Next Article in Journal
Advancements in Glioma Care: Focus on Emerging Neurosurgical Techniques
Next Article in Special Issue
Can Combining Hyaluronic Acid and Physiotherapy in Knee Osteoarthritis Improve the Physicochemical Properties of Synovial Fluid?
Previous Article in Journal
Platelet-Rich Plasma (PRP) in Dermatology: Cellular and Molecular Mechanisms of Action
Previous Article in Special Issue
The Classification of Suspected Predominant Nociplastic Pain in People with Moderate and Severe Haemophilia: A Secondary Exploratory Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathogenic Mechanisms and Therapeutic Approaches in Obesity-Related Knee Osteoarthritis

1
Department of Rheumatology, Medical University of Sofia, 1431 Sofia, Bulgaria
2
Clinic of Rheumatology, University Hospital ‘St. Ivan Rilski’, 1612 Sofia, Bulgaria
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(1), 9; https://doi.org/10.3390/biomedicines12010009
Submission received: 23 November 2023 / Revised: 10 December 2023 / Accepted: 14 December 2023 / Published: 20 December 2023

Abstract

:
The knee is the joint most frequently involved in osteoarthritis, a common joint disorder in the adult population that is associated with significant chronic joint pain, reduced mobility and quality of life. Recent studies have established an association between obesity and the development of knee osteoarthritis that goes beyond the increased mechanical load on the knees as weight-bearing joints. This link is based on the maintenance of a chronic low-grade inflammation, altered secretion of adipokines by the adipose tissue and development of sarcopenia. Major adipokines involved in the pathogenesis of obesity-related knee osteoarthritis include adiponectin, which appears to have a protective effect, as well as leptin, resistin and visfatin, which are associated with higher pain scores and more severe structural damage. Joint pain in knee osteoarthritis may be both nociceptive and neuropathic and is the result of complex mechanisms driven by nerve growth factor, calcitonin gene-related peptide and pro-inflammatory cytokines. The role of endogenous cannabinoids and gut microbiota in common mechanisms between obesity and knee pain has recently been studied. The aim of the present review is to highlight major pathogenic mechanisms in obesity-related knee osteoarthritis with special attention on pain and to comment on possible therapeutic approaches.

1. Introduction

Osteoarthritis (OA) is a common joint disorder in the adult population and is widely associated with chronic joint pain, reduced mobility and impaired productivity [1,2,3]. The knee is the most commonly affected joint, followed by the interphalangeal joints of the hand and the hip [4]. The development of OA is related to a plethora of factors such as age, sex, genetics, trauma and increased mechanical load, as well as various concomitant diseases. In particular, obesity-related OA, also referred to as metabolic syndrome-associated OA, has recently been recognized as a separate entity [2,3,5]. It has been shown that obesity is related to knee OA not simply through an increased mechanical load on weight-bearing joints but through the maintenance of chronic inflammation, altered secretion of adipokines and development of sarcopenia [2,3]. In addition, a higher degree of obesity has been associated with a higher pain score [3]. Recent recommendations on the therapeutic management of knee OA, however, do not include specific guidelines in cases of obesity-related knee OA [6]. Therefore, the aim of this review was to outline the key pathogenic aspects in obesity-related knee OA and to present current understanding of pain mechanisms and therapeutic approaches while commenting, where possible, on those that may be beneficial in knee OA associated with metabolic syndrome.

2. Joint Alterations in Knee OA

Knee OA is a chronic degenerative joint disease that not only involves the joint cartilage and the underlying subchondral bone, but also the synovial membrane, adjacent muscles and ligaments, as well as Hoffa’s fat pad in a pattern that has been termed ‘whole joint disease’ [7,8].
Articular cartilage is a connective tissue structure built of specialized cells called chondrocytes and an extracellular matrix (ECM) that is normally rich in water (more than 70%) and also contains various organic components. These include collagen type II, glycosamino-glycans (chondroitin sulfate and keratan sulfate) and protein molecules such as decorin, aggrecan and fibromodulin. These ECM components are produced by chondrocytes and their synthesis is the subject of finely tuned regulation that involves various proteolytic enzymes [8,9,10]. The organization of the ECM is quite complex. A network of collagen type II fibrils is intertwined with proteoglycan aggregates made up of a ‘core protein’ (aggrecan) that is bound to glycosamino-glycans and further connected to a ‘backbone’ of hyaluronic acid [8,9]. The disruption of this complex organization marks the onset of OA. Two groups of proteolytic enzymes have been implicated in the degradation of the cartilage ECM. A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS) is a family of enzymes responsible for the cleavage of aggrecan from the hyaluronic acid (in particular ADAMTS-4 and 5), while matrix metalloproteinases (MMPs), especially MMP-13, break down the collagen network [11,12]. These ECM alterations then lead to proliferation and hypertrophy of the chondrocytes whose ‘pro-synthetic’ phenotype is changed into a ‘pro-inflammatory’ one, leading to the production of potent mediators of inflammation such as interleukin 6 (IL-6), tumor necrosis factor-alpha (TNFα), interleukin 1β (IL-1β), which further stimulate the breakdown of the ECM, contribute to the occurrence of pain and activate the synovial membrane [8,13]. These pro-inflammatory mediators upregulate the production of ADAMTS, MMPs, cathepsins, cyclooxygenase-2 (COX-2) as well as reactive oxygen species (ROS) which can directly cleave collagen and aggrecan, thereby intensifying cartilage degradation [14]. The significance of proteolytic enzymes has also been demonstrated in animal studies, where knockout mice lacking ADAMTS-5 or MMP-13 did not develop experimental OA [15,16]. In addition to ECM degradation by ADAMTS and MMPs, the de novo synthesis of collagen type II and aggrecan is suppressed in chondrocytes of joints with OA [17].
A major molecular pathway implicated in the development of OA is the nuclear factor kappa-B (NF-kB) signaling pathway [18]. The nuclear translocation of NF-kB and its binding to regulatory segments in DNA initiates the cascade of cartilage degradation, as it leads to increased production of MMP-1, MMP-9 and MMP-13, ADAMTS-4 and 5 and secretion of the pro-inflammatory cytokines, which is then further stimulated through a positive feedback loop [19,20]. The mitogen-activated protein kinase (MAPK) pathway has been shown to upregulate the release of MMP-1 and MMP-13, increase collagen degradation and yield higher levels of TNFα and IL-1β through activation of regulatory gene p38 [21]. In addition, Wnt/β-catenin is another molecular pathway with a key role in OA. Upon binding to its receptor, Wnt leads to the activation of proteins which inhibit the cleavage of β-catenin and allow it to translocate into the nucleus. This is associated with an increase in MMP-13, ADAMTS-4 and 5 and reduced cartilage thickness [22]. Furthermore, Wnt/β-catenin inhibition has been shown to reduce cartilage degradation and ameliorate OA severity in a mouse model [23].
Articular cartilage degradation, however, is not the only hallmark of OA. Subchondral bone changes in the early stages of OA include remodeling of the subchondral plate, increased porosity, decreased thickness of the trabeculae and bone mineral density. Late-stage OA, on the other hand, is characterized by subchondral bone sclerosis with increased trabecular thickness [24,25]. In addition, subchondral bone cysts in weight-bearing segments of the joint have been described as containing a high number of osteoblasts, osteoclasts and osteoprogenitor cells, which indicates significant bone turnover [26]. These osteoblasts show stronger alkaline phosphatase activity and release higher levels of transforming growth factor β1 (TGFβ1), insulin-like growth factor 1 (IGF1), vascular endothelial growth factor (VEGF) and receptor activator of nuclear factor kappa beta-ligand (RANKL) compared to normal osteoblasts [25]. Synovial inflammation has been associated with pain, increased cartilage degradation and radiographic progression with narrowing of the joint space [8,27]. The synovium of OA joints is rich in macrophages, mast cells, T- and B-cells and the synovial fluid contains a variety of pro-inflammatory mediators, including TNFα, IL-6, IL-1β, TGFβ1, VEGF, leukotrienes and prostaglandins, which can stimulate the release of proteolytic enzymes by chondrocytes [27,28,29,30]. The role of synovial macrophages in particular has been widely investigated in OA. Their ability to recognize danger-associated molecular patterns (DAMPs) such as cartilage fragments and intracellular proteins from damaged cells leads to an increased secretion of pro-inflammatory cytokines [31]. Classically, two distinct phenotypes of synovial macrophages have been described. M1, or pro-inflammatory macrophages, have been associated with progression of OA through amplification of MMP-1 and 3 and ADAMTS-4 and 5 and downregulation of collagen and aggrecan synthesis. M2, or anti-inflammatory macrophages promote a ‘protective environment’ in the OA joint through secretion of IL-4 and 10 [31]. The M1/M2 ratio has been found to be markedly elevated in OA knees and correlates with radiographic progression [32]. Finally, pathological manifestations in Hoffa’s fat pad include an increase in vascularization and fibrosis, thickening of the interlobular septa, infiltration of inflammatory cells and higher levels of VEGF and IL-6 [33].

3. The Link between Obesity and Knee OA

While aging remains the primary cause for development of OA, overweight (defined as a body mass index (BMI) > 25 kg/m2) and obesity (defined as a BMI > 30 kg/m2) represent major risk factors associated with knee OA [2,3,34]. In fact, obesity in younger age groups (20–29 and 30–39) has been associated with a higher incidence of knee but not of hip OA, suggesting that the impact of obesity predates that of physiological aging [35]. In addition, higher pain scores, impaired mobility and lower physical activity have all been associated with increased BMI levels [3]. The higher mechanical load exerted on the knees as weight-bearing joints leads to impaired cartilage homeostasis [2]. Animal studies found this to be associated with reduced cartilage thickness, rapid degeneration, increased subchondral bone thickness and occurrence of bone marrow lesions [36,37]. Weight overloading of joints has been shown to upregulate the secretion of TNFα and IL-1β thus mediating the degradation of cartilage ECM [38]. Furthermore, the higher thigh girth seen in overweight patients changes the alignment of lower leg joints—greater abduction of the hip and varus deformity of the knee lead to predominant load transfer along the medial portion of the knee where articular cartilage suffers earlier damage [2]. While increased body weight leading to altered mechanical load on the knees can certainly account for some of the risk of OA development, obesity has also been implicated in OA of non-weight-bearing joints which hints at the role of its systemic effects [3]. Different measurements of increased body weight such as BMI, fat percentage, abdominal obesity and amount of visceral abdominal tissue have all been associated with development of hand OA, while higher BMI correlates with pain intensity in subjects with hand OA [38]. In fact, it has been proposed that obesity’s role in the development and progression of OA combines the impact of increased body weight and the establishment of a chronic ‘micro-inflammatory state’ [39]. Therefore, obesity-related OA can be categorized as a separate type of secondary OA, different from aging-associated primary OA and other cases of OA secondary to trauma or inflammatory arthritis.
Recent studies have elaborated on the role of adipose tissue as a potential source of cytokines, chemokines and other mediators, collectively known as adipokines [40]. These mediators promote either cartilage degradation or the preservation of its integrity through complex interactions (Table 1) [38,40]. In addition, obesity leads to a change in the cellular profile of resident macrophages, from the anti-inflammatory M2 phenotype to the pro-inflammatory M1 phenotype, which stimulates the release of pro-inflammatory cytokines such as TNFα, IL-6 and IL-1β [41].
One major adipokine implicated in the development of knee OA with an apparently contrasting impact is adiponectin. Higher serum levels of adiponectin have been associated with the formation of osteophytes, narrowing of the joint space and higher radiographic score [42]. On the other hand, adiponectin appears to have anti-inflammatory properties by altering the phenotype of resident adipose tissue macrophages from M1 to M2 and by decreasing the levels of TNFα [43]. Moreover, adiponectin has been shown to lower serum levels of triglycerides and free fatty acids, thereby reducing the level of oxidative stress [44]. At the joint level, adiponectin upregulates the expression of the tissue inhibitor of MMP-2 (TIMP-2), which also counters the action of ADAMTS and inhibits the IL-1β-stimulated expression of MMP-13 [44,45].
The serum levels of another major adipokine, leptin, are also increased in overweight patients [46]. They correlate with a higher BMI, a higher Western Ontario and McMaster Universities Arthritis Index (WOMAC) pain score and structural damage [47]. On the other hand, the decrease of serum leptin seen in weight loss has been associated with the improvement of patients’ symptoms [46]. Chondrocytes from OA joints have also demonstrated higher leptin levels and this local increase in the concentration of this particular adipokine upregulates a plethora of pro-inflammatory cytokines such as IL-6, IL-1β, vascular cell adhesion molecule-1 (VCAM-1) and prostaglandin E2, as well as multiple MMP subtypes (namely MMP-1, 3, 9 and 13) by chondrocytes, which promote cartilage degradation [48,49]. In addition, leptin stimulates synovial fibroblasts to release IL-6 and IL-8 and mediates subchondral bone remodeling by increasing the alkaline phosphatase activity of osteoblasts, where its expression is higher than that of normal osteoblasts [50].
Resistin, an adipokine associated with the progression and severity of knee OA, drives the production of MMPs and pro-inflammatory cytokines by chondrocytes and promotes structural damage [51,52,53]. Its serum levels are higher in OA patients compared to healthy controls and correlate with the development of cartilage defects and bone marrow lesions, which are associated with higher pain score and more pronounced structural alterations [53]. Visfatin is a pro-inflammatory adipokine implicated in the pathogenesis of OA through its upregulation of TNFα, IL-6, IL-1β, MMP-3 and 13 and ADAMTS-4 and 5. It is among the major promoters of cartilage degradation by inhibiting the synthesis of collagen type II and proteoglycans with high molecular weight. Serum and synovial fluid levels of visfatin show a positive correlation with structural damage, markers indicating degradation of collagen type II and aggrecan, CRP levels and OA symptoms [54].
Dyslipidemia, another major aspect of obesity, as well as the associated lipotoxicity have recently been implicated in OA [55,56]. Altered lipid metabolism and an upregulation of two key enzymes—25-hydroxycholesterol 7α-hydroxylase and cholesterol 25-hydroxylase—have been reported in OA and likely play a key role in structural damage through the promotion of MMP and ADAMTS. This, in turn, has been associated with structural changes such as synovitis, formation of osteophytes and subchondral bone sclerosis [57]. Cartilage degradation is also worsened through the action of reactive oxygen species (ROS) and free fatty acids which induce mitochondrial dysfunction in chondrocytes and stimulate the release of pro-inflammatory cytokines [58,59,60]. A recent cross-sectional study found higher levels of total cholesterol, triglycerides and low-density lipoprotein (LDL) in patients with symptomatic knee OA [61]. These data confirmed earlier observations that hypertriglyceridemia and decreased high-density lipoprotein (HDL) are associated with more severe knee pain [62]. In addition, higher levels of LDL provoke synovial inflammation and ectopic bone formation by stimulating synovial cells and are associated with higher knee pain [55].
Age-related muscle tissue loss and dysfunction also play a key role in the development of knee OA. However, it is a special type of body composition entity, known as sarcopenic obesity, which has recently been studied in the context of knee OA and appears to be more tightly associated with it than non-sarcopenic obesity [63]. It refers to cases in which the increase in body weight caused by obesity is compensated for by a loss of muscle mass [63]. Sarcopenic obesity has a complex pathogenesis which is influenced by age-related changes in physical activity, nutrition and hormonal profile (such as altered secretion of insulin, parathyroid hormone, sex hormones and vitamin D) [63]. Adipokines, as mediators of systemic chronic inflammation, are another factor behind progressive muscle loss. The serum levels of leptin are negatively correlated with skeletal muscle mass and are increased in subjects with sarcopenic obesity [64]. This is thought to be a result of decreased number of leptin receptors and development of leptin resistance, which in turn causes elevated systemic levels of TNFα, IL-6 and promotes insulin resistance [64]. Conversely, high adiponectin levels have been reported in the serum of sarcopenic subjects as a compensatory mechanism aimed at countering the breakdown of their muscle proteins [65]. Furthermore, adiponectin levels are low in obese patients and subjects with insulin resistance [64]. This suggests that obese patients who subsequently develop sarcopenia would have low serum adiponectin levels and thus be unable to compensate for muscle loss. Sarcopenia has recently been found in as many as 45.2% of cases of knee OA, which is twice as high as in healthy controls [66]. A possible pathogenic mechanism linking the two entities could be an impaired afferent input from the OA knee leading to an altered efferent motor neuron impact on the quadriceps [66]. The latter causes quadricep inhibition and weakness and is a separate risk factor for knee OA progression and OA-related pain.

4. Pain in Knee OA

While the hallmark of OA is the degradation of articular cartilage, blood vessels and nerve endings are not present inside the cartilage and so it cannot generate pain sensation directly [67,68]. However, the synovium, joint capsule, adjacent ligaments, subchondral bone and Hoffa’s fat pad are all richly innervated by sensory and sympathetic nerve fibers and can produce nociceptive pain. Nociceptor nerve terminals register stimuli of mechanic, chemical or thermal origin and release an array of neuropeptides, including substance P (SP) and calcitonin gene-related peptide (CGRP) [69]. The pain signal is then transmitted along the dorsal root ganglia to the posterior horn of the spinal cord and the bodies of the sensory neurons, leading to an upregulation of the NOD-like receptor (NLR) family pyrin domain—containing protein 3 (NLRP3) inflammasome, C-C motif chemokine ligand 2 (CCL2) and its receptor (CCR2) and the Wnt/β-catenin pathway [68]. Additional local release of SP and CGRP recruits second-order neurons through which the pain stimulus is ultimately conveyed via ascending pathways to the central nervous system (CNS) where the conscious awareness of pain takes place [70,71,72].
Apart from nociceptive pain arising from damaged tissue, knee pain in OA may also be driven by neuropathic pathways [68]. Cytokines such as TNFα, IL-6, IL-1β and nerve growth factor (NGF) and chemokines such as CCL2 are implicated not only in the occurrence of nociceptive pain but also in the peripheral sensitization of sensory nerve fibers [73]. This persistence of the pain stimulus ultimately causes central sensitization leading to hyperalgesia and tenderness around the joint, which is typical of neuropathic pain [70,71]. These findings are supported by the fact that patients with less severe joint alterations may experience debilitating persistent joint pain [74]. In addition, pain intensity and central sensitization in OA patients were positively correlated with NGF levels in the synovial fluid [75]. While OA pain is usually located at the site of the affected joint and elicited during weight bearing, exercise, walking and climbing or descending stairs and is relieved at rest, increased central sensitization leading to neuropathic pain may be associated with pain at rest, poorly located pain around the joint structures and postoperative pain persistence following total joint arthroplasty [68,75,76]. Multiple pain mediators including growth factors, cytokines and chemokines have been studied in the context of both nociceptive and neuropathic pathways in OA and are reviewed below.
NGF is a primary cytokine regulating pain stimulus transduction which is upregulated in OA joints through increased mechanical stress and the action of IL-1β TNFα and visfatin, which promote its synthesis by synoviocytes, chondrocytes, macrophages, mast cells and neutrophils [77,78]. NGF acts on tropomyosin receptor kinase A (TrkA) which leads to the upregulation of an array of inflammatory cytokines, as well as neurotransmitters such as SP and CGRP, which are involved both in peripheral and central pain mechanisms. Knee OA models based on destabilization of the medial meniscus have been associated with higher expression of NGF and pain-related behavioral changes in mice [79]. A recent study on a model of knee OA induced by medial meniscectomy found that NGF expression in the synovium increased in early OA and subsequently decreased in advanced stages but continued to increase in osteochondral channels and bone marrow [80]. These changes were mirrored by similar time- and site-specific increases in CGRP-mediated sensory innervation [80]. Consequently, a number of clinical trials attempted to use anti-NGF antibodies for treatment of OA but results of rapidly progressive cartilage degradation halted these investigations. These therapeutic interventions will be discussed further below.
The generation and maintenance of pain patterns in OA including mechanical allodynia and movement-evoked pain are associated with molecular changes in sensory neurons of the dorsal root ganglia, including chemokines and their receptors [68]. CCL2 and its interaction with receptor CCR2 has recently been implicated in the development of knee OA pain through local accumulation of macrophages and monocytes [81]. On the other hand, blockade of CCL2/CCR2 signaling led to a decrease in macrophage recruitment and ameliorated the severity of knee synovitis and cartilage lesions in a mouse model of OA [82]. The intra-articular injection of recombinant CCL2 caused knee hyperalgesia by directly stimulating sensory neurons rich in CCR2. Conversely, the administration of a receptor antagonist resolved the established hyperalgesia [83]. These data suggest that CCL2 acts both through local recruitment of inflammatory cells and subsequent action of mediators on nociceptors, as well as through direct activation of sensory neurons and upregulated afferent pain transduction.
CGRP is a neuropeptide largely found in nociceptive neurons in the dorsal root ganglia and in nerve fibers reaching the posterior horn of the spinal cord [68]. In addition, CGRP acts upon synoviocytes, endothelial and inflammatory cells, thus influencing local angiogenesis and inflammation. Elevated levels of CGRP have been found in the serum, synovial fluid and synovial tissue of OA patients and have been correlated with pain intensity [84]. Moreover, CGRP-containing nerve fibers can directly regulate bone remodeling through promotion of osteogenesis and inhibition of osteoclastogenesis [84]. These findings are supported by the established association between serum and synovial fluid levels of CGRP and osteophyte formation in the knee as a whole and in the medial compartment alone [85]. In addition, as OA progresses, sensory nerve terminals rich in CGRP grow along newly formed blood vessels into channels that penetrate articular cartilage [86]. Therefore, joint structures that are normally not innervated might subsequently become sources of pain [87]. Nevertheless, the precise role of CGRP in OA pain remains to be fully elucidated.
As outlined above, pro-inflammatory cytokines such as TNF-α, IL-1β and IL-6 have been well studied in the pathogenesis of OA, however their role in pain mechanisms remain controversial. Notably, clinical trials of anti-cytokine medication in OA patients did not lead to satisfactory pain relief [68]. An earlier study found no correlation between IL-6 levels and WOMAC pain score [88]. This was later challenged by other authors who established a statistically significant positive correlation between IL-6 levels in the synovial fluid and WOMAC pain score [89]. One study found that while synovial levels of TNFα correlated with total WOMAC score, pain during movement and while at rest, synovial levels of IL-1β were inversely associated with knee pain [90]. Injection of TNF induced persistent sensitization of peripheral sensory neurons in the knee, assessed through development of mechanical allodynia. The concomitant administration of TNFα-inhibitor etanercept or a COX-inhibitor reversed that development [91]. It was later reported that IL-1β and IL-6 were negatively correlated with knee pain score and Kellgren-Lawrence score and hypothesized that pain in the early stage of knee OA may be of inflammatory origin and driven by the above cytokines. Conversely, in late-stage OA other sources of pain rather than inflammation need to be investigated [92]. These observations are further supported by an earlier report which found that levels of TNFα and IL-6 were significantly higher in early as opposed to late-stage knee OA [93]. IL-1β has been shown to increase the production of proteolytic enzymes by chondrocytes and to promote osteoclast differentiation [94]. In addition, it is directly involved in pain genesis by upregulating the expression of NGF [95]. A recent paper on an experimental model of knee OA found that a deficiency in type 1 interleukin-1 receptor (IL-1R1) did not alter the course of cartilage degradation but prevented the occurrence of pain [96]. The NLRP3 inflammasome is part of the innate immune system which regulates active IL-1β [68]. As previously demonstrated, the activation of the inflammasome leads to an upregulation of IL-1β, followed by the release of other pro-inflammatory cytokines and the synthesis of proteolytic enzymes such as MMP-13 and ADAMTS5, which directly cause synovial inflammation and cartilage degradation [97]. On the other hand, OA-related risk factors such as oxidized LDL, elevated cholesterol, hydroxyapatite crystals and others are recognized as DAMPs which trigger the NLRP3 pathway and lead to a specialized type of programmed cell death of the chondrocytes, known as pyroptosis [98]. These pyroptotic chondrocytes release IL-1β and TNFα, which in turn promote the release of MMPs and ADAMTS causing further cartilage damage in a vicious cycle. In addition, the crosstalk between neutrophils and macrophages may also be implicated. Neutrophil extracellular traps (NETs) may prime macrophages and promote the activation of the NLRP3 inflammasome, which upregulates IL-1β and IL-18 that further stimulate neutrophils to form NETs—A cascade which has been recently studied in inflammatory conditions such as atherosclerosis and rheumatoid arthritis [99,100]. Since inflammasomes are involved in the low-grade inflammation seen in obesity and metabolic syndrome, they are likely key structures in the development of obesity-related OA. The inhibition of the NLRP3 inflammasome in a rat model of knee OA via an intra-articular injection of the anti-inflammatory molecule dexmedetomidine improved pain and cartilage tissue damage, which was associated with lower levels of TNFα, IL-1β, IL-6, MMP-13 and an upregulation of collagen type II [101].
These data underscore the complexity of pain generation and maintenance in OA and provide a platform for further studies into pain mechanisms in obesity-related knee OA. As outlined above, obesity-related knee OA is associated with higher levels of the pro-inflammatory cytokines (and, therefore, with increased joint pain and structural damage) and an altered adipokine profile. To the best of our knowledge, the link between adipokines and pain at the molecular level has only been studied in a 2014 paper, where the authors found that visfatin increases the synthesis of NGF and can modulate pain occurrence [77]. Clinical outcome-based studies identified that a higher degree of obesity is associated with a higher pain and disability score [3]. An imaging-based study found that the occurrence of bone marrow lesions on magnetic resonance imaging (MRI) was associated with various components of dyslipidemia, including hypercholesterolemia, hypertriglyceridemia and low levels of high-density lipoproteins (HDL) [102]. A recent paper by Valdes attempted to ‘connect the dots’ between obesity and OA pain on the molecular level [103]. The transient receptor potential cation channel vanilloid subfamily member 1 (TRPV1) is involved in the modulation of nociceptive pain afferents to the spinal cord and has been associated with OA pain. TRPV1 knockout mice did not develop hypertension and impaired glucose tolerance and did not have an increased nociception when placed on a high-fat diet compared to controlled wild-type mice on the same diet, thus suggesting a common molecular pathway between OA and metabolic syndrome [104]. The action of some peripheral endogenous cannabinoids appears to provide a second link between OA and obesity. These mediators control appetite and energy metabolism and can reduce pain by acting on TRPV1 [105]. Cannabinoid receptor antagonists with peripheral action reduce body weight and improve dyslipidemia and insulin resistance and may prove beneficial as analgesics in OA [106]. The gut microbiome and its function provide another interesting association between metabolic syndrome and pain in OA. It has recently been demonstrated that gut microbiome dysbiosis is associated with OA pain and structural damage [107]. On the other hand, modulation of the gut microbiome via a higher dietary fiber intake is associated with lower body weight, total cholesterol and systolic blood pressure and protects against metabolic syndrome, coronary artery disease and diabetes [103]. In addition, an experimental study in mice revealed that obesity can itself modulate the gut microbiome by increasing the number of key pro-inflammatory species, which then drive the onset of systemic inflammation, accompanied by macrophage recruitment to the synovium and progression of OA damage [108]. These common molecular mechanisms, however, merit further investigation in future studies.

5. Therapeutic Approaches in Knee OA with Possible Benefit in Obesity-Related Knee OA

While recent years have seen an abundant amount of research into the pathogenesis of obesity-related OA, no specific therapeutic guidelines have been published [6]. The American College of Rheumatology (ACR) and the Osteoarthritis Research Society International (OARSI) have regularly updated their guidelines for the management of knee OA in general and they should be applied to obesity-related OA as well (Table 2) [109,110]. Nevertheless, before deciding on a particular therapeutic approach one should consider any possible adverse outcomes stemming from other possible obesity-associated conditions such as hypertension, insulin resistance or diabetes and dyslipidemia, to name a few.
Both ACR and OARSI guidelines strongly recommend exercise and weight loss which are of particular importance in subjects with obesity-related knee OA. Weight loss of ≥5% has been associated with better functional improvement and less pain intensity [109]. Another study found that losing between 10 and 20% of baseline body weight had a better impact on clinical and mechanical outcome as opposed to weight loss of <10%, and the results were statistically significant [111]. A randomized control trial of 89 patients with obesity and knee OA found that subjects who had adhered to a low-calorie diet and were frequently followed by a dietician achieved a mean weight loss of 10.9 kg and a statistically significant decrease in the WOMAC pain score [112]. Furthermore, weight loss decreases the impact of mechanical load on the knee and strategies to achieve it are a major element of efforts to slow down knee OA progression [2]. Literature data suggest that weight loss is associated with structural benefits in the knee such as increased proteoglycan content in the ECM and reduction in cartilage thickness loss, as well as increased serum levels of cartilage synthesis markers and decreased serum levels of cartilage degradation markers [113,114]. In one randomized control trial, 289 obese subjects were instructed to follow an exercise program aimed at strengthening the quadriceps over a period of two years, which was associated with a statistically significant decrease in knee pain [115]. Resistance training is also important in promoting muscle strength which improves joint mobility and reduces pain in 50–70% of patients with obesity-related knee OA [2]. Finally, exercise and weight loss are pivotal in the management of traditional cardiovascular risk factors associated with obesity [6].
Oral non-steroidal anti-inflammatory drugs (NSAIDs) and intra-articular injections of corticosteroids (CSs) are strongly recommended in knee OA in the general population. However, such drugs should be used with caution in patients with obesity-related knee OA who have a greater cardiovascular risk due to comorbidities associated with metabolic syndrome [6]. It is therefore advisable to prefer topical over oral NSAIDs in these patients, considering their equivalent therapeutic effect but more favorable safety profile due to a much slower absorption [116]. Intra-articular injections of either hyaluronic acid or CSs have been universally used with a favourable therapeutic outcome and few adverse effects. ACR’s guidelines, however, while strongly recommending the use of intra-articular CSs, do not include a statement on the use of hyaluronic acid [109]. OARSI’s guidelines, on the other hand, suggest the use of intra-articular injections of CSs as a means to provide short-term pain relief, whereas injections of hyaluronic acid are recommended for pain relief over a longer period (12 weeks and beyond) and are considered a safer option compared to repeated CS injections [110]. However, one must consider that the effect of intra-articular injections is lower in patients with higher BMI [6]. The use of anti-obesity drugs has been suggested for patients with BMI > 30 but has not yet been fully studied in associated knee OA [117].
An interesting therapeutic approach has recently been suggested based on the molecular subtype of knee OA and identified pathogenic mechanisms [118]. The authors distinguished between four separate subtypes of progressive knee OA, each represented by the presence of distinct molecular profiles, summarized in Table 3. In addition, they suggested pathogenesis-based preferential treatment options, which are indicated in Table 4.
Considering that obesity-related knee OA shares features of all the above molecular subtypes, it could be difficult to point to a single therapeutic approach. A comprehensive literature review on the repeated intra-articular administration of hyaluronic acid at 6-month-long intervals found pain reduction from baseline and sustained or further reduced pain over the course of repeated injections, with the longest follow up period being 25 months [119]. A post-hoc analysis of the prospective, double-blind, randomized, multicenter and parallel-group ‘HAV-2012 trial’ compared pain and function scores before and six months after intra-articular viscosupplementation, and juxtaposed the results with weight status (obese vs. non-obese) and structural changes (assessed through mild, moderate or severe radiological severity based on OARSI’s 1, 2 or 3 grade) [120]. The study found that WOMAC pain score decreased significantly in all patient subgroups—obese, OARSI grade 3, obese and OARSI grade 3, neither obese nor OARSI grade 3. However, the WOMAC pain score was significantly lower in non-obese versus obese patients and in patients with knee OA OARSI grade 1 or 2 versus grade 3. Still, in patients who responded to treatment, the pain score at baseline and its subsequent decease were not related to weight status and radiological severity. Therefore, the authors concluded that while it was less likely for obese patients to respond to treatment, for those reporting benefit from the therapy, pain relief would be similar to that in non-obese patients [120]. Based on these findings, we would suggest that the intra-articular administration of hyaluronic acid could be recommended in obese patients. However, if no clinical benefit (i.e., pain relief, improved mobility) is reported by the patient at six months from baseline, then repeated injections would not be advisable. A recent meta-analysis on a total of 3793 patients, 1067 of whom received a combination of glucosamine and chondroitin as treatment for knee OA, found a statistically significant improvement in the total WOMAC score and the joint space narrowing in the combination group, but no difference in the pain score assessed on a visual analog scale (VAS) [121]. Considering the need to avoid NSAIDs and CSs in patients with obesity-related knee OA, glucosamine, chondroitin and some natural antioxidants such as curcumin, oleuropein and ginger extracts, may be preferred owing to their favorable benefit/risk ratio and potential positive impact on both metabolic syndrome and OA [6]. Experimental animal studies with curcumin showed a marked chondroprotective role via suppression of aggrecan degradation, inhibition of MMP-3, 8 and 13 and ADAMTS-5 and upregulation of the synthesis of collagen type II [122,123,124]. Oleuropein, a potent antioxidant extracted from olive tree leaves and found in olive oil, has anti-obesity effects and lowers systemic inflammation, blood pressure and cholesterol levels [125]. In a study on human chondrocytes, oleuropein downregulated the IL-1β-induced activation of the NFκB pathway and reduced the levels of MMP-1, MMP-13, and ADAMTS5, thus countering cartilage matrix degradation [126]. However, a recent randomized control trial on 124 subjects with knee pain taking oleuropein as a dietary supplement on a daily basis for 6 months showed no difference to placebo with regard to serum levels of inflammatory and cartilage remodeling biomarkers and a significant effect was only reported in a subgroup of patients with severe walking pain at baseline [127].
As indicated in Table 4, bisphosphonates may have beneficial effect in knee OA of the bone-remodeling subtype. One meta-analysis of seven randomized, placebo-controlled trials of bisphosphonates in knee OA did not show an improvement in either symptoms or radiographic progression [128]. However, the authors suggested that bisphosphonates may be potentially beneficial in patients with high subchondral bone turnover. A recent randomized, double-blinded, placebo-controlled trial of zoledronic acid in 223 participants with knee OA, which used bone marrow lesions seen on MRI as evidence of subchondral bone remodeling, found that an early infusion of zoledronic acid did not reduce knee pain or the size of bone marrow lesions significantly over 24 months [129]. In addition, it has been reported that bisphosphonates may slow down radiographic progression, but their results were significant only in non-overweight patients with mild radiographic damage at baseline [130]. Their secondary analysis in obese patients found no significant effect [130]. These data show that the use of bisphosphonates in knee OA is likely not sufficiently beneficial and there is currently not enough evidence to support their use in cases of obesity-related knee OA. The use of calcitonin was also refuted by two randomized, double-blind, multi-center, placebo-controlled trials, which found no effect on joint space narrowing and non-significant effect on the total WOMAC score [131].
While initially considered especially promising in treating chronic OA pain, NGF inhibitors demonstrated considerable side effects including osteonecrosis and progressive cartilage damage, leading to a temporary suspension of clinical trials, which were resumed in 2012 [68,132]. Currently, lower doses of the anti-NGF monoclonal antibody tanezumab (2.5 to 5 mg, administered subcutaneously) have shown a statistically significant improvement in WOMAC pain and WOMAC function score, while having a favorable safety profile and an occurrence of rapidly-progressive OA in only 1.4–2.8% of subjects [133]. Another strategy to limit the adverse effects of NGF blockade could be to target its receptor, TrKA. A significant reduction in WOMAC pain and functional benefits has been reported in patients with painful knee OA following intra-articular administration of a TrKA inhibitor and an acceptable safety profile, with transient and self-limited reactions at the injection site [134]. Another novel therapeutic strategy targets TRPV-1 through intra-articular injections of capsaicin and resiniferatoxin (RTX). Highly purified synthetic trans-capsaicin, which specifically targets TRPV1-containing pain nociceptors while sparing sensory fibers for touch or pressure, has shown a rapid and significant impact on WOMAC walking pain scores and a favorable safety profile [135,136]. An injectable form of RTX has recently been developed and given a ‘breakthrough therapy’ status by the United States Food and Drug Administration (FDA) [137]. It is currently being investigated for the treatment of knee OA pain in a Phase III clinical trial but preliminary results from the Phase IB trial showed that 83% of patients had a statistically significant, clinically meaningful decrease in the WOMAC walking pain score [138]. Side effects included injection site pain, nausea, vomiting and headache but were manageable and well tolerated [138]. Despite these promising data, a separate analysis of these novel molecules in obesity-related knee OA is still lacking.
Anti-cytokines have long been studied as a possible disease-modifying treatment in OA, however results remain controversial. An open-label randomized control trial comparing the intra-articular administration of TNFα-inhibitor adalimumab versus hyaluronic acid in patients with moderate to severe knee OA reported that pain improvement on VAS and decrease of the WOMAC pain score were significantly greater in the adalimumab group [139]. A similar study of etanercept versus hyaluronic acid in moderate to severe knee OA also found a greater decrease in pain on VAS at post-injection weeks one and two and in the WOMAC pain score on post-injection week four in the group treated with the TNFα-inhibitor [140]. Conversely, the recent OKINADA randomized clinical trial on the efficacy and safety of adalimumab in inflammatory OA of the knee found no significant improvement in pain and joint function in patients with established radiographic knee OA [141]. A novel drug currently under investigation in patients with painful knee OA is the monoclonal antibody MEDI7352, which binds specifically to NGF and TNFα, thus blocking both mediators of pain and inflammation [68]. Autologous conditioned serum (ACS) is another therapeutic option with pain-modifying and anti-inflammatory properties, mainly mediated by the blockade of IL-1′s action via the IL-1 receptor antagonist (IL-1Ra). It is derived from the patient’s own blood and is rich in cytokines and growth factors secreted by platelets upon stimulation with glass beads [142]. A crucial step in the preparation is the incubation time, varying anywhere between 1 and 24 h, however a recent study found that incubation for 3 h had the optimal ratio between anti- to pro-inflammatory cytokines, with higher IL-1Ra levels and lower TNFα levels [143]. In addition, levels of IL-1Ra and platelet-derived growth factor (PDGF) were significantly higher in ACS than in platelet-rich plasma (PRP), suggesting better clinical efficacy [143]. Other authors compared the efficacy of intra-articular administration of ACS, PRP, hyaluronic acid and GCs and found that both blood-derived products performed better in terms of pain relief and functional improvement without a statistically significant difference between them [144]. Other studies, however, showed higher pain reduction and functional improvement with ACS compared to PRP, likely due to the higher IL-1Ra levels, decrease in IL-1β and improved synovial fluid viscosity [145,146]. Furthermore, it has been found that 67% of subjects with painful knee OA who had failed previous standard therapy, including physiotherapy and PRP, had a reduction of pain assessed on VAS one month after administration of ACS and this result persisted at 6 and 12 months of follow-up [147]. A notable finding from this study was that responders had significantly higher levels of IL-1Ra in their ACS compared to non-responders, underscoring the potential benefits of intra-articular IL-1β blockade in knee OA [147]. These reports suggest that anti-cytokine therapy could have potential benefits in obesity-related knee OA considering their role in maintaining the low-grade inflammatory state characteristic of obesity, however they are yet to be studied in such conditions. Finally, anti-obesity drugs have been proposed in patients with BMI > 30 and associated metabolic disorders, but their role in the therapy of concomitant knee OA requires further investigation [47].

6. Conclusions

Obesity-related knee OA is a separate entity that is not simply related to increased mechanical load but represents a disorder with complex pathogenesis driven by the chronic micro-inflammatory state, altered adipokine profile and sarcopenia seen in obese individuals. It is associated with higher pain score, decreased joint functionality and poorer quality of life. While recent years have seen a spike in research in that field, much remains to be elucidated. Future studies should focus on possible correlations between serum and synovial levels of adipokines such as leptin, resistin and adiponectin and mediators such as NGF and CGRP in order to address the molecular basis of already established clinical associations between their expression and pain severity. These could be essential in developing better pain management strategies. While current treatment guidelines do not feature specific recommendations for obesity-related knee OA, there is sufficient evidence to support exercise and weight loss. Viscosupplementation, glucosamine, chondroitin and natural antioxidants may also have potential benefits, whereas oral NSAIDs and GC injections should be avoided due to increased cardiovascular risk. Novel therapeutic approaches including intra-articular administration of inhibitors of key pain mediators such as NGF and TRPV-1 have yielded promising early results but are yet to be studied in the context of obesity.

Author Contributions

R.S.—conceptualization, supervision, writing—review and editing. G.K.—conceptualization, writing—original draft, writing—review and editing. P.E.—conceptualization, writing—review and editing. S.M.—conceptualization, supervision, project administration. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Palazzo, C.; Nguyen, C.; Lefevre-Colau, M.M.; Rannou, F.; Poiraudeau, S. Risk factors and burden of osteoarthritis. Ann. Phys. Rehabil. Med. 2016, 59, 134–138. [Google Scholar] [CrossRef]
  2. Chen, L.; Zheng, J.J.Y.; Li, G.; Yuan, J.; Ebert, J.R.; Li, H.; Papadimitriou, J.; Wang, Q.; Wood, D.; Jones, C.W.; et al. Pathogenesis and clinical management of obesity-related knee osteoarthritis: Impact of mechanical loading. J. Orthop. Translat. 2020, 24, 66–75. [Google Scholar] [CrossRef]
  3. Raud, B.; Gay, C.; Guiguet-Auclair, C.; Bonnin, A.; Gerbaud, L.; Pereira, B.; Duclos, M.; Boirie, Y.; Coudeyre, E. Level of obesity is directly associated with the clinical and functional consequences of knee osteoarthritis. Sci. Rep. 2020, 10, 3601. [Google Scholar] [CrossRef]
  4. Hunter, D.J.; Bierma-Zeinstra, S. Osteoarthritis. Lancet 2019, 393, 1745–1759. [Google Scholar] [CrossRef]
  5. Jansen, N.E.J.; Molendijk, E.; Schiphof, D.; van Meurs, J.B.J.; Oei, E.H.G.; van Middelkoop, M.; Bierma-Zeinstra, S.M.A. Metabolic syndrome and the progression of knee osteoarthritis on MRI. Osteoarthr. Cartil. 2023, 31, 647–655. [Google Scholar] [CrossRef]
  6. Conrozier, T. How to Treat Osteoarthritis in Obese Patients? Curr. Rheumatol. Rev. 2020, 16, 99–104. [Google Scholar] [CrossRef]
  7. Martel-Pelletier, J.; Barr, A.J.; Cicuttini, F.M.; Conaghan, P.G.; Cooper, C.; Goldring, M.B.; Goldring, S.R.; Jones, G.; Teichtahl, A.J.; Pelletier, J.P. Osteoarthritis. Nat. Rev. Dis. Primers 2016, 2, 16072. [Google Scholar] [CrossRef]
  8. 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]
  9. Monov, S.; Shumnalieva, R.; Sheytanov, I.; Kolarov, Z.; Rashkov, R. The effect of nonsteroidal anti-inflammatory drugs on articular cartilage metabolism. Balneoclimatologia 2013, 1, 267–273. [Google Scholar]
  10. Goldring, M.B.; Marcu, K.B. Cartilage homeostasis in health and rheumatic diseases. Arthritis Res. Ther. 2009, 11, 224. [Google Scholar] [CrossRef]
  11. Stanton, H.; Rogerson, F.M.; East, C.J.; Golub, S.B.; Lawlor, K.E.; Meeker, C.T.; Little, C.B.; Last, K.; Farmer, P.J.; Campbell, I.K.; et al. ADAMTS5 is the major aggrecanase in mouse cartilage in vivo and in vitro. Nature 2005, 434, 648–652. [Google Scholar] [CrossRef]
  12. Loeser, R.F. Molecular mechanisms of cartilage destruction: Mechanics, inflammatory mediators, and aging collide. Arthritis Rheum. 2006, 54, 1357–1360. [Google Scholar] [CrossRef]
  13. Roseti, L.; Desando, G.; Cavallo, C.; Petretta, M.; Grigolo, B. Articular Cartilage Regeneration in Osteoarthritis. Cells 2019, 8, 1305. [Google Scholar] [CrossRef]
  14. Ashruf, O.S.; Ansari, M.Y. Natural Compounds: Potential Therapeutics for the Inhibition of Cartilage Matrix Degradation in Osteoarthritis. Life 2022, 13, 102. [Google Scholar] [CrossRef]
  15. Wang, M.; Sampson, E.R.; Jin, H.; Li, J.; Ke, Q.H.; Im, H.J.; Chen, D. MMP13 is a critical target gene during the progression of osteoarthritis. Arthritis Res. Ther. 2013, 15, R5. [Google Scholar] [CrossRef]
  16. Glasson, S.S.; Askew, R.; Sheppard, B.; Carito, B.; Blanchet, T.; Ma, H.L.; Flannery, C.R.; Peluso, D.; Kanki, K.; Yang, Z.; et al. Deletion of active ADAMTS5 prevents cartilage degradation in a murine model of osteoarthritis. Nature 2005, 434, 644–648. [Google Scholar] [CrossRef]
  17. Maldonado, M.; Nam, J. The role of changes in extracellular matrix of cartilage in the presence of inflammation on the pathology of osteoarthritis. BioMed Res. Int. 2013, 2013, 284873. [Google Scholar] [CrossRef]
  18. Nam, J.; Aguda, B.D.; Rath, B.; Agarwal, S. Biomechanical thresholds regulate inflammation through the NF-kappaB pathway: Experiments and modeling. PLoS ONE 2009, 4, e5262. [Google Scholar] [CrossRef]
  19. Rigoglou, S.; Papavassiliou, A.G. The NF-kappaB signalling pathway in osteoarthritis. Int. J. Biochem. Cell Biol. 2013, 45, 2580–2584. [Google Scholar] [CrossRef]
  20. Chow, Y.Y.; Chin, K.Y. The Role of Inflammation in the Pathogenesis of Osteoarthritis. Mediat. Inflamm. 2020, 2020, 8293921. [Google Scholar] [CrossRef]
  21. Li, Z.; Dai, A.; Yang, M.; Chen, S.; Deng, Z.; Li, L. p38MAPK Signaling Pathway in Osteoarthritis: Pathological and Therapeutic Aspects. J. Inflamm. Res. 2022, 15, 723–734. [Google Scholar] [CrossRef]
  22. Zhou, Y.; Wang, T.; Hamilton, J.L.; Chen, D. Wnt/beta-catenin Signaling in Osteoarthritis and in Other Forms of Arthritis. Curr. Rheumatol. Rep. 2017, 19, 53. [Google Scholar] [CrossRef]
  23. Lietman, C.; Wu, B.; Lechner, S.; Shinar, A.; Sehgal, M.; Rossomacha, E.; Datta, P.; Sharma, A.; Gandhi, R.; Kapoor, M.; et al. Inhibition of Wnt/beta-catenin signaling ameliorates osteoarthritis in a murine model of experimental osteoarthritis. JCI Insight 2018, 3, e96308. [Google Scholar] [CrossRef]
  24. Li, G.; Yin, J.; Gao, J.; Cheng, T.S.; Pavlos, N.J.; Zhang, C.; Zheng, M.H. Subchondral bone in osteoarthritis: Insight into risk factors and microstructural changes. Arthritis Res. Ther. 2013, 15, 223. [Google Scholar] [CrossRef]
  25. Zhu, X.; Chan, Y.T.; Yung, P.S.H.; Tuan, R.S.; Jiang, Y. Subchondral Bone Remodeling: A Therapeutic Target for Osteoarthritis. Front. Cell Dev. Biol. 2021, 8, 607764. [Google Scholar] [CrossRef]
  26. Chen, Y.; Wang, T.; Guan, M.; Zhao, W.; Leung, F.K.; Pan, H.; Cao, X.; Guo, X.E.; Lu, W.W. Bone turnover and articular cartilage differences localized to subchondral cysts in knees with advanced osteoarthritis. Osteoarthr. Cartil. 2015, 23, 2174–2183. [Google Scholar] [CrossRef]
  27. 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]
  28. Mora, J.C.; Przkora, R.; Cruz-Almeida, Y. Knee osteoarthritis: Pathophysiology and current treatment modalities. J. Pain. Res. 2018, 11, 2189–2196. [Google Scholar] [CrossRef]
  29. De Lange-Brokaar, B.J.; Ioan-Facsinay, A.; van Osch, G.J.; Zuurmond, A.M.; Schoones, J.; Toes, R.E.; Huizinga, T.W.; Kloppenburg, M. Synovial inflammation, immune cells and their cytokines in osteoarthritis: A review. Osteoarthr. Cartil. 2012, 20, 1484–1499. [Google Scholar] [CrossRef]
  30. 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]
  31. Thomson, A.; Hilkens, C.M.U. Synovial Macrophages in Osteoarthritis: The Key to Understanding Pathogenesis? Front. Immunol. 2021, 12, 678757. [Google Scholar] [CrossRef]
  32. Liu, B.; Zhang, M.; Zhao, J.; Zheng, M.; Yang, H. Imbalance of M1/M2 macrophages is linked to severity level of knee osteoarthritis. Exp. Ther. Med. 2018, 16, 5009–5014. [Google Scholar] [CrossRef]
  33. Zeng, N.; Yan, Z.P.; Chen, X.Y.; Ni, G.X. Infrapatellar Fat Pad and Knee Osteoarthritis. Aging Dis. 2020, 11, 1317–1328. [Google Scholar] [CrossRef]
  34. Sowers, M.R.; Karvonen-Gutierrez, C.A. The evolving role of obesity in knee osteoarthritis. Curr. Opin. Rheumatol. 2010, 22, 533–537. [Google Scholar] [CrossRef]
  35. Gelber, A.C.; Hochberg, M.C.; Mead, L.A.; Wang, N.Y.; Wigley, F.M.; Klag, M.J. Body mass index in young men and the risk of subsequent knee and hip osteoarthritis. Am. J. Med. 1999, 107, 542–548. [Google Scholar] [CrossRef]
  36. Roemhildt, M.L.; Beynnon, B.D.; Gauthier, A.E.; Gardner-Morse, M.; Ertem, F.; Badger, G.J. Chronic in vivo load alteration induces degenerative changes in the rat tibiofemoral joint. Osteoarthr. Cartil. 2013, 21, 346–357. [Google Scholar] [CrossRef]
  37. Matheny, J.B.; Goff, M.G.; Pownder, S.L.; Koff, M.F.; Hayashi, K.; Yang, X.; Bostrom, M.P.G.; van der Meulen, M.C.H.; Hernandez, C.J. An in vivo model of a mechanically-induced bone marrow lesion. J. Biomech. 2017, 64, 258–261. [Google Scholar] [CrossRef]
  38. Sobieh, B.H.; El-Mesallamy, H.O.; Kassem, D.H. Beyond mechanical loading: The metabolic contribution of obesity in osteoarthritis unveils novel therapeutic targets. Heliyon 2023, 9, e15700. [Google Scholar] [CrossRef]
  39. Duclos, M. Osteoarthritis, obesity and type 2 diabetes: The weight of waist circumference. Ann. Phys. Rehabil. Med. 2016, 59, 157–160. [Google Scholar] [CrossRef]
  40. Wang, T.; He, C. Pro-inflammatory cytokines: The link between obesity and osteoarthritis. Cytokine Growth Factor. Rev. 2018, 44, 38–50. [Google Scholar] [CrossRef]
  41. Vieira-Potter, V.J. Inflammation and macrophage modulation in adipose tissues. Cell Microbiol. 2014, 16, 1484–1492. [Google Scholar] [CrossRef]
  42. Xu, H.; Kang, J.H.; Choi, S.E.; Park, D.J.; Kweon, S.S.; Lee, Y.H.; Kim, H.Y.; Lee, J.K.; Shin, M.H.; Lee, S.S. Increased adiponectin levels are associated with higher radiographic scores in the knee joint, but not in the hand joint. Sci. Rep. 2021, 11, 1842. [Google Scholar] [CrossRef]
  43. Ilia, I.; Nitusca, D.; Marian, C. Adiponectin in Osteoarthritis: Pathophysiology, Relationship with Obesity and Presumptive Diagnostic Biomarker Potential. Diagnostics 2022, 12, 455. [Google Scholar] [CrossRef]
  44. Jiang, H.; Pu, Y.; Li, Z.H.; Liu, W.; Deng, Y.; Liang, R.; Zhang, X.M.; Zuo, H.D. Adiponectin, May Be a Potential Protective Factor for Obesity-Related Osteoarthritis. Diabetes Metab. Syndr. Obes. 2022, 15, 1305–1319. [Google Scholar] [CrossRef]
  45. Cabral-Pacheco, G.A.; Garza-Veloz, I.; Castruita-De la Rosa, C.; Ramirez-Acuña, J.M.; Perez-Romero, B.A.; Guerrero-Rodriguez, J.F.; Martinez-Avila, N.; Martinez-Fierro, M.L. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int. J. Mol. Sci. 2020, 21, 9739. [Google Scholar] [CrossRef]
  46. Yan, M.; Zhang, J.; Yang, H.; Sun, Y. The role of leptin in osteoarthritis. Medicine 2018, 97, e0257. [Google Scholar] [CrossRef]
  47. Shumnalieva, R.; Kotov, G.; Monov, S. Obesity-Related Knee Osteoarthritis-Current Concepts. Life 2023, 13, 1650. [Google Scholar] [CrossRef]
  48. Vuolteenaho, K.; Koskinen, A.; Kukkonen, M.; Nieminen, R.; Päivärinta, U.; Moilanen, T.; Moilanen, E. Leptin enhances synthesis of proinflammatory mediators in human osteoarthritic cartilage—Mediator role of NO in leptin-induced PGE2, IL-6, and IL-8 production. Mediat. Inflamm. 2009, 2009, 345838. [Google Scholar] [CrossRef]
  49. Koskinen, A.; Vuolteenaho, K.; Nieminen, R.; Moilanen, T.; Moilanen, E. Leptin enhances MMP-1, MMP-3 and MMP-13 production in human osteoarthritic cartilage and correlates with MMP-1 and MMP-3 in synovial fluid from OA patients. Clin. Exp. Rheumatol. 2011, 29, 57–64. [Google Scholar]
  50. Ait Eldjoudi, D.; Cordero Barreal, A.; Gonzalez-Rodríguez, M.; Ruiz-Fernández, C.; Farrag, Y.; Farrag, M.; Lago, F.; Capuozzo, M.; Gonzalez-Gay, M.A.; Mera Varela, A.; et al. Leptin in Osteoarthritis and Rheumatoid Arthritis: Player or Bystander? Int. J. Mol. Sci. 2022, 23, 2859. [Google Scholar] [CrossRef] [PubMed]
  51. Shumnalieva, R.; Monov, S.; Kolarov, Z.; Rashkov, R. The role of Resistin in rheumatoid arthritis and other autoimmune diseases. Rheumatology 2013, 4, 30–33. [Google Scholar]
  52. Zheng, S.; Xu, J.; Xu, S.; Zhang, M.; Huang, S.; He, F.; Yang, X.; Xiao, H.; Zhang, H.; Ding, C. Association between circulating adipokines, radiographic changes, and knee cartilage volume in patients with knee osteoarthritis. Scand. J. Rheumatol. 2016, 45, 224–229. [Google Scholar] [CrossRef]
  53. Zhu, J.; Ruan, G.; Cen, H.; Meng, T.; Zheng, S.; Wang, Y.; Li, B.; Zhu, Z.; Han, W.; Winzenberg, T.; et al. Association of serum levels of inflammatory markers and adipokines with joint symptoms and structures in participants with knee osteoarthritis. Rheumatology 2022, 61, 1044–1052. [Google Scholar] [CrossRef]
  54. Azamar-Llamas, D.; Hernández-Molina, G.; Ramos-Ávalos, B.; Furuzawa-Carballeda, J. Adipokine Contribution to the Pathogenesis of Osteoarthritis. Mediat. Inflamm. 2017, 2017, 5468023. [Google Scholar] [CrossRef]
  55. Xiong, J.; Long, J.; Chen, X.; Li, Y.; Song, H. Dyslipidemia Might Be Associated with an Increased Risk of Osteoarthritis. BioMed. Res. Int. 2020, 2020, 3105248. [Google Scholar] [CrossRef]
  56. Pragasam, S.S.J.; Venkatesan, V. Metabolic Syndrome Predisposes to Osteoarthritis: Lessons from Model System. Cartilage 2021, 13, 1598S–1609S. [Google Scholar] [CrossRef]
  57. Choi, W.S.; Lee, G.; Song, W.H.; Koh, J.T.; Yang, J.; Kwak, J.S.; Kim, H.E.; Kim, S.K.; Son, Y.O.; Nam, H.; et al. The CH25H-CYP7B1-RORα axis of cholesterol metabolism regulates osteoarthritis. Nature 2019, 566, 254–258. [Google Scholar] [CrossRef]
  58. Alvarez-Garcia, O.; Rogers, N.H.; Smith, R.G.; Lotz, M.K. Palmitate has proapoptotic and proinflammatory effects on articular cartilage and synergizes with interleukin-1. Arthritis Rheumatol. 2014, 66, 1779–1788. [Google Scholar] [CrossRef]
  59. Hashimoto, K.; Akagi, M. The role of oxidation of low-density lipids in pathogenesis of osteoarthritis: A narrative review. J. Int. Med. Res. 2020, 48, 300060520931609. [Google Scholar] [CrossRef] [PubMed]
  60. Thijssen, E.; van Caam, A.; van der Kraan, P.M. Obesity and osteoarthritis, more than just wear and tear: Pivotal roles for inflamed adipose tissue and dyslipidaemia in obesity-induced osteoarthritis. Rheumatology 2015, 54, 588–600. [Google Scholar] [CrossRef] [PubMed]
  61. Andersson, M.; Haglund, E.; Aili, K.; Bremander, A.; Bergman, S. Associations between metabolic factors and radiographic knee osteoarthritis in early disease—A cross-sectional study of individuals with knee pain. BMC Musculoskelet. Disord. 2022, 23, 938. [Google Scholar] [CrossRef] [PubMed]
  62. Pan, F.; Tian, J.; Cicuttini, F.; Jones, G. Metabolic syndrome and trajectory of knee pain in older adults. Osteoarthr. Cartil. 2020, 28, 45–52. [Google Scholar] [CrossRef] [PubMed]
  63. Lee, S.; Kim, T.-N.; Kim, S.-H. Sarcopenic obesity is more closely associated with knee osteoarthritis than is nonsarcopenic obesity: A cross-sectional study. Arthritis Rheum. 2012, 64, 3947–3954. [Google Scholar] [CrossRef] [PubMed]
  64. Spanoudaki, M.; Giaginis, C.; Mentzelou, M.; Bisbinas, A.; Solovos, E.; Papadopoulos, K.; Paliokas, I.; Zidrou, C.; Cheimaras, A.; Hassapidou, M.; et al. Sarcopenia and Sarcopenic Obesity and Osteoarthritis: A Discussion among Muscles, Fat, Bones, and Aging. Life 2023, 13, 1242. [Google Scholar] [CrossRef] [PubMed]
  65. Komici, K.; Dello Iacono, A.; De Luca, A.; Perrotta, F.; Bencivenga, L.; Rengo, G.; Rocca, A.; Guerra, G. Adiponectin and Sarcopenia: A Systematic Review with Meta-Analysis. Front. Endocrinol. 2021, 12, 576619. [Google Scholar] [CrossRef] [PubMed]
  66. Pegreffi, F.; Balestra, A.; De Lucia, O.; Smith, L.; Barbagallo, M.; Veronese, N. Prevalence of Sarcopenia in Knee Osteoarthritis: A Systematic Review and Meta-Analysis. J. Clin. Med. 2023, 12, 1532. [Google Scholar] [CrossRef]
  67. Salaffi, F.; Ciapetti, A.; Carotti, M. The sources of pain in osteoarthritis: A pathophysiological review. Reum. Ital. J. Rheumatol. 2014, 66, 57–71. [Google Scholar] [CrossRef] [PubMed]
  68. Yu, H.; Huang, T.; Lu, W.W.; Tong, L.; Chen, D. Osteoarthritis Pain. Int. J. Mol. Sci. 2022, 23, 4642. [Google Scholar] [CrossRef]
  69. Stockl, S.; Eitner, A.; Bauer, R.J.; Konig, M.; Johnstone, B.; Grassel, S. Substance P and Alpha-calcitonin gene-related peptide differentially affect human osteoarthritic and healthy chondrocytes. Front. Immunol. 2021, 12, 722884. [Google Scholar] [CrossRef]
  70. Fu, K.; Robbins, S.R.; McDougall, J.J. Osteoarthritis: The genesis of pain. Rheumatology 2018, 57, iv43–iv50. [Google Scholar] [CrossRef]
  71. Soni, A.; Wanigasekera, V.; Mezue, M.; Cooper, C.; Javaid, M.K.; Andrew, J.P.; Tracey, I. Central sensitization in knee osteoarthritis: Relating presurgical brainstem neuroimaging and PainDETECT-based patient stratification to arthroplasty outcome. Arthritis Rheumatol. 2019, 71, 550–560. [Google Scholar] [CrossRef] [PubMed]
  72. Vincent, T.L. Peripheral pain mechanisms in osteoarthritis. Pain 2020, 161, S138–S146. [Google Scholar] [CrossRef] [PubMed]
  73. Miller, R.E.; Miller, R.J.; Malfait, A.M. Osteoarthritis joint pain: The cytokine connection. Cytokine 2014, 70, 185–193. [Google Scholar] [CrossRef] [PubMed]
  74. Hattori, T.; Shimo, K.; Niwa, Y.; Tokiwa, Y.; Matsubara, T. Association of chronic pain with radiologic severity and central sensitization in hip osteoarthritis patients. J. Pain. Res. 2021, 14, 1153–1160. [Google Scholar] [CrossRef] [PubMed]
  75. Ohashi, Y.; Fukushima, K.; Inoue, G.; Uchida, K.; Koyama, T.; Tsuchiya, M.; Uchiyama, K.; Takahira, N.; Takaso, M. Central sensitization inventory scores correlate with pain at rest in patients with hip osteoarthritis: A retrospective study. BMC Musculoskelet. Disord. 2020, 21, 595. [Google Scholar] [CrossRef] [PubMed]
  76. Ohashi, Y.; Fukushima, K.; Uchida, K.; Koyama, T.; Tsuchiya, M.; Saito, H.; Uchiyama, K.; Takahira, N.; Inoue, G.; Takaso, M. Adverse effects of higher preoperative pain at rest, a central sensitization-related symptom, on outcomes after total hip arthroplasty in patients with osteoarthritis. J. Pain. Res. 2021, 14, 3345–3352. [Google Scholar] [CrossRef]
  77. Pecchi, E.; Priam, S.; Gosset, M.; Pigenet, A.; Sudre, L.; Laiguillon, M.C.; Berenbaum, F.; Houard, X. Induction of nerve growth factor expression and release by mechanical and inflammatory stimuli in chondrocytes: Possible involvement in osteoarthritis pain. Arthritis Res. Ther. 2014, 16, R16. [Google Scholar] [CrossRef]
  78. Malfait, A.M.; Miller, R.E.; Miller, R.J. Basic mechanisms of pain in osteoarthritis: Experimental observations and new perspectives. Rheum. Dis. Clin. N. Am. 2021, 47, 165–180. [Google Scholar] [CrossRef]
  79. McNamee, K.E.; Burleigh, A.; Gompels, L.L.; Feldmann, M.; Allen, S.J.; Williams, R.O.; Dawbarn, D.; Vincent, T.L.; Inglis, J.J. Treatment of murine osteoarthritis with TrkAd5 reveals a pivotal role for nerve growth factor in non-inflammatory joint pain. Pain 2010, 149, 386–392. [Google Scholar] [CrossRef]
  80. Aso, K.; Walsh, D.A.; Wada, H.; Izumi, M.; Tomitori, H.; Fujii, K.; Ikeuchi, M. Time course and localization of nerve growth factor expression and sensory nerve growth during progression of knee osteoarthritis in rats. Osteoarthr. Cartil. 2022, 30, 1344–1355. [Google Scholar] [CrossRef]
  81. Miller, R.E.; Tran, P.B.; Das, R.; Ghoreishi-Haack, N.; Ren, D.; Miller, R.J.; Malfait, A.M. CCR2 chemokine receptor signaling mediates pain in experimental osteoarthritis. Proc. Natl. Acad. Sci. USA 2012, 109, 20602–20607. [Google Scholar] [CrossRef] [PubMed]
  82. Raghu, H.; Lepus, C.M.; Wang, Q.; Wong, H.H.; Lingampalli, N.; Oliviero, F.; Punzi, L.; Giori, N.J.; Goodman, S.B.; Chu, C.R.; et al. CCL2/CCR2, but not CCL5/CCR5, mediates monocyte recruitment, inflammation and cartilage destruction in osteoarthritis. Ann. Rheum. Dis. 2017, 76, 914–922. [Google Scholar] [CrossRef] [PubMed]
  83. Ishihara, S.; Obeidat, A.M.; Wokosin, D.L.; Ren, D.; Miller, R.J.; Malfait, A.M.; Miller, R.E. The role of intra-articular neuronal CCR2 receptors in knee joint pain associated with experimental osteoarthritis in mice. Arthritis Res. Ther. 2021, 23, 103. [Google Scholar] [CrossRef] [PubMed]
  84. Nekomoto, A.; Nakasa, T.; Ikuta, Y.; Ding, C.; Miyaki, S.; Adachi, N. Feasibility of administration of calcitonin gene-related peptide receptor antagonist on attenuation of pain and progression in osteoarthritis. Sci. Rep. 2023, 13, 15354. [Google Scholar] [CrossRef]
  85. Liu, F.; Su, F.; Zhang, T.; Liu, R.; Liu, N.; Dong, T. Relationship between knee osteophytes and calcitonin gene-related peptide concentrations of serum and synovial fluid in knee of osteoarthritis. Medicine 2023, 102, e34691. [Google Scholar] [CrossRef]
  86. Suri, S.; Gill, S.E.; Massena de Camin, S.; Wilson, D.; McWilliams, D.F.; Walsh, D.A. Neurovascular invasion at the osteochondral junction and in osteophytes in osteoarthritis. Ann. Rheum. Dis. 2007, 66, 1423–1428. [Google Scholar] [CrossRef]
  87. Walsh, D.A.; Mapp, P.I.; Kelly, S. Calcitonin gene-related peptide in the joint: Contributions to pain and inflammation. Br. J. Clin. Pharmacol. 2015, 80, 965–978. [Google Scholar] [CrossRef]
  88. Brenner, S.S.; Klotz, U.; Alscher, D.M.; Mais, A.; Lauer, G.; Schweer, H.; Seyberth, H.W.; Fritz, P.; Bierbach, U. Osteoarthritis of the knee—Clinical assessments and inflammatory markers. Osteoarthr. Cartil. 2004, 12, 469–475. [Google Scholar] [CrossRef]
  89. Radojčić, M.R.; Thudium, C.S.; Henriksen, K.; Tan, K.; Karlsten, R.; Dudley, A.; Chessell, I.; Karsdal, M.A.; Bay-Jensen, A.C.; Crema, M.D.; et al. Biomarker of extracellular matrix remodelling C1M and proinflammatory cytokine interleukin 6 are related to synovitis and pain in end-stage knee osteoarthritis patients. Pain 2017, 158, 1254–1263. [Google Scholar] [CrossRef]
  90. Leung, Y.Y.; Huebner, J.L.; Haaland, B.; Wong, S.B.S.; Kraus, V.B. Synovial fluid pro-inflammatory profile differs according to the characteristics of knee pain. Osteoarthr. Cartil. 2017, 25, 1420–1427. [Google Scholar] [CrossRef]
  91. Richter, F.; Natura, G.; Loser, S.; Schmidt, K.; Viisanen, H.; Schaible, H.G. Tumor necrosis factor causes persistent sensitization of joint nociceptors to mechanical stimuli in rats. Arthritis Rheum. 2010, 62, 3806–3814. [Google Scholar] [CrossRef] [PubMed]
  92. Li, L.; Li, Z.; Li, Y.; Hu, X.; Zhang, Y.; Fan, P. Profiling of inflammatory mediators in the synovial fluid related to pain in knee osteoarthritis. BMC Musculoskelet. Disord. 2020, 21, 99. [Google Scholar] [CrossRef] [PubMed]
  93. Barker, T.; Rogers, V.E.; Henriksen, V.T.; Aguirre, D.; Trawick, R.H.; Rasmussen, G.L.; Momberger, N.G. Serum cytokines are increased and circulating micronutrients are not altered in subjects with early compared to advanced knee osteoarthritis. Cytokine 2014, 68, 133–136. [Google Scholar] [CrossRef] [PubMed]
  94. Levescot, A.; Chang, M.H.; Schnell, J.; Nelson-Maney, N.; Yan, J.; Martinez-Bonet, M.; Grieshaber-Bouyer, R.; Lee, P.Y.; Wei, K.; Blaustein, R.B.; et al. IL-1beta-driven osteoclastogenic Tregs accelerate bone erosion in arthritis. J. Clin. Investig. 2021, 131, e141008. [Google Scholar] [CrossRef]
  95. Kusakabe, T.; Sawaji, Y.; Endo, K.; Suzuki, H.; Konishi, T.; Maekawa, A.; Murata, K.; Yamamoto, K. DUSP-1 induced by PGE2 and PGE1 attenuates IL-1beta-activated MAPK signaling, leading to suppression of NGF expression in human intervertebral disc cells. Int. J. Mol. Sci. 2021, 23, 371. [Google Scholar] [CrossRef]
  96. Mailhot, B.; Christin, M.; Tessandier, N.; Sotoudeh, C.; Bretheau, F.; Turmel, R.; Pellerin, È.; Wang, F.; Bories, C.; Joly-Beauparlant, C.; et al. Neuronal interleukin-1 receptors mediate pain in chronic inflammatory diseases. J. Exp. Med. 2020, 217, e20191430. [Google Scholar] [CrossRef]
  97. Yu, H.; Yao, S.; Zhou, C.; Fu, F.; Luo, H.; Du, W.; Jin, H.; Tong, P.; Chen, D.; Wu, C.; et al. Morroniside attenuates apoptosis and pyroptosis of chondrocytes and ameliorates osteoarthritic development by inhibiting NF-kappaB signaling. J. Ethnopharmacol. 2021, 266, 113447. [Google Scholar] [CrossRef]
  98. Chang, X.; Kang, Y.; Yang, Y.; Chen, Y.; Shen, Y.; Jiang, C.; Shen, Y. Pyroptosis: A Novel Intervention Target in the Progression of Osteoarthritis. J. Inflamm. Res. 2022, 15, 3859–3871. [Google Scholar] [CrossRef]
  99. Singh, P.; Kumar, N.; Singh, M.; Kaur, M.; Singh, G.; Narang, A.; Kanwal, A.; Sharma, K.; Singh, B.; Napoli, M.D.; et al. Neutrophil Extracellular Traps and NLRP3 Inflammasome: A Disturbing Duo in Atherosclerosis, Inflammation and Atherothrombosis. Vaccines 2023, 11, 261. [Google Scholar] [CrossRef]
  100. Chen, B.; Wang, Y.; Chen, G. New Potentiality of Bioactive Substances: Regulating the NLRP3 Inflammasome in Autoimmune Diseases. Nutrients 2023, 15, 4584. [Google Scholar] [CrossRef]
  101. Cheng, F.; Yan, F.F.; Liu, Y.P.; Cong, Y.; Sun, K.F.; He, X.M. Dexmedetomidine inhibits the NF-κB pathway and NLRP3 inflammasome to attenuate papain-induced osteoarthritis in rats. Pharm. Biol. 2019, 57, 649–659. [Google Scholar] [CrossRef] [PubMed]
  102. Pan, F.; Tian, J.; Mattap, S.M.; Cicuttini, F.; Jones, G. Association between metabolic syndrome and knee structural change on MRI. Rheumatology 2020, 59, 185–193. [Google Scholar] [CrossRef] [PubMed]
  103. Valdes, A.M. Metabolic syndrome and osteoarthritis pain: Common molecular mechanisms and potential therapeutic implications. Osteoarthr. Cartil. 2020, 28, 7–9. [Google Scholar] [CrossRef] [PubMed]
  104. Marshall, N.J.; Liang, L.; Bodkin, J.; Dessapt-Baradez, C.; Nandi, M.; Collot-Teixeira, S.; Smillie, S.J.; Lalgi, K.; Fernandes, E.S.; Gnudi, L.; et al. A role for TRPV1 in influencing the onset of cardiovascular disease in obesity. Hypertension 2013, 61, 246–252. [Google Scholar] [CrossRef] [PubMed]
  105. Storozhuk, M.V.; Zholos, A.V. TRP Channels as Novel Targets for Endogenous Ligands: Focus on Endocannabinoids and Nociceptive Signalling. Curr. Neuropharmacol. 2018, 16, 137–150. [Google Scholar] [CrossRef] [PubMed]
  106. Simon, V.; Cota, D. Mechanisms in endocrinology: Endocannabinoids and metabolism: Past, present and future. Eur. J. Endocrinol. 2017, 176, R309–R324. [Google Scholar] [CrossRef] [PubMed]
  107. Boer, C.G.; Radjabzadeh, D.; Medina-Gomez, C.; Garmaeva, S.; Schiphof, D.; Arp, P.; Koet, T.; Kurilshikov, A.; Fu, J.; Ikram, M.A.; et al. Intestinal microbiome composition and its relation to joint pain and inflammation. Nat. Commun. 2019, 10, 4881. [Google Scholar] [CrossRef] [PubMed]
  108. Schott, E.M.; Farnsworth, C.W.; Grier, A.; Lillis, J.A.; Soniwala, S.; Dadourian, G.H.; Bell, R.D.; Doolittle, M.L.; Villani, D.A.; Awad, H.; et al. Targeting the gut microbiome to treat the osteoarthritis of obesity. JCI Insight 2018, 3, e95997. [Google Scholar] [CrossRef]
  109. Kolasinski, S.L.; Neogi, T.; Hochberg, M.C.; Oatis, C.; Guyatt, G.; Block, J.; Callahan, L.; Copenhaver, C.; Dodge, C.; Felson, D.; et al. 2019 American College of Rheumatology/Arthritis Foundation Guideline for the Management of Osteoarthritis of the Hand, Hip, and Knee. Arthritis Care Res. 2020, 72, 149–162. [Google Scholar] [CrossRef]
  110. Bannuru, R.R.; Osani, M.C.; Vaysbrot, E.E.; Arden, N.K.; Bennell, K.; Bierma-Zeinstra, S.M.A.; Kraus, V.B.; Lohmander, L.S.; Abbott, J.H.; Bhandari, M.; et al. OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthr. Cartil. 2019, 27, 1578–1589. [Google Scholar] [CrossRef]
  111. Messier, S.P.; Resnik, A.E.; Beavers, D.P.; Mihalko, S.L.; Miller, G.D.; Nicklas, B.J.; deVita, P.; Hunter, D.J.; Lyles, M.F.; Eckstein, F.; et al. Intentional Weight Loss in Overweight and Obese Patients with Knee Osteoarthritis: Is More Better? Arthritis Care Res. 2018, 70, 1569–1575. [Google Scholar] [CrossRef] [PubMed]
  112. Bliddal, H.; Leeds, A.R.; Stigsgaard, L.; Astrup, A.; Christensen, R. Weight loss as treatment for knee osteoarthritis symptoms in obese patients: 1-year results from a randomised controlled trial. Ann. Rheum. Dis. 2011, 70, 1798–1803. [Google Scholar] [CrossRef] [PubMed]
  113. Anandacoomarasamy, A.; Leibman, S.; Smith, G.; Caterson, I.; Giuffre, B.; Fransen, M.; Sambrook, P.N.; March, L. Weight loss in obese people has structure-modifying effects on medial but not on lateral knee articular cartilage. Ann. Rheum. Dis. 2012, 71, 26–32. [Google Scholar] [CrossRef] [PubMed]
  114. Richette, P.; Poitou, C.; Garnero, P.; Vicaut, E.; Bouillot, J.L.; Lacorte, J.M.; Basdevant, A.; Clément, K.; Bardin, T.; Chevalier, X. Benefits of massive weight loss on symptoms, systemic inflammation and cartilage turnover in obese patients with knee osteoarthritis. Ann. Rheum. Dis. 2011, 70, 139–144. [Google Scholar] [CrossRef] [PubMed]
  115. Jenkinson, C.M.; Doherty, M.; Avery, A.J.; Read, A.; Taylor, M.A.; Sach, T.H.; Silcocks, P.; Muir, K.R. Effects of dietary intervention and quadriceps strengthening exercises on pain and function in overweight people with knee pain: Randomised controlled trial. BMJ 2009, 339, b3170. [Google Scholar] [CrossRef] [PubMed]
  116. Rannou, F.; Pelletier, J.P.; Martel-Pelletier, J. Efficacy and safety of topical NSAIDs in the management of osteoarthritis: Evidence from real-life setting trials and surveys. Semin. Arthritis Rheum. 2016, 45 (Suppl. S4), S18–S21. [Google Scholar] [CrossRef]
  117. Deveza, L.; Hunter, D. An update on the treatment of osteoarthritis in obese patients. Expert. Opin. Pharmacother. 2016, 17, 753–755. [Google Scholar] [CrossRef]
  118. Lv, Z.; Yang, Y.X.; Li, J.; Fei, Y.; Guo, H.; Sun, Z.; Lu, J.; Xu, X.; Jiang, Q.; Ikegawa, S.; et al. Molecular Classification of Knee Osteoarthritis. Front. Cell Dev. Biol. 2021, 9, 725568. [Google Scholar] [CrossRef]
  119. Altman, R.; Hackel, J.; Niazi, F.; Shaw, P.; Nicholls, M. Efficacy and safety of repeated courses of hyaluronic acid injections for knee osteoarthritis: A systematic review. Semin. Arthritis Rheum. 2018, 48, 168–175. [Google Scholar] [CrossRef]
  120. Conrozier, T.; Eymard, F.; Chouk, M.; Chevalier, X. Impact of obesity, structural severity and their combination on the efficacy of viscosupplementation in patients with knee osteoarthritis. BMC Musculoskelet. Disord. 2019, 20, 376. [Google Scholar] [CrossRef]
  121. Meng, Z.; Liu, J.; Zhou, N. Efficacy and safety of the combination of glucosamine and chondroitin for knee osteoarthritis: A systematic review and meta-analysis. Arch. Orthop. Trauma. Surg. 2023, 143, 409–421. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, Z.; Leong, D.J.; Xu, L.; He, Z.; Wang, A.; Navati, M.; Kim, S.J.; Hirsh, D.M.; Hardin, J.A.; Cobelli, N.J.; et al. Curcumin slows osteoarthritis progression and relieves osteoarthritis-associated pain symptoms in a post-traumatic osteoarthritis mouse model. Arthritis Res Ther. 2016, 18, 128. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, J.; Ma, J.; Gu, J.H.; Wang, F.Y.; Shang, X.S.; Tao, H.R.; Wang, X. Regulation of type II collagen, matrix metalloproteinase-13 and cell proliferation by interleukin-1beta is mediated by curcumin via inhibition of NF-kappaB signaling in rat chondrocytes. Mol. Med. Rep. 2017, 16, 1837–1845. [Google Scholar] [CrossRef] [PubMed]
  124. Nicoliche, T.; Maldonado, D.C.; Faber, J.; Silva, M. Evaluation of the articular cartilage in the knees of rats with induced arthritis treated with curcumin. PLoS ONE 2020, 15, e0230228. [Google Scholar] [CrossRef] [PubMed]
  125. Mikami, T.; Kim, J.; Park, J.; Lee, H.; Yaicharoen, P.; Suidasari, S.; Yokozawa, M.; Yamauchi, K. Olive leaf extract prevents obesity, cognitive decline, and depression and improves exercise capacity in mice. Sci. Rep. 2021, 11, 12495. [Google Scholar] [CrossRef] [PubMed]
  126. Feng, Z.; Li, X.; Lin, J.; Zheng, W.; Hu, Z.; Xuan, J.; Ni, W.; Pan, X. Oleuropein inhibits the IL-1beta-induced expression of inflammatory mediators by suppressing the activation of NF-kappaB and MAPKs in human osteoarthritis chondrocytes. Food Funct. 2017, 8, 3737–3744. [Google Scholar] [CrossRef] [PubMed]
  127. Horcajada, M.N.; Beaumont, M.; Sauvageot, N.; Poquet, L.; Saboundjian, M.; Costes, B.; Verdonk, P.; Brands, G.; Brasseur, J.; Urbin-Choffray, D.; et al. An oleuropein-based dietary supplement may improve joint functional capacity in older people with high knee joint pain: Findings from a multicentre-RCT and post hoc analysis. Adv Musculoskelet Dis. 2022, 14, 1759720X211070205. [Google Scholar] [CrossRef] [PubMed]
  128. Vaysbrot, E.E.; Osani, M.C.; Musetti, M.C.; McAlindon, T.E.; Bannuru, R.R. Are bisphosphonates efficacious in knee osteoarthritis? A meta-analysis of randomized controlled trials. Osteoarthr. Cartil. 2018, 26, 154–164. [Google Scholar] [CrossRef]
  129. Cai, G.; Aitken, D.; Laslett, L.; Hill, C.; March, L.; Wluka, A.E.; Wang, Y.; Blizzard, L.; Cicuttini, F.; Winzenberg, T.; et al. OP0016 A multicentre randomised controlled trial of zoledronic acid for osteoarthritis of the knee with bone marrow lesions. Ann. Rheum. Dis. 2018, 77, 57–58. [Google Scholar]
  130. Hayes, K.N.; Giannakeas, V.; Wong, A.K.O. Bisphosphonate Use Is Protective of Radiographic Knee Osteoarthritis Progression Among those With Low Disease Severity and Being Non-Overweight: Data From the Osteoarthritis Initiative. J. Bone Miner. Res. 2020, 35, 2318–2326. [Google Scholar] [CrossRef]
  131. Karsdal, M.A.; Byrjalsen, I.; Alexandersen, P.; Bihlet, A.; Andersen, J.R.; Riis, B.J.; Bay-Jensen, A.C.; Christiansen, C.; CSMC021C2301/2 investigators. Treatment of symptomatic knee osteoarthritis with oral salmon calcitonin: Results from two phase 3 trials. Osteoarthr. Cartil. 2015, 23, 532–543. [Google Scholar] [CrossRef] [PubMed]
  132. Siddiq, M.A.B.; Clegg, D.; Jansen, T.L.; Rasker, J.J. Emerging and New Treatment Options for Knee Osteoarthritis. Curr. Rheumatol. Rev. 2022, 18, 20–32. [Google Scholar] [CrossRef] [PubMed]
  133. Berenbaum, F.; Blanco, F.J.; Guermazi, A.; Miki, K.; Yamabe, T.; Viktrup, L.; Junor, R.; Carey, W.; Brown, M.T.; West, C.R.; et al. Subcutaneous tanezumab for osteoarthritis of the hip or knee: Efficacy and safety results from a 24-week randomised phase III study with a 24-week follow-up period. Ann. Rheum. Dis. 2020, 79, 800–810. [Google Scholar] [CrossRef] [PubMed]
  134. Krupka, E.; Jiang, G.L.; Jan, C. Efficacy and safety of intra-articular injection of tropomyosin receptor kinase A inhibitor in painful knee osteoarthritis: A randomized, double-blind and placebo-controlled study. Osteoarthr. Cartil. 2019, 27, 1599–1607. [Google Scholar] [CrossRef] [PubMed]
  135. Ghouri, A.; Conaghan, P.G. Update on novel pharmacological therapies for osteoarthritis. Ther. Adv. Musculoskelet. Dis. 2019, 11, 1759720X19864492. [Google Scholar] [CrossRef] [PubMed]
  136. Campbell, J.N.; Stevens, R.; Hanson, P.; Connolly, J.; Meske, D.S.; Chung, M.K.; Lascelles, B.D.X. Injectable Capsaicin for the Management of Pain Due to Osteoarthritis. Molecules 2021, 26, 778. [Google Scholar] [CrossRef] [PubMed]
  137. Szallasi, A. Resiniferatoxin: Nature’s Precision Medicine to Silence TRPV1-Positive Afferents. Int. J. Mol. Sci. 2023, 24, 15042. [Google Scholar] [CrossRef] [PubMed]
  138. Leiman, D.; Minkowitz, H.; Levitt, R.C.; Solanki, D.; Horn, D.; Janfaza, D.; Sarno, D.; Albores-Ibarra, N.; Bai, X.; Takeshita, K.; et al. Preliminary results from a phase 1b double-blind study to assess the safety, tolerability, and efficacy of intra-articular administration of resiniferatoxin or placebo for the treatment of moderate to severe pain due to osteoarthritis of the knee. Osteoarthr. Cartil. 2020, 28, S138. [Google Scholar] [CrossRef]
  139. Wang, J. Efficacy and safety of adalimumab by intra-articular injection for moderate to severe knee osteoarthritis: An open-label randomized controlled trial. J. Int. Med. Res. 2018, 46, 326–334. [Google Scholar] [CrossRef]
  140. Ohtori, S.; Orita, S.; Yamauchi, K.; Eguchi, Y.; Ochiai, N.; Kishida, S.; Kuniyoshi, K.; Aoki, Y.; Nakamura, J.; Ishikawa, T.; et al. Efficacy of Direct Injection of Etanercept into Knee Joints for Pain in Moderate and Severe Knee Osteoarthritis. Yonsei Med. J. 2015, 56, 1379–1383. [Google Scholar] [CrossRef]
  141. Maksymowych, W.; Bessette, L.; Lambert, R.G.; Carapellucci, A.; Appleton, C.T. OP0229 Osteoarthritis of the knee, inflammation, and the effect of adalimumab (OKINADA): A randomized placebo-controlled trial. Ann. Rheum. Dis. 2022, 81, 151–152. [Google Scholar] [CrossRef]
  142. Baltzer, A.W.; Moser, C.; Jansen, S.A.; Krauspe, R. Autologous conditioned serum (Orthokine) is an effective treatment for knee osteoarthritis. Osteoarthr. Cartil. 2009, 17, 152–160. [Google Scholar] [CrossRef] [PubMed]
  143. Cheng, P.G.; Yang, K.D.; Huang, L.G.; Wang, C.H.; Ko, W.S. Comparisons of Cytokines, Growth Factors and Clinical Efficacy between Platelet-Rich Plasma and Autologous Conditioned Serum for Knee Osteoarthritis Management. Biomolecules 2023, 13, 555. [Google Scholar] [CrossRef] [PubMed]
  144. Khurana, A.; Goyal, A.; Kirubakaran, P.; Akhand, G.; Gupta, R.; Goel, N. Efficacy of Autologous Conditioned Serum (ACS), Platelet-Rich Plasma (PRP), Hyaluronic Acid (HA) and Steroid for Early Osteoarthritis Knee: A Comparative Analysis. Indian J. Orthop. 2021, 55, 217–227. [Google Scholar] [CrossRef] [PubMed]
  145. 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. Iran. J. Allergy Asthma Immunol. 2020, 19, 243–252. [Google Scholar] [CrossRef]
  146. Shirokova, L.; Noskov, S.; Gorokhova, V.; Reinecke, J.; Shirokova, K. Intra-Articular Injections of a Whole Blood Clot Secretome, Autologous Conditioned Serum, Have Superior Clinical and Biochemical Efficacy Over Platelet-Rich Plasma and Induce Rejuvenation-Associated Changes of Joint Metabolism: A Prospective, Controlled Open-Label Clinical Study in Chronic Knee Osteoarthritis. Rejuvenation Res. 2020, 23, 401–410. [Google Scholar]
  147. Leone, R.; de Rosa, A.; Iudicone, P.; Fioravanti, D.; Capua, G.; Rossetti, F.; Isgro, M.A.; Pierelli, L. Pain control and functional improvement in patients treated by autologous conditioned serum after failure of platelet rich plasma treatments in knee osteoarthritis. Transfus. Med. 2021, 31, 357–364. [Google Scholar] [CrossRef]
Table 1. Major adipokines implicated in obesity-related knee OA and their impact.
Table 1. Major adipokines implicated in obesity-related knee OA and their impact.
AdipokineProposed Role in Knee OA
AdiponectinProtective; decreases the levels of TNFα; lowers IL-1β-stimulated expression of MMP-13; promotes TIMP-2 which inhibits MMP-2 and ADAMTS.
LeptinHarmful; upregulates IL-6, IL-1β, prostaglandin E2, MMP-1, 3, 9 and 13; mediates subchondral bone remodeling; correlates with higher pain score.
ResistinHarmful; upregulates MMPs and pro-inflammatory cytokines; correlates with development of cartilage and bone marrow lesions and higher pain score.
VisfatinHarmful; upregulates TNFα, IL-6, IL-1β, MMP-3 and 13 and ADAMTS-4 and 5; correlates with structural damage and OA symptoms.
ADAMTS–A Disintegrin and Metalloproteinase with Thrombospondin motifs; IL-1β–interleukin-1 beta; IL-6–interleukin-6; MMP–matrix metalloproteinase; TIMP–tissue inhibitor of matrix metalloproteinase-2; TNFα–tumor necrosis factor alpha.
Table 2. ACR and OARSI guidelines on the management of knee osteoarthritis by strength of recommendation [109,110].
Table 2. ACR and OARSI guidelines on the management of knee osteoarthritis by strength of recommendation [109,110].
Type of RecommendationACROARSI
Strongly recommendedExercise
Self-efficacy and self-management programs
Weight loss
Cane
Tibiofemoral knee brace
Tai Chi
Oral and topical NSAIDs
Intraarticular CSs
Core recommendation–Arthritis Education; Structured Land-Based Exercise Programs (Type 1–strengthening and/or cardio and/or balance training/neuromuscular exercise OR Type 2–Mind-body Exercise including Tai Chi or Yoga) with or without Dietary Weight Management
Topical NSAIDs
Conditionally recommendedHeat, Therapeutic cooling
Cognitive behavioral therapy
Acupuncture
Kinesiotaping
Balance training
Patellofemoral knee brace
Yoga
Acetaminophen
Tramadol
Duloxetine
Topical capsaicin
Aquatic exercise
Gait aids
Self-management programs
Cognitive behavioral therapy with exercise
Non-selective NSAIDs with or without PPIs (excluding patients with GI or CV comorbidities)
COX-2 inhibitors (excluding patients with CV comorbidities)
Intraarticular CSs
Intraarticular hyaluronic acid
COX-2–cyclooxygenase-2; CSs–corticosteroids; CV–cardiovascular; GI–gastrointestinal; NSAIDs–non-steroidal anti-inflammatory drugs; PPIs–proton pump inhibitors.
Table 3. Molecular subtypes of knee osteoarthritis and key mediators [118].
Table 3. Molecular subtypes of knee osteoarthritis and key mediators [118].
Knee Osteoarthritis SubtypeRepresentative Molecules
Cartilage degradation-driven subtypeCTX-II, C2C, C2M, C-Col X
Bone remodeling-driven subtypeALP, CTX-I, C1M, NTX-I
Pain-driven subtypeBradykinin, CGRP, hs-CRP, NGF
Inflammation-driven subtypeIL-1β, IL-1Ra, IL-6, TNFα
ALP–alkaline phosphatase; CGRP–calcitonin gene-related peptide; CTX-I–C-telopeptide fragments of collagen type I; CTX-II–C-telopeptide fragments of collagen type II; C2C–cleavage neoepitope of collagen type II; C1M–fragments of collagen type I degraded my matrix metalloproteinases; C2M–fragments of collagen type II degraded my matrix metalloproteinases; C-Col X–C-terminus of collagen type X; hs-CRP–high sensitive C-reactive protein; IL-1β–interleukin-1 beta; IL-1Ra–interleukin-1 receptor antagonist; IL-6–interleukin-6; NGF–nerve growth factor; NTX-I–N-telopeptide of collagen type I; TNFα–tumor necrosis factor alpha.
Table 4. Potential pathogenesis-based preferential therapeutic approaches based on molecular subtype of knee osteoarthritis [118].
Table 4. Potential pathogenesis-based preferential therapeutic approaches based on molecular subtype of knee osteoarthritis [118].
Knee Osteoarthritis SubtypeTreatment PrinciplePotential Beneficial Therapies
Cartilage degradation-driven subtypeSupplementation of cartilage extracellular matrix componentsHyaluronic acid, chondroitin, glucosamine, undenatured collagen type II
Bone remodeling-driven subtypeAnti-bone resorptionBisphosphonates, calcitonin, osteoprotegerin
Pain-driven subtypeAnalgesia, anti-inflammationNSAIDs, opioids, CGRP inhibitor, NGF inhibitor, capsaicin
Inflammation-driven subtypeAnti-inflammationTNFα-inhibitor, IL-1-inhibitor, IL-1Ra, NSAIDs, COX-2-inhibitor
CGRP–calcitonin gene-related peptide; COX-2–cyclooxygenase 2; IL-1–interleukin-1; IL-1Ra–interleukin-1 receptor antagonist; NSAIDs–non-steroidal anti-inflammatory drugs; NGF–nerve growth factor.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shumnalieva, R.; Kotov, G.; Ermencheva, P.; Monov, S. Pathogenic Mechanisms and Therapeutic Approaches in Obesity-Related Knee Osteoarthritis. Biomedicines 2024, 12, 9. https://doi.org/10.3390/biomedicines12010009

AMA Style

Shumnalieva R, Kotov G, Ermencheva P, Monov S. Pathogenic Mechanisms and Therapeutic Approaches in Obesity-Related Knee Osteoarthritis. Biomedicines. 2024; 12(1):9. https://doi.org/10.3390/biomedicines12010009

Chicago/Turabian Style

Shumnalieva, Russka, Georgi Kotov, Plamena Ermencheva, and Simeon Monov. 2024. "Pathogenic Mechanisms and Therapeutic Approaches in Obesity-Related Knee Osteoarthritis" Biomedicines 12, no. 1: 9. https://doi.org/10.3390/biomedicines12010009

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