J.; Hernandez-Lamas, M. Intra-Articular Injections of Platelet-Rich Plasma versus Hyaluronic Acid in the Treatment of Osteoarthritic Knee Pain: A Randomized Clinical Trial in the Context of the Spanish National Health Care System. Int. J. Mol. Sci. 2016, 17, 71064. [Google Scholar] [CrossRef] [PubMed]
  113. Huang, G.; Hua, S.; Yang, T.; Ma, J.; Yu, W.; Chen, X. Platelet-rich Plasma Shows Beneficial Effects for Patients with Knee Osteoarthritis by Suppressing Inflammatory Factors. Exp. Ther. Med. 2018, 15, 3096–3102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Filardo, G.; Kon, E.; Di Martino, A.; Di Matteo, B.; Merli, M.L.; Cenacchi, A.; Fornasari, P.M.; Marcacci, M. Platelet-Rich Plasma vs Hyaluronic Acid to Treat Knee Degenerative Pathology: Study Design and Preliminary Results of a Randomized Controlled Trial. BMC Musculoskelet. Disord. 2012, 13, 229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Raeissadat, S.A.; Rayegani, S.M.; Hassanabadi, H.; Fathi, M.; Ghorbani, E.; Babaee, M.; Azma, K. Knee Osteoarthritis Injection Choices: Platelet- Rich Plasma (PRP) versus Hyaluronic Acid (A One-Year Randomized Clinical Trial). Clin. Med. Insights Arthritis Musculoskelet Disord. 2015, 8, CMAMD.S17894. [Google Scholar] [CrossRef] [PubMed]
  116. Spaková, T.; Rosocha, J.; Lacko, M.; Harvanová, D.; Gharaibeh, A. Treatment of Knee Joint Osteoarthritis with Autologous Platelet-Rich Plasma in Comparison with Hyaluronic Acid. Am. J. Phys. Med. Rehabil. 2012, 91, 411–417. [Google Scholar] [CrossRef] [PubMed]
  117. Patel, S.; Dhillon, M.S.; Aggarwal, S.; Marwaha, N.; Jain, A. Treatment With Platelet-Rich Plasma Is More Effective Than Placebo for Knee Osteoarthritis: A Prospective, Double-Blind, Randomized Trial. Am. J. Sports Med. 2013, 41, 356–364. [Google Scholar] [CrossRef] [PubMed]
  118. Tavassoli, M.; Janmohammadi, N.; Hosseini, A.; Khafri, S.; Esmaeilnejad-Ganji, S.M. Single- and Double-Dose of Platelet-Rich Plasma versus Hyaluronic Acid for Treatment of Knee Osteoarthritis: A Randomized Controlled Trial. WJO 2019, 10, 310–326. [Google Scholar] [CrossRef]
  119. Wu, Y.-T.; Hsu, K.-C.; Li, T.-Y.; Chang, C.-K.; Chen, L.-C. Effects of Platelet-Rich Plasma on Pain and Muscle Strength in Patients With Knee Osteoarthritis. Am. J. Phys. Med. Rehabil. 2018, 97, 248–254. [Google Scholar] [CrossRef]
  120. Lin, K.-Y.; Yang, C.-C.; Hsu, C.-J.; Yeh, M.-L.; Renn, J.-H. Intra-Articular Injection of Platelet-Rich Plasma Is Superior to Hyaluronic Acid or Saline Solution in the Treatment of Mild to Moderate Knee Osteoarthritis: A Randomized, Double-Blind, Triple-Parallel, Placebo-Controlled Clinical Trial. Arthrosc. J. Arthrosc. Relat. Surg. 2019, 35, 106–117. [Google Scholar] [CrossRef]
  121. Yu, W.; Xu, P.; Huang, G.; Liu, L. Clinical Therapy of Hyaluronic Acid Combined with Platelet-rich Plasma for the Treatment of Knee Osteoarthritis. Exp. Ther. Med. 2018, 16, 2119–2125. [Google Scholar] [CrossRef] [Green Version]
  122. Xu, Z.; He, Z.; Shu, L.; Li, X.; Ma, M.; Ye, C. Intra-Articular Platelet-Rich Plasma Combined With Hyaluronic Acid Injection for Knee Osteoarthritis Is Superior to Platelet-Rich Plasma or Hyaluronic Acid Alone in Inhibiting Inflammation and Improving Pain and Function. Arthrosc. J. Arthrosc. Relat. Surg. 2021, 37, 903–915. [Google Scholar] [CrossRef] [PubMed]
  123. Tucker, J.D.; Goetz, L.L.; Duncan, M.B.; Gilman, J.B.; Elmore, L.W.; Sell, S.A.; McClure, M.J.; Quagliano, P.V.; Martin, C.C. Randomized, Placebo-Controlled Analysis of the Knee Synovial Environment Following Platelet-Rich Plasma Treatment for Knee Osteoarthritis. PM&R 2021, 13, 707–719. [Google Scholar] [CrossRef]
  124. Raeissadat, S.A.; Gharooee Ahangar, A.; Rayegani, S.M.; Minator Sajjadi, M.; Ebrahimpour, A.; Yavari, P. Platelet-Rich Plasma-Derived Growth Factor vs Hyaluronic Acid Injection in the Individuals with Knee Osteoarthritis: A One Year Randomized Clinical Trial. JPR 2020, 13, 1699–1711. [Google Scholar] [CrossRef] [PubMed]
  125. Park, Y.-B.; Kim, J.-H.; Ha, C.-W.; Lee, D.-H. Clinical Efficacy of Platelet-Rich Plasma Injection and Its Association With Growth Factors in the Treatment of Mild to Moderate Knee Osteoarthritis: A Randomized Double-Blind Controlled Clinical Trial As Compared With Hyaluronic Acid. Am. J. Sports Med. 2021, 49, 487–496. [Google Scholar] [CrossRef]
  126. Dulic, O.; Rasovic, P.; Lalic, I.; Kecojevic, V.; Gavrilovic, G.; Abazovic, D.; Maric, D.; Miskulin, M.; Bumbasirevic, M. Bone Marrow Aspirate Concentrate versus Platelet Rich Plasma or Hyaluronic Acid for the Treatment of Knee Osteoarthritis. Medicina 2021, 57, 1193. [Google Scholar] [CrossRef]
  127. Raeissadat, S.A.; Ghazi Hosseini, P.; Bahrami, M.H.; Salman Roghani, R.; Fathi, M.; Gharooee Ahangar, A.; Darvish, M. The Comparison Effects of Intra-Articular Injection of Platelet Rich Plasma (PRP), Plasma Rich in Growth Factor (PRGF), Hyaluronic Acid (HA), and Ozone in Knee Osteoarthritis; a One Year Randomized Clinical Trial. BMC Musculoskelet. Disord. 2021, 22, 134. [Google Scholar] [CrossRef]
  128. Elksniņš-Finogejevs, A.; Vidal, L.; Peredistijs, A. Intra-Articular Platelet-Rich Plasma vs Corticosteroids in the Treatment of Moderate Knee Osteoarthritis: A Single-Center Prospective Randomized Controlled Study with a 1-Year Follow Up. J. Orthop. Surg. Res. 2020, 15, 257. [Google Scholar] [CrossRef]
  129. Pishgahi, A.; Abolhasan, R.; Shakouri, S.K.; Soltani-Zangbar, M.S.; Dareshiri, S.; Ranjbar Kiyakalayeh, S.; Khoeilar, A.; Zamani, M.; Motavalli Khiavi, F.; Pourabbas Kheiraddin, B.; et al. Effect of Dextrose Prolotherapy, Platelet Rich Plasma and Autologous Conditioned Serum on Knee Osteoarthritis: A Randomized Clinical Trial. IJAAI 2020. [Google Scholar] [CrossRef]
  130. Dório, M.; Pereira, R.M.R.; Luz, A.G.B.; Deveza, L.A.; de Oliveira, R.M.; Fuller, R. Efficacy of Platelet-Rich Plasma and Plasma for Symptomatic Treatment of Knee Osteoarthritis: A Double-Blinded Placebo-Controlled Randomized Clinical Trial. BMC Musculoskelet. Disord. 2021, 22, 822. [Google Scholar] [CrossRef]
  131. Kesiktas, F.N.; Dernek, B.; Sen, E.I.; Albayrak, H.N.; Aydin, T.; Yildiz, M. Comparison of the Short-Term Results of Single-Dose Intra-Articular Peptide with Hyaluronic Acid and Platelet-Rich Plasma Injections in Knee Osteoarthritis: A Randomized Study. Clin. Rheumatol. 2020, 39, 3057–3064. [Google Scholar] [CrossRef]
  132. Lana, J.F.S.D.; Weglein, A.; Sampson, S.E.; Vicente, E.F.; Huber, S.C.; Souza, C.V.; Ambach, M.A.; Vincent, H.; Urban-Paffaro, A.; Onodera, C.M.K.; et al. Randomized Controlled Trial Comparing Hyaluronic Acid, Platelet-Rich Plasma and the Combination of Both in the Treatment of Mild and Moderate Osteoarthritis of the Knee. J. Stem. Cells Regen. Med. 2016, 12, 69–78. [Google Scholar] [PubMed]
  133. Bastos, R.; Mathias, M.; Andrade, R.; Amaral, R.J.F.C.; Schott, V.; Balduino, A.; Bastos, R.; Miguel Oliveira, J.; Reis, R.L.; Rodeo, S.; et al. Intra-Articular Injection of Culture-Expanded Mesenchymal Stem Cells with or without Addition of Platelet-Rich Plasma Is Effective in Decreasing Pain and Symptoms in Knee Osteoarthritis: A Controlled, Double-Blind Clinical Trial. Knee Surg. Sports Traumatol. Arthrosc. 2020, 28, 1989–1999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Bastos, R.; Mathias, M.; Andrade, R.; Bastos, R.; Balduino, A.; Schott, V.; Rodeo, S.; Espregueira-Mendes, J. Intra-Articular Injections of Expanded Mesenchymal Stem Cells with and without Addition of Platelet-Rich Plasma Are Safe and Effective for Knee Osteoarthritis. Knee Surg. Sports Traumatol. Arthrosc. 2018, 26, 3342–3350. [Google Scholar] [CrossRef]
  135. Görmeli, G.; Görmeli, C.A.; Ataoglu, B.; Çolak, C.; Aslantürk, O.; Ertem, K. Multiple PRP Injections Are More Effective than Single Injections and Hyaluronic Acid in Knees with Early Osteoarthritis: A Randomized, Double-Blind, Placebo-Controlled Trial. Knee Surg. Sports Traumatol. Arthrosc. 2017, 25, 958–965. [Google Scholar] [CrossRef] [PubMed]
  136. Kavadar, G.; Demircioglu, D.T.; Celik, M.Y.; Emre, T.Y. Effectiveness of Platelet-Rich Plasma in the Treatment of Moderate Knee Osteoarthritis: A Randomized Prospective Study. J. Phys. Ther. Sci. 2015, 27, 3863–3867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Simental-Mendía, M.; Acosta-Olivo, C.A.; Hernández-Rodríguez, A.N.; Santos-Santos, O.R.; de la Garza-Castro, S.; Peña-Martínez, V.M.; Vilchez-Cavazos, F. Intraarticular Injection of Platelet-Rich Plasma in Knee Osteoarthritis: Single versus Triple Application Approach. Pilot Study. Acta Reum. Port. 2019, 44, 138–144. [Google Scholar]
  138. Raeissadat, S.A.; Rayegani, S.M.; Ahangar, A.G.; Abadi, P.H.; Mojgani, P.; Ahangar, O.G. Efficacy of Intra-Articular Injection of a Newly Developed Plasma Rich in Growth Factor (PRGF) Versus Hyaluronic Acid on Pain and Function of Patients with Knee Osteoarthritis: A Single-Blinded Randomized Clinical Trial. Clin. Med. Insights Arthritis Musculoskelet. Disord. 2017, 10, 117954411773345. [Google Scholar] [CrossRef]
  139. Vaquerizo, V.; Plasencia, M.Á.; Arribas, I.; Seijas, R.; Padilla, S.; Orive, G.; Anitua, E. Comparison of Intra-Articular Injections of Plasma Rich in Growth Factors (PRGF-Endoret) Versus Durolane Hyaluronic Acid in the Treatment of Patients with Symptomatic Osteoarthritis: A Randomized Controlled Trial. Arthrosc. J. Arthrosc. Relat. Surg. 2013, 29, 1635–1643. [Google Scholar] [CrossRef]
  140. Sánchez, M.; Fiz, N.; Azofra, J.; Usabiaga, J.; Aduriz Recalde, E.; Garcia Gutierrez, A.; Albillos, J.; Gárate, R.; Aguirre, J.J.; Padilla, S.; et al. A Randomized Clinical Trial Evaluating Plasma Rich in Growth Factors (PRGF-Endoret) Versus Hyaluronic Acid in the Short-Term Treatment of Symptomatic Knee Osteoarthritis. Arthrosc. J. Arthrosc. Relat. Surg. 2012, 28, 1070–1078. [Google Scholar] [CrossRef]
  141. Smith, P.A. Intra-Articular Autologous Conditioned Plasma Injections Provide Safe and Efficacious Treatment for Knee Osteoarthritis: An FDA-Sanctioned, Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Am. J. Sports Med. 2016, 44, 884–891. [Google Scholar] [CrossRef]
  142. Cerza, F.; Carnì, S.; Carcangiu, A.; Di Vavo, I.; Schiavilla, V.; Pecora, A.; De Biasi, G.; Ciuffreda, M. Comparison Between Hyaluronic Acid and Platelet-Rich Plasma, Intra-Articular Infiltration in the Treatment of Gonarthrosis. Am. J. Sports Med. 2012, 40, 2822–2827. [Google Scholar] [CrossRef] [PubMed]
  143. Di Martino, A.; Di Matteo, B.; Papio, T.; Tentoni, F.; Selleri, F.; Cenacchi, A.; Kon, E.; Filardo, G. Platelet-Rich Plasma Versus Hyaluronic Acid Injections for the Treatment of Knee Osteoarthritis: Results at 5 Years of a Double-Blind, Randomized Controlled Trial. Am. J. Sports Med. 2019, 47, 347–354. [Google Scholar] [CrossRef] [PubMed]
  144. Luo, D.; Wan, X.; Liu, J.; Tong, T. Optimally Estimating the Sample Mean from the Sample Size, Median, Mid-Range, and/or Mid-Quartile Range. Stat. Methods Med. Res. 2018, 27, 1785–1805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Higgins, J.; Thomas, J.; Chandler, J.; Cumpston, M.; Li, T.; Page, M.; Welch, V. Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (Updated February 2021). Available online: www.training.cochrane.org/handbook (accessed on 10 January 2022).
  146. R Core Team A Language and Environment for Statistical Computing; R Foundation for Statistical Computing: Vienna, Austria, 2018; Available online: http://www.R-project.org/ (accessed on 10 January 2022).
Figure 1. Molecular pathways involved in platelet activation during the inflammatory process.
Figure 1. Molecular pathways involved in platelet activation during the inflammatory process.
Cells 11 01080 g001
Figure 2. Flow chart of study inclusion.
Figure 2. Flow chart of study inclusion.
Cells 11 01080 g002
Figure 3. Forest plot for VAS scores comparing PRP versus control (CI, confidence interval; IV, inverse variance; SD, standard deviation; Cells 11 01080 i001, mean with 95% confidence interval for total and subtotals; Cells 11 01080 i002, mean with 95% confidence interval for source data).
Figure 3. Forest plot for VAS scores comparing PRP versus control (CI, confidence interval; IV, inverse variance; SD, standard deviation; Cells 11 01080 i001, mean with 95% confidence interval for total and subtotals; Cells 11 01080 i002, mean with 95% confidence interval for source data).
Cells 11 01080 g003
Figure 4. Forest plot for WOMAC scores (CI, confidence interval; IV, inverse variance; SD, standard deviation; Cells 11 01080 i003, mean with 95% confidence interval for total and subtotals; Cells 11 01080 i004, mean with 95% confidence interval for source data).
Figure 4. Forest plot for WOMAC scores (CI, confidence interval; IV, inverse variance; SD, standard deviation; Cells 11 01080 i003, mean with 95% confidence interval for total and subtotals; Cells 11 01080 i004, mean with 95% confidence interval for source data).
Cells 11 01080 g004
Figure 5. Forest plot for IKDC scores (CI, confidence interval; IV, inverse variance; SD, standard deviation; Cells 11 01080 i005, mean with 95% confidence interval for total and subtotals; Cells 11 01080 i006, mean with 95% confidence interval for source data).
Figure 5. Forest plot for IKDC scores (CI, confidence interval; IV, inverse variance; SD, standard deviation; Cells 11 01080 i005, mean with 95% confidence interval for total and subtotals; Cells 11 01080 i006, mean with 95% confidence interval for source data).
Cells 11 01080 g005
Figure 6. Forest plot for KOOS sub-scores (ADL, activities of daily living; CI, confidence interval; IV, inverse variance; QoL, quality of life; SD, standard deviation; Cells 11 01080 i007, mean with 95% confidence interval for total and subtotals; Cells 11 01080 i008, mean with 95% confidence interval for source data).
Figure 6. Forest plot for KOOS sub-scores (ADL, activities of daily living; CI, confidence interval; IV, inverse variance; QoL, quality of life; SD, standard deviation; Cells 11 01080 i007, mean with 95% confidence interval for total and subtotals; Cells 11 01080 i008, mean with 95% confidence interval for source data).
Cells 11 01080 g006aCells 11 01080 g006bCells 11 01080 g006c
Figure 7. Forest plot for adverse events (CI, confidence interval; M-H, Mantel-Heanszel; Cells 11 01080 i009, odds ratio with 95% confidence interval for total and subtotals; Cells 11 01080 i010, odds ratio with 95% confidence interval for source data).
Figure 7. Forest plot for adverse events (CI, confidence interval; M-H, Mantel-Heanszel; Cells 11 01080 i009, odds ratio with 95% confidence interval for total and subtotals; Cells 11 01080 i010, odds ratio with 95% confidence interval for source data).
Cells 11 01080 g007
Figure 8. Risk of bias analysis for PRP application in osteoarthritis ( Cells 11 01080 i011, low risk of bias; Cells 11 01080 i012, unclear risk of bias; Cells 11 01080 i013, high risk of bias).
Figure 8. Risk of bias analysis for PRP application in osteoarthritis ( Cells 11 01080 i011, low risk of bias; Cells 11 01080 i012, unclear risk of bias; Cells 11 01080 i013, high risk of bias).
Cells 11 01080 g008
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tramś, E.; Malesa, K.; Pomianowski, S.; Kamiński, R. Role of Platelets in Osteoarthritis—Updated Systematic Review and Meta-Analysis on the Role of Platelet-Rich Plasma in Osteoarthritis. Cells 2022, 11, 1080. https://doi.org/10.3390/cells11071080

AMA Style

Tramś E, Malesa K, Pomianowski S, Kamiński R. Role of Platelets in Osteoarthritis—Updated Systematic Review and Meta-Analysis on the Role of Platelet-Rich Plasma in Osteoarthritis. Cells. 2022; 11(7):1080. https://doi.org/10.3390/cells11071080

Chicago/Turabian Style

Tramś, Ewa, Kamila Malesa, Stanisław Pomianowski, and Rafał Kamiński. 2022. "Role of Platelets in Osteoarthritis—Updated Systematic Review and Meta-Analysis on the Role of Platelet-Rich Plasma in Osteoarthritis" Cells 11, no. 7: 1080. https://doi.org/10.3390/cells11071080

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop