Next Article in Journal
Importance of Genetic Polymorphisms in MT1 and MT2 Genes in Metals Homeostasis and Their Relationship with the Risk of Acute Pancreatitis Occurrence in Smokers—Preliminary Findings
Next Article in Special Issue
A Novel Method Facilitating the Simple and Low-Cost Preparation of Human Osteochondral Slice Explants for Large-Scale Native Tissue Analysis
Previous Article in Journal
Effect and Biocompatibility of a Cross-Linked Hyaluronic Acid and Polylactide-co-glycolide Microcapsule Vehicle in Intratympanic Drug Delivery for Treating Acute Acoustic Trauma
Previous Article in Special Issue
Vibrational Spectroscopy in Assessment of Early Osteoarthritis—A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Physical Exercise and Nutraceuticals in Modulating Molecular Pathways of Osteoarthritis

1
Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
2
Physical Medicine and Rehabilitation Unit, Department of Neurosciences, ASST Carlo Poma, 46100 Mantova, Italy
3
Physical Medicine and Rehabilitation, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
4
Translational Medicine, Dipartimento Attività Integrate Ricerca e Innovazione (DAIRI), Azienda Ospedaliera S.S. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(11), 5722; https://doi.org/10.3390/ijms22115722
Submission received: 29 April 2021 / Revised: 23 May 2021 / Accepted: 26 May 2021 / Published: 27 May 2021
(This article belongs to the Special Issue Osteoarthritis: From Molecular Pathways to Therapeutic Advances)

Abstract

:
Osteoarthritis (OA) is a painful and disabling disease that affects millions of patients. Its etiology is largely unknown, but it is most likely multifactorial. OA pathogenesis involves the catabolism of the cartilage extracellular matrix and is supported by inflammatory and oxidative signaling pathways and marked epigenetic changes. To delay OA progression, a wide range of exercise programs and naturally derived compounds have been suggested. This literature review aims to analyze the main signaling pathways and the evidence about the synergistic effects of these two interventions to counter OA. The converging nutrigenomic and physiogenomic intervention could slow down and reduce the complex pathological features of OA. This review provides a comprehensive picture of a possible signaling approach for targeting OA molecular pathways, initiation, and progression.

1. Introduction

Osteoarthritis (OA) is one of the most common degenerative musculoskeletal disorders, characterized by a progressive loss of joint cartilage, synovial inflammation, formation of osteophytes, and subchondral bone remodeling [1,2,3]. This detrimental condition has a complex pathogenesis due to its multifactorial nature [4,5,6]. More specifically, the aberrant expression of degradative proteases or catabolic mediators might be induced in the chondrocytes, which contribute to cartilage erosion [7,8]. This imbalance between anabolic and catabolic processes might damage the structural integrity of the joint cartilage, resulting in stiffness, pain, and limited range of motion (ROM) in the later stages of OA [9,10]. Thus, subsequent loss of function, increased disability, lower performance in the activities of daily living (ADL), and reduction of health-related quality of life (HRQoL) are common findings in these patients [11].
In this scenario, an early diagnosis supported by the detailed understanding of the molecular pathways underpinning OA could help to develop tailored conservative therapeutic approaches aimed to avoid surgical and joint replacement treatments [12]. To date, several non-surgical treatments have been proposed in the last years for OA patients, including pharmacological treatments (e.g., acetaminophen, nonsteroidal anti-inflammatory drugs (NSAIDs), duloxetine, and opioids) [13], intra-articular injections with hyaluronic acid and glucocorticoids [14], focal muscle vibration [15], intra-articular oxygen-ozone therapy [16], radiofrequency ablation of genicular nerves [17], adipose-derived mesenchymal stem cell therapy [18], and platelet-rich plasma injections [19]. However, physical exercise is recommended by several guidelines as the first-line intervention in OA patients [13,20,21], which plays a crucial role in the prevention and treatment of the disabling sequelae of this severe chronic disease [22,23].
Nowadays, other alternative therapeutic interventions to treat OA have come to the fore, such as nutraceuticals, defined as substances that can be considered as a food or part of a food and provides medical or health benefits, including OA prevention and treatment [24,25]. In vitro and in vivo studies showed that epigenetic changes are triggered by micronutrients commonly present in diets (e.g., vitamins, carotenoids, and flavonoids) that might modulate OA mediator pathways [26,27,28]. Indeed, myoblasts and chondrocytes seem to share similar pathological targets and pathways and close anatomical location, suggesting the possible existence of a paracrine network [29]. In this context, an adequate prescription of physical exercise and nutritional supplementation might have a positive impact not only in terms of OA molecular pathways modulation, but also in terms of functioning and HRQoL improvement.
Therefore, in the present comprehensive review, we sought to describe the state-of-the-art about the role that physical exercise and nutraceuticals might play in the complex management of OA, in terms of modulation of its molecular pathways.

2. Osteoarthritis Molecular Pathways

The pathogenic involvement in OA pathogenesis of pro-inflammatory cytokines released by chondrocytes and synoviocytes is well known and described in the scientific literature [7]. These cellular mediator patterns develop from cell signaling and gene expression pathways, which amplify the already altered cellular transduction, releasing additional inflammatory compounds and enzymes [30].

2.1. Reactive Oxygen Species

OA etiopathogenesis is influenced by several genetic and environmental factors not already fully explained. Recent studies have shown the involvement of oxidative stress and reactive oxygen species (ROS) in OA onset and progression [31]. ROS are unstable and highly reactive oxygen-containing free radicals combined with molecules to achieve chemical stability. Cells have developed antioxidant systems to scavenge ROS and maintain intracellular redox milieu. However, ROS are also key components of many physiological processes, and, at moderate concentrations, they act as indispensable second messengers. Their activity is based on the alterations of the cellular chemical environment, consisting of oxidative modification of proteins, influencing signal transduction, gene regulation, and cell cycling, in a complex process called redox biology [32]. The ability of cells to discriminate between the opposing effects of ROS is dependent on intensity, duration, and context of signaling and cellular redox status. When the cellular antioxidant capacity is insufficient to detoxify, ROS reacts with DNA, proteins, and lipids, disrupting their normal structure, impairing function, and leading to cytotoxicity in a process called oxidative stress [33].
Articular cartilage exists in relatively hypoxic conditions as a unique tissue thriving in a mechanically active environment. Despite living in a low O2 environment, chondrocytes are characterized by abundant and active mitochondria that contribute to adenosine triphosphate (ATP) production [34]. Disruption of mitochondrial function, leading to increased levels of intracellular ROS, has been hypothesized to disrupt cartilage homeostasis and is considered as one of the main contributors of OA-related cartilage damage [35].
In physiological conditions, ROS are produced at low concentrations in chondrocytes mitochondria through oxidative phosphorylation and in cytoplasm by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. The overproduction of ROS observed in OA impairs mitochondrial functions through mtDNA damage, inducing chondrocyte senescence and apoptosis, increasing cartilage degradation, and reducing matrix synthesis along with subchondral bone disfunction and synovial inflammation [36,37,38]. Evidence for ROS implication in cartilage degradation comes from the presence of lipid peroxidation products, nitrite, and nitrated products in the biological fluids in OA animal models [36]. On the contrary, antioxidant enzyme concentrations are decreased in OA patients, confirming the role that oxidative stress might play in OA pathogenesis [39].
Prolonged oxidative stress induces in the cartilaginous tissue the synthesis of large amounts of proteolytic enzymes, which in turn favors the shift towards catabolic reactions through several kinases such as p38 mitogen-activated protein kinase/nuclear factor kappa-light-chain-enhancer of activated B cells (p38MAPK/NF-κB) and activin receptor-like kinase 1 (ALK1) pathways [30]. In response to the proinflammatory stimuli, the overproduction of nitric oxide (NO) suppresses the cartilage matrix synthesis, enhances matrix metalloproteinases (MMPs) activity and induces chondrocyte apoptosis [40]. Taken together, all these mechanisms increase ROS concentrations leading to the membrane potential alterations that result in the release of cytochrome c and increase in Caspase-3 activity leading to apoptosis [41]. Lastly, oxidative stress-mediated inflammation is also responsible for the increased rate of hyaluronan degradation in synovial fluid [42].
On the other hand, chondrocytes respond to hypoxia and ROS damage, increasing the production of hypoxia-inducible factor-1 (HIF-1). HIF-1α can alleviate hypoxia-induced apoptosis, senescence, and matrix degradation in chondrocytes through mitophagy enhancing BCL2/adenovirus E1B 19-kDa-interacting protein 3 (BNIP3) expression [43].

2.2. Cellular Apoptosis

In the context of cellular autophagy, the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT)/mechanistic target of rapamycin (mTOR) survival pathway plays a crucial role in OA pathogenesis. However, the role of Wnt/β-catenin and extracellular signal-regulated kinases (ERKs) pathway in OA pathogenesis is less elucidated [44]. In PI3K/mTOR signaling, AKT phosphorylated by PI3K modulates mTOR through a direct activation or an indirect block of one of its inhibitors. Activation of PI3K/mTOR pathway curbs the pro-apoptotic machinery resulting in enhanced cell survival through Caspase-3 inhibition [44,45]. Indeed, PI3K/mTOR is inactivated in IL-1b mediated chondrocyte apoptosis [46]. Alongside these mechanisms, Wnt proteins stand out through two characteristic pathways: β-catenin-independent pathway and β-catenin-dependent pathway [47]. In the independent process, the β-catenin undergoes phosphorylation, resulting in ubiquitination and consequent proteasomal degradation. On the other hand, in the β-catenin-dependent pathway, after the binding of Wnt ligands to the transmembrane frizzled (Fzd) receptor, the disheveled protein prevents the phosphorylation of β-catenin inducing the translocation into the nucleus where it stimulates the transcription of anti-apoptotic genes including c-Myc and cyclin D1 [48,49]. Lastly, recent studies seem to show that ERKs might be able to increase rather than decrease the levels of anabolic biomarkers in degenerative human chondrocyte [50,51,52].

2.3. Pro-Inflammatory Signaling

Alongside chondrocyte apoptosis, the scientific literature has recently focused on the mediation of inflammatory gene expression and inflammatory pathways role in OA pathogenesis. NF-κB is a protein complex that controls transcription of DNA and cytokine production [53]. In unstimulated cells, the NF-κB dimers are sequestered in the cytoplasm by a family of inhibitors called inhibitors of κB (IκBs) [54]. High Mobility Group Box 1 (HMGB1) is a non-histone DNA binding protein and is considered a damage-associated molecular pattern protein (DAMP) [55]. The binding of the extracellular ligand HMGB1 to the advanced glycation end product receptor (RAGE) has been described to activate IκB kinase (IKK), resulting in phosphorylation and degradation of IκBα [56].
Subsequently, p65 protein is released and phosphorylated for NF-κB heterodimer formation, and this complex is then translocated from the cytoplasm to the nucleus, inducing the expression of several genes such as cyclooxygenase (COX), Matrix metalloproteinases (MMPs), and pro inflammatory cytokines [10].
This pathway is associated with mitogen-activated protein kinase (MAPK) signaling which constitutes a family of serine/threonine kinases (p38 MAPK; c-Jun amino-terminal kinase (JNK); and ERK1/2) characterized by a multilevel crosstalk and activated by a large array of inflammatory and stressful conditions [57,58]. In addition to the ERKs, the activated p38 MAPK can translocate into the nucleus upregulating kinases and transcription factors promoting cell autophagy in some cells while enhancing survival, growth, and differentiation in other cells [57]. Lastly, the phosphorylation of JNK via specific tyrosine and threonine residues is able to increase pro-inflammatory gene expression [58].

2.4. Anti-Inflammatory Signaling

Nuclear erythroid factor 2 (Nrf2) is a distress mediator involved in oxidative stress reduction [59]. It operates as a transcription factor binding to the antioxidant response elements (AREs) in the promoter region of antioxidant genes. In unstimulated chondrocytes, Nrf2 is kept quiescent through the association with kelch-like ECH-associated protein 1 (Keap1) in the cytosol, hence restricting its translocation into the nucleus to bind the AREs [60]. DJ-1 is a sensitive redox protein that serves as a stabilizer of Nrf2; in DJ-1 knock-out animal models, the downregulation of Nrf2 could lead to an increase in oxidative stress. In the same context, heme oxygenase-1 (HO-1) is an Nrf2 downstream effector and both proteins are compensatory mediators that recognize pro-oxidative stressors and induce the production of antioxidant enzymes to neutralize the ROS [61]. Emerging evidence showed that the activation of DJ-1/Nrf2 signaling in chondrocytes is crucial to protecting these cells against oxidative insults, downregulating HMGB1 and NF-κB protein expression, and activating the PI3K/mTOR pathway [62].
In summary, the activation of NF kB, p38MAPK, and JNK is involved in the expression of several inflammatory genes that boost OA pathogenesis and progression. On the other hand, the upregulation of NRf2 could allow the expression of genes leading to a suppression of inflammation and inhibition of the activation of NF-κB. In this scenario, the PI3K/mTOR, Wnt, and ERK pathways appear to block cell apoptosis and promote proliferation.

3. Physical Exercise as a Modulator of Osteoarthritis Molecular Pathways

Physical exercise has been recognized as a safe, effective, and multifaceted therapeutic treatment to reduce pain and disability in OA patients [13,20,22,23,63].
The American College of Sports Medicine guidelines and EULAR recommend physical activity and exercise as positive and effective interventions on physical fitness as well as disease-specific and general outcomes in people with hip and knee OA. Moreover, these guidelines suggest that physical activity and exercise should be considered as integral parts of standard care [64,65].
Recommended exercise programs include land-based therapeutic exercise and water physiotherapy interventions [66,67]. Indeed, a recent systematic review confirmed that Pilates, aerobic, and strengthening exercise programs performed for 8–12 weeks, 3–5 sessions per week, with each session lasting 1 h, could be considered as an effective treatment in OA patients [68].
The exact protective mechanism of action of physical exercise on cartilage is not yet known, albeit several studies performed on animal models highlighted key molecules that could help define the best therapeutic exercise regimen.
Exercise modulates transcription in several metabolic pathways associated with extracellular matrix (ECM) biosynthesis and inflammation/immune responses in normal cartilage of rats undergoing treadmill walking (12 m/min, 45 min/day for 2, 5, or 15 days). More specifically, low-intensity exercises were able to regulate the different pathways involving PI3K-AKT, NF-κB, Ras, Rap, MAPK, cAMP, and Ptgs2. Moreover, exercise was able to suppress the expression of genes involved in ECM degradation, bone formation, and initiation of pro-inflammatory cascades, which are known to be upregulated in OA (Mmp9, Mmp8, Igf1, ColIa1, Adamts3, Adamts14). Conversely, exercise can upregulate genes involved in ECM synthesis that are commonly downregulated in OA (Chrdl2, Tnfrsf11b, Timp4, Thbs2, Tgfb1, Mmp3, Il1r1, Il1r2, Cilp, and Bmp5) [29]. Taken together, these results suggest a crucial and multifaceted role of physical exercise in several molecular pathways involved in OA pathogenesis and progression.

3.1. Chondroprotective Role of Physical Exercise

OA animal models have been widely used in the research field to investigate the role of physical exercise in modulating molecular pathways involved in OA pathogenesis, highlighting a positive effect on cartilage preservation.
Iijima and colleagues showed that low-speed treadmill walking exercise (12 m/min for 30 min/day, 5 days/week performed for 2–4 weeks) prevented the progression of post-traumatic bone and cartilage lesions and increased BMP-2 and BMP-6 expression in the chondrocytes of the joint superficial zone of 24 male Wistar rats with induced damage to knee joints [69].
Assis et al. studied the effects of aerobic exercise performed with treadmill, training 3 days/week at 16 m/min for 50 min/day for 8 weeks, on an animal experimental model of knee OA. At the end of the treatment protocol, the exercise group had a better pattern of cartilage organization with less fibrillation and irregularities along the articular surface and a lower degenerative process, compared with controls. Moreover, animals in the exercise group had a lower chondrocyte nuclear or nucleolar expression of IL-1β, Caspase-3, and MMP-13, confirming the ability of aerobic exercise to downregulate proinflammatory, proteolytic, and apoptotic pathways [70]. Similarly, treadmill exercise with moderate-intensity (18 m/min, 60 min/day, 5 days/week for 8 weeks) could have a chondroprotective effect in an OA animal model through the inhibition of NF-κB expression, resulting in a potent inhibition of MMP-13 gene expression [71].
Moderate-intensity exercise performed for only 4 weeks can also inhibit nuclear translocation of HDAC3/NF-κB complex, leading to the decreased expression of inflammatory proteins and cartilage protection in a rat model of OA. Similarly, in isolated primary rat chondrocytes mimicking OA-like pathologies, RGFP966, an HDAC3-specific inhibitor, could reduce IL-1β-induced inflammation and ROS production through the inhibition of nuclear translocation of NF-κB. Moreover, the same results were observed in a OA rat model, suggesting a role of RGFP966 in exerting exercise mediated chondroprotective effects [72]. Lastly, Castrogiovanni et al. demonstrated that a moderate-intensity physical activity protocol could lead to a decreased expression of OA-related biomarkers (IL-1, TNFα, MMP-13) and an increased expression of chondroprotective ones (IL-4, IL-10, and lubricin) in the synovium of an OA-induced rat model, highlighting the beneficial effects of exercise on cartilage preservation [73].

3.2. Anti-Inflammatory and Anti-Apoptotic Role of Physical Exercise on Murine Models

The anti-inflammatory effects of physical exercise in OA have been widely investigated on murine models. Here, a short review of the main findings showing the positive effects of physical exercise at different levels in OA damaged joints will be performed.
One hour treadmill activity showed an increase in Maresin-1 content, a strong anti-inflammatory molecule, in intra-articular lavage fluid of MIA-induced rat OA. Maresin-1 was able to decrease MMP13, activate the PI3k/Akt pathway, and suppress the NF-κB p65 pathway in IL-1β-induced rat fibroblast-like synoviocytes in vitro, suggesting a pivotal role of Maresin-1 in exerting the anti-inflammatory activity of exercise [74].
Similarly, four weeks of moderate exercise in a monosodium iodoacetate (MIA)-induced OA model determined IL-1β reduction and IL-4 increase in both serum and intra-articular lavage fluid, resulting in anti-inflammatory effect at cartilage tissue level. Furthermore, immunohistochemical assays showed that exercise markedly promoted the expression of autophagosome proteins LC3B, SQSTM1, and LC3II in the whole cartilage tissue, suggesting a positive role of exercise in autophagosome’s reaction to pathological stimuli [75].
Moderate intensity treadmill training in MIA-induced OA determined an increase in 15-hydroxyeicosatetraenoic acid (15-HETE) concentration in knee joint. 15-HETE is a well-known anti-inflammatory molecule and was also able to inhibit IL-1β-induced inflammation in primary chondrocytes and increase p-Akt levels in vitro. Moreover, 15-HETE injection in knee OA alleviated cartilage damage through the inhibition of MMP-13 expression and the concomitant increase in COL2 expression in joint cartilage tissue [76].
Other animal experimental models of OA included anterior cruciate ligament transection. A study performed on this specific animal model revealed that 4 months of moderate exercise determined the reduction of Caspase3 expression and concomitant overexpression of Hsp70, an antiapoptotic factor, in both superficial and deep zones of the knee medial compartment [77]. Similarly, in surgically-induced rat knee OA, low-speed treadmill exercise (12 m/min, 5 days/week for 4 weeks) increased pSmad-5, Id1 and BMP-2, BMP-4, BMP-6, and BMP receptor 2 expression in the superficial zone chondrocytes and suppressed cartilage degeneration [69].
In this scenario, it should be noted that physical exercise could provide beneficial effects in OA according to several factors, including adherence to exercise regimens, frequency of exercise, and actual loading of the symptomatic compartment [76,78]. Nam et al. [78] demonstrated that the extent of cartilage damage could play a key role in achieving the optimal effects of exercise. They conducted a study using a common treadmill exercise protocol (speed of 12 m/min for 45 min/day) after MIA-induction damage at knee level in rats, demonstrating that exercise significantly prevented the OA progression, albeit its efficacy appeared to be inversely related to the extent of cartilage damage. Transcriptome-wide gene expression analysis revealed that gentle treadmill activity started 1 day post–MIA OA induction significantly suppressed inflammation-associated genes through NF-κB network downregulation, with crucial effects on apoptosis and cell cycle (Cdkn1a and Bcl2), matrix breakdown (Mmp12 and Mmp14), and proinflammatory responses (IL15 and IL18) in murine models. On the other hand, a delayed intervention after grade 1 cartilage damage could be less effective in suppressing proinflammatory genes or upregulating matrix synthesis. These effects were mainly related to Sox9 suppression leading to the down-regulation of alkaline phosphatase, Cilp, Cilp2, and Mgp, which are crucial mediators required for correct matrix assembly [78].

3.3. Beneficial Effects of Physical Exercise on Osteoarthritis Patients

Evidence observed on rat and mouse OA models have provided further insights that have been translated to humans since the early disease stages. The beneficial effect of exercise has been already showed in subjects at high risk of developing OA and alterations in articular cartilage composition could be a good marker of early OA pathological modifications [79].
In a randomized trial, patients at a high risk of developing radiographic OA for partial medial meniscus resection were subjected to 4 months weight-bearing exercise protocol. Cartilage quality, comprising GAG, was evaluated through fixed-charge density tissue measurement by gadolinium-enhanced MRI (dGEMRIC). The exercise group showed an improvement in dGEMRIC, compared with controls (p = 0.036), suggesting that human cartilage has a potential to adapt to loading changes [80]. This adaptation consequent to exercises varies between different locations within the joint, and the largest beneficial effect is observed in the load-bearing cartilage, such as the lateral posterior cartilage [81].
dGEMRIC index changes were also investigated in the study of Munukka et al. The authors evaluated 87 postmenopausal women with mild knee OA performing leisure-time physical activity (LTPA). After a period of 12 months, a linear relationship between higher LTPA level and increased dGEMRIC index changes in the posterior region of the lateral and medial femoral cartilage, were observed. These results suggested that higher LTPA level is related to regional increases in estimated glycosaminoglycan content of tibiofemoral cartilage [82]. Similarly, a randomized controlled trial conducted on 43 post-menopausal women with mild knee OA performing lower limb aquatic resistance training, showed a significantly reduction of T2-MRI scores in the posterior region of the medial femoral condyle after 4 months of physical activity. A decrease in T2 scores is suggestive of improved integrity and orientation of the collagen fibers and a decrease in hydration of articular cartilage [82].
Another promising technique to assess OA-related cartilage modifications is the microdialysis method. Helmark et al. obtained information about the effect of a single-leg knee-extension protocol on cartilage biomarkers and cytokines, both inside the joint as well as in the synovium, in a group of women affected by knee OA. In this RCT, over a period of three hours, a single bout of mechanical loading resulted in a significant decrease in cartilage oligomeric matrix proteins (COMP) and an increase in the anti-inflammatory mediator IL-10 in both intra-articular and peri-synovium spaces, compared with controls (p < 0.05). Significant increases were also registered for IL-6 and IL-8 in both groups, whereas TNF-α increased peri-synovially in the experimental group only [83].
Similarly, a pilot RCT performed by Hunt et al. demonstrated that a 10-week exercise protocol, consisting of hip abductors, quadriceps, and hamstrings muscle strengthening, performed on seventeen subjects with radiographically confirmed medial tibiofemoral OA, could lead to a reduction in serum of the cartilage degradation marker COMP levels, compared with controls (2.11 log U/L vs. 2.36 log U/L; p < 0.04). Moreover, a significant relationship emerged between dynamic measure of knee joint load and the ratio between two molecular markers increased in OA patient biological fluids (the urinary C-telopeptide of type II collagen/serum C-propeptide of type II procollagen ratio β = 1.11, 95% CI = 0.15, 2.07; p = 0.04) [84]. Lastly, the combined nutritional and exercise intervention also seem to act on cartilage biomarkers. A recent study demonstrated that an 18-month period aerobic walking and strength training in combination with an intensive diet determined the reduction of serum collagen type I (C1M) and II (C2M) in obese elderly patients affected by tibiofemoral OA [85].

3.4. Physical Exercise as an Antioxidant Intervention

The role of physical activity on oxidative stress and redox biology balance has recently obtained an emerging interest in the scientific literature. It is already known that physical activity might improve antioxidant defenses and lowers lipid peroxidation levels in both adults and aged individuals [86]. At cellular level, the beneficial role of physical exercise in OA comes through the increased expression of chondrogenic transcription factors SOX9 in circulating mesenchymal progenitors associated with autophagy. Autophagy regulates mitochondrial activity in stem cells to provide the best metabolic conditions, thus limiting ROS production and preventing metabolic stress and genome damage [87].
Furthermore, aerobic exercise modulates the chondrocyte response according to the balance between redox milieu and mechano-transduction mechanisms. Chondrocytes subjected to mechanical stimulation activate the NF-κB signaling pathway, which is considered a possible link between loading and chondrocytic responses to proinflammatory cytokines [88].
Nrf2 is a stress response protein in OA chondrocytes with anti-oxidative and anti-apoptotic function and acts via ERK1/2/ELK1-P70S6K-P90RSK signaling axis in vitro [89]. Physical exercise could activate Nrf2 in response to the ROS increase, leading to the expression of antioxidant genes such as phase-II antioxidant enzymes such as glutathione and NADPH through Nrf2/Maf/ARE pathways activation [90].
Although researchers and clinicians have demonstrated a particular attention to the role of physical exercise antioxidant effects on OA, at present, evidence in literature about this issue is scarce. In MIA-induced OA rat model, 8 weeks of endurance exercise training (13 m/min, 50 min/day for 3 days/week) promoted free radicals generation that could stimulate both exercise adaptation and mitochondrial biogenesis. In this scenario, exercise promoted a significant increase in myeloperoxidase and superoxide dismutase activity in the articular capsule [91]. Similarly, a two-group cross-sectional study showed that strengthening exercises could changes antioxidant status of systemic markers in knee OA patients. More specifically, ten obese women affected by knee OA, following an acute bout of isokinetic exercise, showed an immediate post-exercise rise of non-enzymatic antioxidant capacity of serum sustained by scavenging activity of 1,1-diphenyl-2-picrylhydrazyl. This finding suggests that the up-regulation of the body’s antioxidant defense could be probably related to the mobilization of antioxidant reserves in plasma [92].
In the complex management of OA, physical exercise turned out to be effective also in synergy with other innovative therapies (i.e., mesenchymal cell implants or injections). In this scenario, autologous chondrocyte implants and bone marrow-derived mesenchymal cell implants or injections are innovative therapeutic interventions in OA focused on articular cartilage and subchondral bone damage treatment [93,94,95]. Studies in humans showed that exercise might enhance joint recruitment of bone marrow-derived mesenchymal stem cells and upregulates the expression of osteogenic and chondrogenic genes (Runx, MSx1, Sox9, COL2A1, ATG3), osteogenic microRNAs, and osteogenic growth factors (BMP2, BMP6) [87,96,97]. In experimental rodent models, physical exercise enhanced the osteogenic potential of bone marrow-derived mesenchymal stem cells and reduced their adipogenic potential. Moreover, exercise performed after stem cell implantation could enhance stem cell transplant viability [98,99,100,101,102]. Taken together, these results suggest a complex and multifaceted role of physical exercise on different antioxidant pathways corroborating the physiological basis supporting its efficacy in the treatment of OA.

3.5. Challenges and Potential Controversies

Despite its countless scientifically proven beneficial effects, physical exercise in certain conditions could be detrimental to articular cartilage health. Studies on human subjects reported that even moderate exercise, recommended as a treatment for OA, might increase markers of cartilage degradation in OA-affected joints [103,104]. Due to these controversies, the investigation of the imbalances in the molecular pathways involved in OA pathogenesis and progression could help in providing more insights about this controversial issue.
In an exercise-induced OA rat model, two weeks of 16 m/min treadmill activity showed that Ctnnb1, β-catenin, and Wnt-3a participate in the pro-inflammatory OA pathogenesis through an abnormal activation of the Wnt/β-catenin pathway due to excessive mechanical load [105]. Similarly a high-impact exercise protocol comprising flexion, extension, and compression of the limbs for two weeks resulted in IL-1β overexpression and IL-10 down-regulation in an advanced stage of OA induced in a rat model [106]. Siebelt et al. highlighted the impact of a 6-week treadmill intense exercise protocol in rats highlighting the loss of proteoglycan content in the exercised compared with sedentary OA joints induced by papain injection [104]. In surgically-induced rat knee OA, the same treadmill velocity was not able to modulate the effect of BMP pathway on OA pathological modifications, leading to the progression of morphological alterations [69]. Similarly, anterior cruciate ligament transected-animals running at a lower speed (18 m/min for 6 weeks) underwent advanced cartilage degradation, compared with the sedentary animals [107].
In humans affected by OA, Jayabalan et al. observed that 45 min of continuous walking resulted in a cumulative increase in COMP concentration, a marker of cartilage turnover, whereas interval walking was associated with COMP concentrations comparable to baseline. This study shed light about the possibility that incorporating resting periods in walking regimens may impact the potentially deleterious effects of longer continuous walking bouts on the knee joint [108].
In conclusion, the above-mentioned controversies might be due to the performance of high-intensity exercises for a long time. Indeed, they are not commonly recommended by the recent International Guidelines for frail OA patients [64,65].

4. Nutrigenomic: Role of Nutraceuticals on Osteoarthritis

Nutraceuticals are considered as dietary supplements including a concentrated form of a presumed bioactive substance, originally derived from a food, aimed to improve health status particularly in “active aging” [109]. In this scenario, nutraceuticals play a pivotal role on osteoarthritis pathogenesis with specific targets in modulating OA pathways (see Table 1).
Long-chain polyunsaturated fatty acids (PUFAs) include alpha-linolenic (ALA), eicosapentaenoic (EPA) and docosahexaenoic (DHA) and are structural components of cell membranes. However, they also exert their action in a high number of intracellular signaling and metabolic pathways [26]. PUFAs and their metabolites act as second messengers when intercalated in the cell membrane, inhibiting the expression of MMP-13 through the inactivation of the p38MAPK and NF-κB p65 pathway, and promoting the activation of the PI3k/Akt signaling [58,74].
In the recent literature, the isolated administration of Chondroitin Sulfate has been shown to increase the transcription of the gene for Cartilage Oligomeric Matrix Protein (COMP) and decrease the expression of Toll-like receptors, thus inhibiting the Nfkb1 pathway in chondrocytes [112].
Glucosamine (GlcN) is an amino monosaccharide component of glycosaminoglycan (GAG) chains, usually in the form of sulfate or hydrochloride salts derived from the chitin of the exoskeleton of crustaceans and from mushrooms [26]. Glucosamine promotes the proliferation of human osteoblasts modulating the mTOR pathway and enhancing β-catenin nuclear translocation with consequent activation of cell proliferation via cyclin D1 expression [49,121,137].
Sulforaphane is an isothiocyanate derived from its glucosinolate precursor glucoraphanin, which is found in edible cruciferous vegetables, especially in broccoli. It can ameliorate ROS-induced oxidative stress and cartilage matrix degradation suppressing pro-inflammatory cytokines such as IL-6, TNF-α, and IL-17, inhibiting the JNK signaling and activating Keap1/Nrf2 pathway [133,134,135].
Berberine is an isoquinoline alkaloid, produced by many plants, as Coptis japonica Makino, Coptis, Berberis petiolaris and B. vulgaris [138]. It modulates the PI3K/Akt and NF-κB pathways regulating cytoskeletal reorganization and dedifferentiation in chondrocytes, improving the progression of OA [111].
Apigenin is a natural product belonging to the flavone class that is the aglycone of several naturally occurring glycosides. As an isolated compound from Cirsium japonicum var. maackii, it blocks MMP3, MMP13, and COX-2 expression through Hif-2α inhibition via the NF-κB and JNK signaling pathways [110]. Therefore, downstream of the NF-κB and JNK signal expression might block the translation of the pro-inflammatory factor HIF2a.
Eupatilin is an O-methylated flavonoid present in Artemisia asiatica (Asteraceae), which suppresses the expression of pro-inflammatory genes in chondrocytes through the reduction of JNK phosphorylation, NF-κB, and MAPK activation pathway [28].
Other nutraceuticals that could have a role in modulating the OA pathways seem to be the green tea catechins (GTCs), which could protect against cartilage loss and reduce the progression of OA through the modulation of NF-κB inhibitor expression and targeting the PI3K/AKT pathway. Another main component of the green tea is the epigallo-catechin-3 gallate that might inhibit NF-κB nuclear translocation by blocking IkB degradation in human chondrocytes stimulated with IL-1β [122,123,124].
Genistein, an isoflavone from soy (Glycine max), inhibits the NF-κB pathway and the consequent expression of catabolic factors, while stimulating the expression of Ho-1, associated with the activation of Nrf-2 in human chondrocytes [118,119,120].
Furthermore, Jaceosidin, an extract of Artemisia argyi, showed to reduce the damage to OA cartilage by blocking the degradation of IκB and inhibiting the nuclear translocation of NF-κB [117].
Quercitin is a hydroxyflavonoid widely contained in the flowers, leaves, and fruits of different plants. It improved OA via dose-dependent effects on the Toll-like Receptor-4 (TRL-4)/NF-κB pathway in vivo, inducing a cytosolic down expression of TLR-4 [129].
Moreover, Wogonin is an O-methylated flavone present in Scutellaria baicalensis with a positive effect on OA, which may be related to the inhibition of pro-inflammatory genes expression via the NF-κB pathway [136].
Curcumin, the main polyphenol component from the roots of turmeric (Curcuma longa), improves the inflammatory pattern through NF-κB signaling inhibition and Nrf2/ARE signaling activation. In addition, curcumin inhibits cell autophagy via ERK1/2 signaling pathway activation [52,113,114,115,116].
Olive oil polyphenols (OOPs), as oleocanthal (OC), oleuropein (OP), tyrosol (TY), and hydroxytyrosol (HT), could modulate inflammatory and degenerative OA molecular patterns. OOPs inhibit pro-inflammatory genes expression through the suppression of NF-κB and MAPK activation, while enhancing Nrf-2 signaling [125,126,127,128].
Lastly, resveratrol, a phenol extracted from the skin of grapes, wines, mulberries, and peanuts, belongs to the phytoalexins that protect plants from microbial infections. Resveratrol increases cell proliferation via stimulation of Wnt signaling dependent pathway and might exert self-limiting mechanism of inflammation through PI3K/Akt and Nrf2 axis activation and concomitant NF-κB inhibition [47,86,139,140].
Therefore, nutraceuticals and physical exercise are involved in molecular pathways OA with apoptotic, pro-, or anti-inflammatory targets (see Figure 1 for further details).

5. Conclusions

In this comprehensive review, we described the main molecular pathways underpinning OA pathogenesis and progression, highlighting the different mechanisms of action and potential targets for innovative therapeutic interventions. Moreover, we described common targets of physical activity and nutraceuticals suggesting a possible therapeutical synergism of these two interventions. The decrease in ROS production upstream of physical exercise can guarantee a beneficial approach regardless of the nutraceutical chosen, whereas the mutual connections on a cellular mediator would suggest the composition of the compounds in an ad hoc logic.
In conclusion, despite the limitations related to the low characterizations of some molecular pathways, the current review provides intriguing insights regarding molecular pathophysiological mechanisms, innovative therapeutical target pathways, and synergistic interventions that could improve the complex framework of OA management.

Author Contributions

Study conceptualization, A.d.S.; methodology, A.d.S.; writing—original draft preparation, A.d.S., N.M., and C.M.; writing—review and editing, M.I. and A.A.; visualization, C.C.; supervision, A.d.S. All authors have read and agreed to the published version of the manuscript.

Funding

Not applicable.

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. Cross, M.; Smith, E.; Hoy, D.; Nolte, S.; Ackerman, I.; Fransen, M.; Bridgett, L.; Williams, S.; Guillemin, F.; Hill, C.L.; et al. The global burden of hip and knee osteoarthritis: Estimates from the Global Burden of Disease 2010 study. Ann. Rheum. Dis. 2014, 73, 1323–1330. [Google Scholar] [CrossRef]
  2. Agel, J.; Akesson, K.; Amadio, P.C.; Anderson, M.; Badley, E.; Balint, G.; Bellamy, N.; Bigos, S.; Bishop, N.; Bivans, B.; et al. The Burden of Musculoskeletal Conditions at the Start of the New Millennium; WHO Technical Report Series; World Health Organization: Geneva, Switzerland, 2003. [Google Scholar]
  3. Martel-Pelletier, J.; Boileau, C.; Pelletier, J.P.; Roughley, P.J. Cartilage in normal and osteoarthritis conditions. Best Pract. Res. Clin. Rheumatol. 2008, 22, 351–384. [Google Scholar] [CrossRef]
  4. Wang, M.N.; Liu, L.; Zhao, L.P.; Yuan, F.; Fu, Y.B.; Xu, X.B.; Li, B. Research of inflammatory factors and signaling pathways in knee osteoarthritis. Zhongguo Gu Shang 2020, 33, 388–392. [Google Scholar] [PubMed]
  5. Hong, J.I.; Park, I.Y.; Kim, H.A. Understanding the molecular mechanisms underlying the pathogenesis of arthritis pain using animal models. Int. J. Mol. Sci. 2020, 21, 533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Al-Modawi, R.N.; Brinchmann, J.E.; Karlsen, T.A. Multi-pathway Protective Effects of MicroRNAs on Human Chondrocytes in an In Vitro Model of Osteoarthritis. Mol. Ther. Nucleic Acids 2019. [Google Scholar] [CrossRef] [Green Version]
  7. Zheng, L.; Zhang, Z.; Sheng, P.; Mobasheri, A. The role of metabolism in chondrocyte dysfunction and the progression of osteoarthritis. Ageing Res. Rev. 2021, 66, 101249. [Google Scholar] [CrossRef]
  8. Kraus, V.B.; Karsdal, M.A. Osteoarthritis: Current Molecular Biomarkers and the Way Forward. Calcif. Tissue Int. 2020, 1–10. [Google Scholar] [CrossRef]
  9. Xu, L.; Li, Y. A Molecular Cascade Underlying Articular Cartilage Degeneration. Curr. Drug Targets 2020. [Google Scholar] [CrossRef]
  10. Chow, Y.Y.; Chin, K.Y. The Role of Inflammation in the Pathogenesis of Osteoarthritis. Mediat. Inflamm. 2020, 2020, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. McDonough, C.M.; Jette, A.M. The contribution of osteoarthritis to functional limitations and disability. Clin. Geriatr. Med. 2010, 26, 387–399. [Google Scholar] [CrossRef] [Green Version]
  12. Iolascon, G.; Gimigliano, F.; Moretti, A.; de Sire, A.; Migliore, A.; Brandi, M.L.; Piscitelli, P. Early osteoarthritis: How to define, diagnose, and manage. A systematic review. Eur. Geriatr. Med. 2017. [Google Scholar] [CrossRef]
  13. McAlindon, T.E.; Bannuru, R.R.; Sullivan, M.C.; Arden, N.K.; Berenbaum, F.; Bierma-Zeinstra, S.M.; Hawker, G.A.; Henrotin, Y.; Hunter, D.J.; Kawaguchi, H.; et al. OARSI guidelines for the non-surgical management of knee osteoarthritis. Osteoarthr. Cartil. 2014. [Google Scholar] [CrossRef] [Green Version]
  14. Santilli, V.; Mangone, M.; Paoloni, M.; Agostini, F.; Alviti, F.; Bernetti, A. Comment on “early efficacy of intra-articular HYADD® 4 (Hymovis®) injections for symptomatic knee osteoarthritis”. Joints 2018, 5, 79. [Google Scholar] [CrossRef] [Green Version]
  15. Rabini, A.; De Sire, A.; Marzetti, E.; Gimigliano, R.; Ferriero, G.; Piazzini, D.B.; Iolascon, G.; Gimigliano, F. Effects of focal muscle vibration on physical functioning in patients with knee osteoarthritis: A randomized controlled trial. Eur. J. Phys. Rehabil. Med. 2015, 51, 513–520. [Google Scholar]
  16. De Sire, A.; Stagno, D.; Minetto, M.A.; Cisari, C.; Baricich, A.; Invernizzi, M. Long-term effects of intra-articular oxygen-ozone therapy versus hyaluronic acid in older people affected by knee osteoarthritis: A randomized single-blind extension study. J. Back Musculoskelet. Rehabil. 2020. [Google Scholar] [CrossRef]
  17. El-Hakeim, E.H.; Elawamy, A.; Kamel, E.Z.; Goma, S.H.; Gamal, R.M.; Ghandour, A.M.; Osman, A.M.; Morsy, K.M. Fluoroscopic guided radiofrequency of genicular nerves for pain alleviation in chronic knee osteoarthritis: A single-blind randomized controlled trial. Pain Physician 2018. [Google Scholar] [CrossRef]
  18. Roato, I.; Ferracini, R. Is the adipose-derived mesenchymal stem cell therapy effective for treatment of knee osteoarthritis? Ann. Transl. Med. 2019. [Google Scholar] [CrossRef]
  19. Southworth, T.M.; Naveen, N.B.; Tauro, T.M.; Leong, N.L.; Cole, B.J. The Use of Platelet-Rich Plasma in Symptomatic Knee Osteoarthritis. J. Knee Surg. 2019. [Google Scholar] [CrossRef]
  20. 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 Rheumatol. 2020. [Google Scholar] [CrossRef]
  21. Meiyappan, K.P.; Cote, M.P.; Bozic, K.J.; Halawi, M.J. Adherence to the American Academy of Orthopaedic Surgeons Clinical Practice Guidelines for Nonoperative Management of Knee Osteoarthritis. J. Arthroplast. 2020. [Google Scholar] [CrossRef]
  22. Mazor, M.; Best, T.M.; Cesaro, A.; Lespessailles, E.; Toumi, H. Osteoarthritis biomarker responses and cartilage adaptation to exercise: A review of animal and human models. Scand. J. Med. Sci. Sport. 2019, 29, 1072–1082. [Google Scholar] [CrossRef]
  23. de Sire, A.; de Sire, R.; Petito, V.; Masi, L.; Cisari, C.; Gasbarrini, A.; Scaldaferri, F.; Invernizzi, M. Gut–joint axis: The role of physical exercise on gut microbiota modulation in older people with osteoarthritis. Nutrients 2020, 12, 574. [Google Scholar] [CrossRef] [Green Version]
  24. Pérez-Lozano, M.L.; Cesaro, A.; Mazor, M.; Esteve, E.; Berteina-Raboin, S.; Best, T.M.; Lespessailles, E.; Toumi, H. Emerging natural-product-based treatments for the management of osteoarthritis. Antioxidants 2021, 10, 265. [Google Scholar] [CrossRef]
  25. Aghamohammadi, D.; Dolatkhah, N.; Bakhtiari, F.; Eslamian, F.; Hashemian, M. Nutraceutical supplements in management of pain and disability in osteoarthritis: A systematic review and meta-analysis of randomized clinical trials. Sci. Rep. 2020, 10. [Google Scholar] [CrossRef] [PubMed]
  26. D’Adamo, S.; Cetrullo, S.; Panichi, V.; Mariani, E.; Flamigni, F.; Borzì, R.M. Nutraceutical Activity in Osteoarthritis Biology: A Focus on the Nutrigenomic Role. Cells 2020, 9, 1232. [Google Scholar] [CrossRef] [PubMed]
  27. Deligiannidou, G.E.; Papadopoulos, R.E.; Kontogiorgis, C.; Detsi, A.; Bezirtzoglou, E.; Constantinides, T. Unraveling natural products’ role in osteoarthritis management—An overview. Antioxidants 2020, 9, 348. [Google Scholar] [CrossRef] [Green Version]
  28. Lee, H.; Zhao, X.; Son, Y.O.; Yang, S. Therapeutic single compounds for osteoarthritis treatment. Pharmaceuticals 2021, 14, 131. [Google Scholar] [CrossRef]
  29. Blazek, A.D.; Nam, J.; Gupta, R.; Pradhan, M.; Perera, P.; Weisleder, N.L.; Hewett, T.E.; Chaudhari, A.M.; Lee, B.S.; Leblebicioglu, B.; et al. Exercise-driven metabolic pathways in healthy cartilage. Osteoarthr. Cartil. 2016. [Google Scholar] [CrossRef] [Green Version]
  30. Hui, W.; Young, D.A.; Rowan, A.D.; Xu, X.; Cawston, T.E.; Proctor, C.J. Oxidative changes and signalling pathways are pivotal in initiating age-related changes in articular cartilage. Ann. Rheum. Dis. 2016. [Google Scholar] [CrossRef] [Green Version]
  31. Li, D.; Xie, G.; Wang, W. Reactive oxygen species: The 2-edged sword of osteoarthritis. Am. J. Med. Sci. 2012, 344, 486–490. [Google Scholar] [CrossRef]
  32. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [Green Version]
  33. Jones, D.P. Radical-free biology of oxidative stress. Am. J. Physiol. Cell Physiol. 2008, 295, C849–C868. [Google Scholar] [CrossRef] [Green Version]
  34. Blanco, F.J.; Rego, I.; Ruiz-Romero, C. The role of mitochondria in osteoarthritis. Nat. Rev. Rheumatol. 2011, 7, 161–169. [Google Scholar] [CrossRef]
  35. Mao, X.; Fu, P.; Wang, L.; Xiang, C. Mitochondria: Potential Targets for Osteoarthritis. Front. Med. 2020, 7, 7. [Google Scholar] [CrossRef]
  36. Lepetsos, P.; Papavassiliou, A.G. ROS/oxidative stress signaling in osteoarthritis. Biochim. Biophys. Acta Mol. Basis Dis. 2016, 1862, 576–591. [Google Scholar] [CrossRef]
  37. Rahmati, M.; Nalesso, G.; Mobasheri, A.; Mozafari, M. Aging and osteoarthritis: Central role of the extracellular matrix. Ageing Res. Rev. 2017, 40, 20–30. [Google Scholar] [CrossRef]
  38. Henrotin, Y.E.; Bruckner, P.; Pujol, J.P.L. The role of reactive oxygen species in homeostasis and degradation of cartilage. Osteoarthr. Cartil. 2003, 11, 747–755. [Google Scholar] [CrossRef] [Green Version]
  39. Regan, E.A.; Bowler, R.P.; Crapo, J.D. Joint fluid antioxidants are decreased in osteoarthritic joints compared to joints with macroscopically intact cartilage and subacute injury. Osteoarthr. Cartil. 2008. [Google Scholar] [CrossRef] [Green Version]
  40. Hwang, H.S.; Kim, H.A. Chondrocyte apoptosis in the pathogenesis of osteoarthritis. Int. J. Mol. Sci. 2015, 16, 26035–26054. [Google Scholar] [CrossRef]
  41. Zahan, O.M.; Serban, O.; Gherman, C.; Fodor, D. The evaluation of oxidative stress in osteoarthritis. Med. Pharm. Rep. 2020, 93, 12–22. [Google Scholar] [CrossRef]
  42. Setti, T.; Arab, M.G.L.; Santos, G.S.; Alkass, N.; Andrade, M.A.P.; Lana, J.F.S.D. The protective role of glutathione in osteoarthritis. J. Clin. Orthop. Trauma 2021, 15, 145–151. [Google Scholar] [CrossRef]
  43. Hu, S.; Zhang, C.; Ni, L.; Huang, C.; Chen, D.; Shi, K.; Jin, H.; Zhang, K.; Li, Y.; Xie, L.; et al. Stabilization of HIF-1α alleviates osteoarthritis via enhancing mitophagy. Cell Death Dis. 2020. [Google Scholar] [CrossRef]
  44. Malemud, C.J. The PI3K/Akt/PTEN/mTOR pathway: A fruitful target for inducing cell death in rheumatoid arthritis? Future Med. Chem. 2015, 7, 1137–1147. [Google Scholar] [CrossRef]
  45. Li, Z.; Wang, J.; Deng, X.; Huang, D.; Shao, Z.; Ma, K. Compression stress induces nucleus pulposus cell autophagy by inhibition of the PI3K/AKT/mTOR pathway and activation of the JNK pathway. Connect. Tissue Res. 2020. [Google Scholar] [CrossRef] [PubMed]
  46. Rao, Z.; Wang, S.; Wang, J. Peroxiredoxin 4 inhibits IL-1β-induced chondrocyte apoptosis via PI3K/AKT signaling. Biomed. Pharmacother. 2017. [Google Scholar] [CrossRef]
  47. Ashrafizadeh, M.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Resveratrol targeting the Wnt signaling pathway: A focus on therapeutic activities. J. Cell. Physiol. 2020, 235, 4135–4145. [Google Scholar] [CrossRef] [PubMed]
  48. Bouaziz, W.; Sigaux, J.; Modrowski, D.; Devignes, C.S.; Funck-Brentano, T.; Richette, P.; Ea, H.K.; Provot, S.; Cohen-Solal, M.; Haÿ, E. Interaction of HIF1α and β-catenin inhibits matrix metalloproteinase 13 expression and prevents cartilage damage in mice. Proc. Natl. Acad. Sci. USA 2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Ma, Y.; Zheng, W.; Chen, H.; Shao, X.; Lin, P.; Liu, X.; Li, X.; Ye, H. Glucosamine promotes chondrocyte proliferation via the Wnt/β-catenin signaling pathway. Int. J. Mol. Med. 2018. [Google Scholar] [CrossRef] [PubMed]
  50. Khan, N.M.; Haseeb, A.; Ansari, M.Y.; Devarapalli, P.; Haynie, S.; Haqqi, T.M. Wogonin, a plant derived small molecule, exerts potent anti-inflammatory and chondroprotective effects through the activation of ROS/ERK/Nrf2 signaling pathways in human Osteoarthritis chondrocytes. Free Radic. Biol. Med. 2017. [Google Scholar] [CrossRef] [Green Version]
  51. Zhao, Y.P.; Liu, B.; Tian, Q.Y.; Wei, J.L.; Richbourgh, B.; Liu, C.J. Progranulin protects against osteoarthritis through interacting with TNF-α and β-Catenin signalling. Ann. Rheum. Dis. 2015. [Google Scholar] [CrossRef] [Green Version]
  52. Li, X.; Feng, K.; Li, J.; Yu, D.; Fan, Q.; Tang, T.; Yao, X.; Wang, X. Curcumin inhibits apoptosis of chondrocytes through activation ERK1/2 signaling pathways induced autophagy. Nutrients 2017, 9, 414. [Google Scholar] [CrossRef]
  53. Imagawa, K.; de Andrés, M.C.; Hashimoto, K.; Pitt, D.; Itoi, E.; Goldring, M.B.; Roach, H.I.; Oreffo, R.O.C. The epigenetic effect of glucosamine and a nuclear factor-kappa B (NF-κB) inhibitor on primary human chondrocytes—Implications for osteoarthritis. Biochem. Biophys. Res. Commun. 2011. [Google Scholar] [CrossRef] [Green Version]
  54. Vincent, T.L. Mechanoflammation in osteoarthritis pathogenesis. Semin. Arthritis Rheum. 2019. [Google Scholar] [CrossRef]
  55. Qin, Y.; Chen, Y.; Wang, W.; Wang, Z.; Tang, G.; Zhang, P.; He, Z.; Liu, Y.; Dai, S.M.; Shen, Q. HMGB1-LPS complex promotes transformation of osteoarthritis synovial fibroblasts to a rheumatoid arthritis synovial fibroblast-like phenotype. Cell Death Dis. 2014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Millerand, M.; Berenbaum, F.; Jacques, C. Danger signals and inflammaging in osteoarthritis. Clin. Exp. Rheumatol. 2019, 37, 48–56. [Google Scholar] [PubMed]
  57. Sun, J.; Nan, G. The Mitogen-Activated Protein Kinase (MAPK) Signaling Pathway as a Discovery Target in Stroke. J. Mol. Neurosci. 2016, 59, 90–98. [Google Scholar] [CrossRef]
  58. Wang, Z.; Guo, A.; Ma, L.; Yu, H.; Zhang, L.; Meng, H.; Cui, Y.; Yu, F.; Yang, B. Docosahexenoic acid treatment ameliorates cartilage degeneration via a p38 MAPK-dependent mechanism. Int. J. Mol. Med. 2016. [Google Scholar] [CrossRef] [Green Version]
  59. Marchev, A.S.; Dimitrova, P.A.; Burns, A.J.; Kostov, R.V.; Dinkova-Kostova, A.T.; Georgiev, M.I. Oxidative stress and chronic inflammation in osteoarthritis: Can NRF2 counteract these partners in crime? Ann. N. Y. Acad. Sci. 2017, 1404, 114–135. [Google Scholar] [CrossRef]
  60. Sun, Z.; Zhang, S.; Chan, J.Y.; Zhang, D.D. Keap1 Controls Postinduction Repression of the Nrf2-Mediated Antioxidant Response by Escorting Nuclear Export of Nrf2. Mol. Cell. Biol. 2007. [Google Scholar] [CrossRef] [Green Version]
  61. Tonelli, C.; Chio, I.I.C.; Tuveson, D.A. Transcriptional Regulation by Nrf2. Antioxid. Redox Signal. 2018, 29, 1727–1745. [Google Scholar] [CrossRef] [Green Version]
  62. He, F.; Ru, X.; Wen, T. NRF2, a transcription factor for stress response and beyond. Int. J. Mol. Sci. 2020, 21, 4777. [Google Scholar] [CrossRef]
  63. Jevsevar, D.S.; Brown, G.A.; Jones, D.L.; Matzkin, E.G.; Manner, P.A.; Mooar, P.; Schousboe, J.T.; Stovitz, S.; Sanders, J.O.; Bozic, K.J.; et al. The American Academy of Orthopaedic Surgeons evidence-based guideline on: Treatment of osteoarthritis of the knee, 2nd edition. J. Bone Jt. Surg. Am. 2013. [Google Scholar] [CrossRef]
  64. Garber, C.E.; Blissmer, B.; Deschenes, M.R.; Franklin, B.A.; Lamonte, M.J.; Lee, I.M.; Nieman, D.C.; Swain, D.P. Quantity and quality of exercise for developing and maintaining cardiorespiratory, musculoskeletal, and neuromotor fitness in apparently healthy adults: Guidance for prescribing exercise. Med. Sci. Sports Exerc. 2011. [Google Scholar] [CrossRef] [PubMed]
  65. Rausch Osthoff, A.K.; Niedermann, K.; Braun, J.; Adams, J.; Brodin, N.; Dagfinrud, H.; Duruoz, T.; Esbensen, B.A.; Günther, K.P.; Hurkmans, E.; et al. 2018 EULAR recommendations for physical activity in people with inflammatory arthritis and osteoarthritis. Ann. Rheum. Dis. 2018, 77, 1251–1260. [Google Scholar] [CrossRef] [Green Version]
  66. Fransen, M. When is physiotherapy appropriate? Best Pract. Res. Clin. Rheumatol. 2004. [Google Scholar] [CrossRef]
  67. Rahmann, A. Exercise for people with hip or knee osteoarthritis: A comparison of land-based and aquatic interventions. Open Access J. Sport. Med. 2010. [Google Scholar] [CrossRef] [Green Version]
  68. Raposo, F.; Ramos, M.; Lúcia Cruz, A. Effects of exercise on knee osteoarthritis: A systematic review. Musculoskelet. Care 2021. [Google Scholar] [CrossRef]
  69. Iijima, H.; Aoyama, T.; Ito, A.; Tajino, J.; Yamaguchi, S.; Nagai, M.; Kiyan, W.; Zhang, X.; Kuroki, H. Exercise intervention increases expression of bone morphogenetic proteins and prevents the progression of cartilage-subchondral bone lesions in a post-traumatic rat knee model. Osteoarthr. Cartil. 2016. [Google Scholar] [CrossRef] [Green Version]
  70. Assis, L.; Milares, L.P.; Almeida, T.; Tim, C.; Magri, A.; Fernandes, K.R.; Medalha, C.; Muniz Renno, A.C. Aerobic exercise training and low-level laser therapy modulate inflammatory response and degenerative process in an experimental model of knee osteoarthritis in rats. Osteoarthr. Cartil. 2016, 24, 169–177. [Google Scholar] [CrossRef] [Green Version]
  71. Yang, Y.; Wang, Y.; Kong, Y.; Zhang, X.; Bai, L. The effects of different frequency treadmill exercise on lipoxin A4 and articular cartilage degeneration in an experimental model of monosodium iodoacetate-induced osteoarthritis in rats. PLoS ONE 2017, 12. [Google Scholar] [CrossRef]
  72. Zhang, H.; Ji, L.; Yang, Y.; Wei, Y.; Zhang, X.; Gang, Y.; Lu, J.; Bai, L. The Therapeutic Effects of Treadmill Exercise on Osteoarthritis in Rats by Inhibiting the HDAC3/NF-KappaB Pathway in vivo and in vitro. Front. Physiol. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  73. Castrogiovanni, P.; Di Rosa, M.; Ravalli, S.; Castorina, A.; Guglielmino, C.; Imbesi, R.; Vecchio, M.; Drago, F.; Szychlinska, M.A.; Musumeci, G. Moderate physical activity as a prevention method for knee osteoarthritis and the role of synoviocytes as biological key. Int. J. Mol. Sci. 2019, 20, 511. [Google Scholar] [CrossRef] [Green Version]
  74. Lu, J.; Feng, X.; Zhang, H.; Wei, Y.; Yang, Y.; Tian, Y.; Bai, L. Maresin-1 suppresses IL-1β-induced MMP-13 secretion by activating the PI3K/AKT pathway and inhibiting the NF-κB pathway in synovioblasts of an osteoarthritis rat model with treadmill exercise. Connect. Tissue Res. 2020. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, X.; Yang, Y.; Li, X.; Zhang, H.; Gang, Y.; Bai, L. Alterations of autophagy in knee cartilage by treatment with treadmill exercise in a rat osteoarthritis model. Int. J. Mol. Med. 2019, 43, 336–344. [Google Scholar] [CrossRef] [PubMed]
  76. Tian, Y.; Gou, J.; Zhang, H.; Lu, J.; Jin, Z.; Jia, S.; Bai, L. The anti-inflammatory effects of 15-HETE on osteoarthritis during treadmill exercise. Life Sci. 2021. [Google Scholar] [CrossRef]
  77. Galois, L.; Etienne, S.; Grossin, L.; Watrin-Pinzano, A.; Cournil-Henrionnet, C.; Loeuille, D.; Netter, P.; Mainard, D.; Gillet, P. Dose-response relationship for exercise on severity of experimental osteoarthritis in rats: A pilot study. Osteoarthr. Cartil. 2004. [Google Scholar] [CrossRef] [Green Version]
  78. Nam, J.; Perera, P.; Liu, J.; Wu, L.C.; Rath, B.; Butterfield, T.A.; Agarwal, S. Transcriptome-wide gene regulation by gentle treadmill walking during the progression of monoiodoacetate-induced arthritis. Arthritis Rheum. 2011. [Google Scholar] [CrossRef] [Green Version]
  79. Bricca, A.; Juhl, C.B.; Steultjens, M.; Wirth, W.; Roos, E.M. Impact of exercise on articular cartilage in people at risk of, or with established, knee osteoarthritis: A systematic review of randomised controlled trials. Br. J. Sports Med. 2019. [Google Scholar] [CrossRef] [Green Version]
  80. Roos, E.M.; Dahlberg, L. Positive effects of moderate exercise on glycosaminoglycan content in knee cartilage: A four-month, randomized, controlled trial in patients at risk of osteoarthritis. Arthritis Rheum. 2005. [Google Scholar] [CrossRef] [Green Version]
  81. Hawezi, Z.K.; Lammentausta, E.; Svensson, J.; Roos, E.M.; Dahlberg, L.E.; Tiderius, C.J. Regional dGEMRIC analysis in patients at risk of osteoar thritis provides additional information about activity related changes in car tilage structure. Acta Radiol. 2016. [Google Scholar] [CrossRef]
  82. Munukka, M.; Waller, B.; Häkkinen, A.; Nieminen, M.T.; Lammentausta, E.; Kujala, U.M.; Paloneva, J.; Kautiainen, H.; Kiviranta, I.; Heinonen, A. Physical Activity Is Related with Cartilage Quality in Women with Knee Osteoarthritis. Med. Sci. Sports Exerc. 2017. [Google Scholar] [CrossRef] [PubMed]
  83. Helmark, I.C.; Mikkelsen, U.R.; Børglum, J.; Rothe, A.; Petersen, M.C.H.; Andersen, O.; Langberg, H.; Kjaer, M. Exercise increases interleukin-10 levels both intraarticularly and peri-synovially in patients with knee osteoarthritis: A randomized controlled trial. Arthritis Res. Ther. 2010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Hunt, M.A.; Pollock, C.L.; Kraus, V.B.; Saxne, T.; Peters, S.; Huebner, J.L.; Sayre, E.C.; Cibere, J. Relationships amongst osteoarthritis biomarkers, dynamic knee joint load, and exercise: Results from a randomized controlled pilot study. BMC Musculoskelet. Disord. 2013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Messier, S.P.; Mihalko, S.L.; Legault, C.; Miller, G.D.; Nicklas, B.J.; DeVita, P.; Beavers, D.P.; Hunter, D.J.; Lyles, M.F.; Eckstein, F.; et al. Effects of intensive diet and exercise on knee joint loads, inflammation, and clinical outcomes among overweight and obese adults with knee osteoarthritis: The IDEA randomized clinical trial. JAMA J. Am. Med. Assoc. 2013. [Google Scholar] [CrossRef]
  86. Simioni, C.; Zauli, G.; Martelli, A.M.; Vitale, M.; Sacchetti, G.; Gonelli, A.; Neri, L.M. Oxidative stress: Role of physical exercise and antioxidant nutraceuticals in adulthood and aging. Oncotarget 2018, 9, 17181–17198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Dalle Carbonare, L.; Mottes, M.; Cheri, S.; Deiana, M.; Zamboni, F.; Gabbiani, D.; Schena, F.; Salvagno, G.L.; Lippi, G.; Valenti, M.T. Increased Gene Expression of RUNX2 and SOX9 in Mesenchymal Circulating Progenitors Is Associated with Autophagy during Physical Activity. Oxid. Med. Cell. Longev. 2019. [Google Scholar] [CrossRef]
  88. Hunter, C.J.; Imler, S.M.; Malaviya, P.; Nerem, R.M.; Levenston, M.E. Mechanical compression alters gene expression and extracellular matrix synthesis by chondrocytes cultured in collagen I gels. Biomaterials 2002. [Google Scholar] [CrossRef]
  89. Khan, N.M.; Ahmad, I.; Haqqi, T.M. Nrf2/ARE pathway attenuates oxidative and apoptotic response in human osteoarthritis chondrocytes by activating ERK1/2/ELK1-P70S6K-P90RSK signaling axis. Free Radic. Biol. Med. 2018, 116, 159–171. [Google Scholar] [CrossRef]
  90. Vargas-Mendoza, N.; Morales-González, Á.; Madrigal-Santillán, E.O.; Madrigal-Bujaidar, E.; Álvarez-González, I.; García-Melo, L.F.; Anguiano-Robledo, L.; Fregoso-Aguilar, T.; Morales-Gonzalez, J.A. Antioxidant and adaptative response mediated by Nrf2 during physical exercise. Antioxidants 2019, 8, 196. [Google Scholar] [CrossRef] [Green Version]
  91. Cifuentes, D.J.; Rocha, L.G.; Silva, L.A.; Brito, A.C.; Rueff-Barroso, C.R.; Porto, L.C.; Pinho, R.A. Decrease in oxidative stress and histological changes induced by physical exercise calibrated in rats with osteoarthritis induced by monosodium iodoacetate. Osteoarthr. Cartil. 2010. [Google Scholar] [CrossRef] [Green Version]
  92. Germanou, E.I.; Chatzinikolaou, A.; Malliou, P.; Beneka, A.; Jamurtas, A.Z.; Bikos, C.; Tsoukas, D.; Theodorou, A.; Katrabasas, I.; Margonis, K.; et al. Oxidative stress and inflammatory responses following an acute bout of isokinetic exercise in obese women with knee osteoarthritis. Knee 2013. [Google Scholar] [CrossRef] [PubMed]
  93. Freitag, J.; Bates, D.; Boyd, R.; Shah, K.; Barnard, A.; Huguenin, L.; Tenen, A. Mesenchymal stem cell therapy in the treatment of osteoarthritis: Reparative pathways, safety and efficacy—A review. BMC Musculoskelet. Disord. 2016, 17, 1–13. [Google Scholar] [CrossRef] [Green Version]
  94. Song, J.S.; Hong, K.T.; Kim, N.M.; Jung, J.Y.; Park, H.S.; Chun, Y.S.; Kim, S.J. Cartilage regeneration in osteoarthritic knees treated with distal femoral osteotomy and intra-lesional implantation of allogenic human umbilical cord blood-derived mesenchymal stem cells: A report of two cases. Knee 2019. [Google Scholar] [CrossRef]
  95. Minas, T.; Ogura, T.; Bryant, T. Autologous chondrocyte implantation. JBJS Essent. Surg. Tech. 2016. [Google Scholar] [CrossRef]
  96. Schmidt, A.; Bierwirth, S.; Weber, S.; Platen, P.; Schinköthe, T.; Bloch, W. Short intensive exercise increases the migratory activity of mesenchymal stem cells. Br. J. Sports Med. 2009. [Google Scholar] [CrossRef]
  97. Valenti, M.T.; Deiana, M.; Cheri, S.; Dotta, M.; Zamboni, F.; Gabbiani, D.; Schena, F.; Dalle Carbonare, L.; Mottes, M. Physical Exercise Modulates miR-21-5p, miR-129-5p, miR-378-5p, and miR-188-5p Expression in Progenitor Cells Promoting Osteogenesis. Cells 2019, 8, 742. [Google Scholar] [CrossRef] [Green Version]
  98. Smith, J.K. Exercise as an adjuvant to cartilage regeneration therapy. Int. J. Mol. Sci. 2020, 21, 9471. [Google Scholar] [CrossRef] [PubMed]
  99. Emmons, R.; Niemiro, G.M.; Owolabi, O.; De Lisio, M. Acute exercise mobilizes hematopoietic stem and progenitor cells and alters the mesenchymal stromal cell secretome. J. Appl. Physiol. 2016. [Google Scholar] [CrossRef]
  100. Bourzac, C.; Bensidhoum, M.; Pallu, S.; Portier, H. Use of adult mesenchymal stromal cells in tissue repair: Impact of physical exercise. Am. J. Physiol. Cell Physiol. 2019, 317, C642–C654. [Google Scholar] [CrossRef] [PubMed]
  101. Ocarino, N.M.; Boeloni, J.N.; Goes, A.M.; Silva, J.F.; Marubayashi, U.; Serakides, R. Osteogenic differentiation of mesenchymal stem cells from osteopenic rats subjected to physical activity with and without nitric oxide synthase inhibition. Nitric Oxide Biol. Chem. 2008. [Google Scholar] [CrossRef] [PubMed]
  102. Hell, R.C.R.; Ocarino, N.M.; Boeloni, J.N.; Silva, J.F.; Goes, A.M.; Santos, R.L.; Serakides, R. Physical activity improves age-related decline in the osteogenic potential of rats’ bone marrow-derived mesenchymal stem cells. Acta Physiol. 2012, 205, 292–301. [Google Scholar] [CrossRef]
  103. Andersson, M.L.E.; Thorstensson, C.A.; Roos, E.M.; Petersson, I.F.; Heinegård, D.; Saxne, T. Serum levels of Cartilage Oligomeric Matrix Protein (COMP) increase temporarily after physical exercise in patients with knee osteoarthritis. BMC Musculoskelet. Disord. 2006, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Siebelt, M.; Groen, H.C.; Koelewijn, S.J.; de Blois, E.; Sandker, M.; Waarsing, J.H.; Müller, C.; van Osch, G.J.V.M.; de Jong, M.; Weinans, H. Increased physical activity severely induces osteoarthritic changes in knee joints with papain induced sulfate-glycosaminoglycan depleted cartilage. Arthritis Res. Ther. 2014, 16. [Google Scholar] [CrossRef] [Green Version]
  105. Liu, S.S.; Zhou, P.; Zhang, Y. Abnormal expression of key genes and proteins in the canonical Wnt/β-catenin pathway of articular cartilage in a rat model of exercise-induced osteoarthritis. Mol. Med. Rep. 2016, 13, 1999–2006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Rojas-Ortega, M.; Cruz, R.; Vega-López, M.A.; Cabrera-González, M.; Hernández-Hernández, J.M.; Lavalle-Montalvo, C.; Kouri, J.B. Exercise modulates the expression of IL-1β and IL-10 in the articular cartilage of normal and osteoarthritis-induced rats. Pathol. Res. Pract. 2015. [Google Scholar] [CrossRef]
  107. Coyle, C.H.; Henry, S.E.; Haleem, A.M.; O’Malley, M.J.; Chu, C.R. Serum CTXii Correlates with Articular Cartilage Degeneration After Anterior Cruciate Ligament Transection or Arthrotomy Followed by Standardized Exercise. Sports Health 2012. [Google Scholar] [CrossRef] [Green Version]
  108. Jayabalan, P.; Gustafson, J.; Sowa, G.A.; Piva, S.R.; Farrokhi, S. A Stimulus-Response Framework to Investigate the Influence of Continuous Versus Interval Walking Exercise on Select Serum Biomarkers in Knee Osteoarthritis. Am. J. Phys. Med. Rehabil. 2019. [Google Scholar] [CrossRef] [PubMed]
  109. Iolascon, G.; Gimigliano, R.; Bianco, M.; de Sire, A.; Moretti, A.; Giusti, A.; Malavolta, N.; Migliaccio, S.; Migliore, A.; Napoli, N.; et al. Are dietary supplements and nutraceuticals effective for musculoskeletal health and cognitive function? A scoping review. J. Nutr. Heal. Aging 2017. [Google Scholar] [CrossRef]
  110. Cho, C.; Kang, L.J.; Jang, D.; Jeon, J.; Lee, H.; Choi, S.; Han, S.J.; Oh, E.; Nam, J.; Kim, C.S.; et al. Cirsium japonicum var. maackii and apigenin block Hif-2α-induced osteoarthritic cartilage destruction. J. Cell. Mol. Med. 2019. [Google Scholar] [CrossRef]
  111. Wong, S.K.; Chin, K.Y.; Ima-Nirwana, S. Berberine and musculoskeletal disorders: The therapeutic potential and underlying molecular mechanisms. Phytomedicine 2020, 73, 152892. [Google Scholar] [CrossRef]
  112. Korotkyi, O.; Huet, A.; Dvorshchenko, K.; Kobyliak, N.; Falalyeyeva, T.; Ostapchenko, L. Probiotic Composition and Chondroitin Sulfate Regulate TLR-2/4-Mediated NF-κB Inflammatory Pathway and Cartilage Metabolism in Experimental Osteoarthritis. Probiotics Antimicrob. Proteins 2021. [Google Scholar] [CrossRef]
  113. Zhang, Y.; Zeng, Y. Curcumin reduces inflammation in knee osteoarthritis rats through blocking TLR4 /MyD88/NF-κB signal pathway. Drug Dev. Res. 2019. [Google Scholar] [CrossRef]
  114. Jiang, C.; Luo, P.; Li, X.; Liu, P.; Li, Y.; Xu, J. Nrf2/ARE is a key pathway for curcumin-mediated protection of TMJ chondrocytes from oxidative stress and inflammation. Cell Stress Chaperones 2020. [Google Scholar] [CrossRef] [PubMed]
  115. Panaro, M.A.; Corrado, A.; Benameur, T.; Paolo, C.F.; Cici, D.; Porro, C. The emerging role of curcumin in the modulation of TLR-4 signaling pathway: Focus on neuroprotective and anti-rheumatic properties. Int. J. Mol. Sci. 2020, 21, 2299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Qiu, B.; Xu, X.; Yi, P.; Hao, Y. Curcumin reinforces MSC-derived exosomes in attenuating osteoarthritis via modulating the miR-124/NF-κB and miR-143/ROCK1/TLR9 signalling pathways. J. Cell. Mol. Med. 2020. [Google Scholar] [CrossRef]
  117. Lee, H.; Jang, D.; Jeon, J.; Cho, C.; Choi, S.; Han, S.J.; Oh, E.; Nam, J.; Park, C.H.; Shin, Y.S.; et al. Seomae mugwort and jaceosidin attenuate osteoarthritic cartilage damage by blocking IκB degradation in mice. J. Cell. Mol. Med. 2020, 24, 8126–8137. [Google Scholar] [CrossRef]
  118. Liu, F.C.; Wang, C.C.; Lu, J.W.; Lee, C.H.; Chen, S.C.; Ho, Y.J.; Peng, Y.J. Chondroprotective effects of genistein against osteoarthritis induced joint inflammation. Nutrients 2019, 11, 1180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Yuan, J.; Ding, W.; Wu, N.; Jiang, S.; Li, W. Protective Effect of Genistein on Condylar Cartilage through Downregulating NF- B Expression in Experimentally Created Osteoarthritis Rats. Biomed Res. Int. 2019. [Google Scholar] [CrossRef] [Green Version]
  120. Zou, Y.; Liu, Q.; Guo, P.; Huang, Y.; Ye, Z.; Hu, J. Anti-chondrocyte apoptosis effect of genistein in treating inflammation-induced osteoarthritis. Mol. Med. Rep. 2020, 22, 2032–2042. [Google Scholar] [CrossRef]
  121. Lv, C.; Wang, L.; Zhu, X.; Lin, W.; Chen, X.; Huang, Z.; Huang, L.; Yang, S. Glucosamine promotes osteoblast proliferation by modulating autophagy via the mammalian target of rapamycin pathway. Biomed. Pharmacother. 2018. [Google Scholar] [CrossRef]
  122. Bai, H.; Zhang, Z.; Li, Y.; Song, X.; Ma, T.; Liu, C.; Liu, L.; Yuan, R.; Wang, X.; Gao, L. L-theanine reduced the development of knee osteoarthritis in rats via its anti-inflammation and anti-matrix degradation actions: In vivo and in vitro study. Nutrients 2020, 12, 1988. [Google Scholar] [CrossRef] [PubMed]
  123. Luk, H.Y.; Appell, C.; Chyu, M.C.; Chen, C.H.; Wang, C.Y.; Yang, R.S.; Shen, C.L. Impacts of green tea on joint and skeletal muscle health: Prospects of translational nutrition. Antioxidants 2020, 9, 1050. [Google Scholar] [CrossRef] [PubMed]
  124. Chang, Y.C.; Liu, H.W.; Chan, Y.C.; Hu, S.H.; Liu, M.Y.; Chang, S.J. The green tea polyphenol epigallocatechin-3-gallate attenuates age-associated muscle loss via regulation of miR-486-5p and myostatin. Arch. Biochem. Biophys. 2020. [Google Scholar] [CrossRef]
  125. Feng, Z.; Li, X.; Lin, J.; Zheng, W.; Hu, Z.; Xuan, J.; Ni, W.; Pan, X. Oleuropein inhibits the IL-1β-induced expression of inflammatory mediators by suppressing the activation of NF-κB and MAPKs in human osteoarthritis chondrocytes. Food Funct. 2017. [Google Scholar] [CrossRef] [PubMed]
  126. Serreli, G.; Deiana, M. Extra Virgin Olive Oil Polyphenols: Modulation of Cellular Pathways Related to Oxidant Species and Inflammation in Aging. Cells 2020, 9, 478. [Google Scholar] [CrossRef] [Green Version]
  127. Varela-Eirín, M.; Carpintero-Fernández, P.; Sánchez-Temprano, A.; Varela-Vázquez, A.; Paíno, C.L.; Casado-Díaz, A.; Continente, A.C.; Mato, V.; Fonseca, E.; Kandouz, M.; et al. Senolytic activity of small molecular polyphenols from olive restores chondrocyte redifferentiation and promotes a pro-regenerative environment in osteoarthritis. Aging 2020. [Google Scholar] [CrossRef]
  128. Chen, Y.L.; Yan, D.Y.; Wu, C.Y.; Xuan, J.W.; Jin, C.Q.; Hu, X.L.; Bao, G.D.; Bian, Y.J.; Hu, Z.C.; Shen, Z.H.; et al. Maslinic acid prevents IL-1β-induced inflammatory response in osteoarthritis via PI3K/AKT/NF-κB pathways. J. Cell. Physiol. 2021. [Google Scholar] [CrossRef]
  129. Zhang, J.; Yin, J.; Zhao, D.; Wang, C.; Zhang, Y.; Wang, Y.; Li, T. Therapeutic effect and mechanism of action of quercetin in a rat model of osteoarthritis. J. Int. Med. Res. 2019. [Google Scholar] [CrossRef]
  130. Farkhondeh, T.; Folgado, S.L.; Pourbagher-Shahri, A.M.; Ashrafizadeh, M.; Samarghandian, S. The therapeutic effect of resveratrol: Focusing on the Nrf2 signaling pathway. Biomed. Pharmacother. 2020, 127, 110234. [Google Scholar] [CrossRef]
  131. Xu, X.; Liu, X.; Yang, Y.; He, J.; Jiang, M.; Huang, Y.; Liu, X.; Liu, L.; Gu, H. Resveratrol exerts anti-osteoarthritic effect by inhibiting TLR4/NF-κB signaling pathway via the TLR4/Akt/FoxO1 axis in IL-1β-stimulated SW1353 cells. Drug Des. Devel. Ther. 2020. [Google Scholar] [CrossRef] [PubMed]
  132. Yi, H.; Zhang, W.; Cui, Z.M.; Cui, S.Y.; Fan, J.B.; Zhu, X.H.; Liu, W. Resveratrol alleviates the interleukin-1β-induced chondrocytes injury through the NF-κB signaling pathway. J. Orthop. Surg. Res. 2020. [Google Scholar] [CrossRef]
  133. Yang, J.; Song, X.; Feng, Y.; Liu, N.; Fu, Z.; Wu, J.; Li, T.; Chen, H.; Chen, J.; Chen, C.; et al. Natural ingredients-derived antioxidants attenuate H2O2-induced oxidative stress and have chondroprotective effects on human osteoarthritic chondrocytes via Keap1/Nrf2 pathway. Free Radic. Biol. Med. 2020. [Google Scholar] [CrossRef] [PubMed]
  134. Moon, S.J.; Jhun, J.; Ryu, J.; Kwon, J.Y.; Kim, S.Y.; Jung, K.A.; Cho, M.L.; Min, J.K. The anti-arthritis effect of sulforaphane, an activator of Nrf2, is associated with inhibition of both B cell differentiation and the production of inflammatory cytokines. PLoS ONE 2021. [Google Scholar] [CrossRef] [PubMed]
  135. Luo, T.; Fu, X.; Liu, Y.; Ji, Y.; Shang, Z. Sulforaphane Inhibits Osteoclastogenesis via Suppression of the Autophagic Pathway. Molecules 2021, 26, 347. [Google Scholar] [CrossRef] [PubMed]
  136. Khan, N.M.; Haseeb, A.; Ansari, M.Y.; Haqqi, T.M. A wogonin-rich-fraction of Scutellaria baicalensis root extract exerts chondroprotective effects by suppressing IL-1β-induced activation of AP-1 in human OA chondrocytes. Sci. Rep. 2017. [Google Scholar] [CrossRef] [Green Version]
  137. Ammendolia, A.; Marotta, N.; Marinaro, C.; Demeco, A.; Mondardini, P.; Costantino, C. High Power Laser Therapy and Glucosamine sulfate in the treatment of knee osteoarthritis: A single blinded randomized controlled trial. Acta Biomed. l’Ateneo Parm. 2021. [Google Scholar] [CrossRef]
  138. Zhang, C.; Sheng, J.; Li, G.; Zhao, L.; Wang, Y.; Yang, W.; Yao, X.; Sun, L.; Zhang, Z.; Cui, R. Effects of berberine and its derivatives on cancer: A systems pharmacology review. Front. Pharmacol. 2020, 10, 1461. [Google Scholar] [CrossRef] [Green Version]
  139. Hou, C.Y.; Tain, Y.L.; Yu, H.R.; Huang, L.T. The effects of resveratrol in the treatment of metabolic syndrome. Int. J. Mol. Sci. 2019, 20, 535. [Google Scholar] [CrossRef] [Green Version]
  140. Hasan, M.M.; Bae, H. An overview of stress-induced resveratrol synthesis in grapes: Perspectives for resveratrol-enriched grape products. Molecules 2017, 22, 294. [Google Scholar] [CrossRef]
Figure 1. Involvement of nutraceuticals and exercise in molecular pathways for apoptotic, pro-, or anti-inflammatory signaling.
Figure 1. Involvement of nutraceuticals and exercise in molecular pathways for apoptotic, pro-, or anti-inflammatory signaling.
Ijms 22 05722 g001
Table 1. Characteristics of the studies included with the nutrigenomic targets.
Table 1. Characteristics of the studies included with the nutrigenomic targets.
Nutraceutical OA Pathways InvolvedModulation ActionMain FindingsJournalAuthorsYear
Apigenin┤ HIF 2α↓ ADAMTS-4
↓ COX-2
↓ MMP
Apigenin blocks osteoarthritis development as Hif-2α inhibitor.Journal of Cellular and Molecular MedicineCho et al. [110]2019
Berberine→ PI3K/AKT
┤ NF-κB
↓ ADAMTS-4
↓ COX-2
↓ MMP
Berberine activates PI3K/Akt and NF-κB pathways.PhytomedicineWong et al. [111]2019
Chondroitin Sulfate┤ NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
Separate administration of chondroitin sulfate raised expression of Comp and reduced TLRs, and NF-κB expressions in cartilage.Probiotics and Antimicrobial ProteinsKorotkyi et al. [112]2021
Curcumin→ ERK1/2↓ Chondrocyte ApoptosisCurcumin inhibits apoptosis of chondrocytes through activation ERK1/2 signaling pathways induced autophagy.NutrientsLi et al. [52]2017
┤NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
Curcumin reduces inflammation in knee osteoarthritis through blocking TLR4/MyD88/NF-κB signal pathway.Drug Development ResearchZhang et al. [113]2018
→ Nrf2/HO-1↑ GPX1,3,4
↑ SOD1
↑ CAT
↑ GST
Curcumin inhibits chondrocytes inflammation through the Nrf2/ARE signaling pathway, thereby exerting cartilage protective effects.Cell Stress and ChaperonesJiang et al. [114]2020
┤ TLR4/NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
Curcumin improve neuroinflammatory process by reducing microglia/macrophage activation and neuronal apoptosis through a mechanism involving the TLR4/NF-κB signaling pathway in microglia/macrophages.International Journal of Molecular SciencesPanaro et al. [115]2020
┤ NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
Curcumin reduces expression of NF-κB and ROCK1.Journal of Cellular and Molecular MedicineQiu et al. [116]2020
Eupatilin┤ NF-κB
┤ JNK
┤ p38MAPK
↓ ADAMTS-4
↓ COX-2
↓ MMP
Eupatilin suppressed expression of MMPs, ADAMTSs in chondrocytes by reducing JNK phosphorylation and NF-κB and MAPK signaling.PharmaceuticalsLee et al. [117]2021
Genistein→ Nrf2/HO-1↑ GPX1,3,4
↑ SOD1
↑ CAT
↑ GST
Genistein downregulates MMPs, ADAMTSs via NF-κB signaling pathway by blocking IκB degradation and activating Keap1/Nrf2 pathway.NutrientsLiu et al. [118]2019
┤ NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
BioMed Research InternationalYaun et al. [119]2019
Molecular Medicine ReportZou et al. [120]2020
Glucosamine→ mTOR↓ Chondrocyte ApoptosisGlucosamine promotes osteoblast proliferation by modulating autophagy via the mTOR pathway.Biomedicine & PharmacotherapyLv et al. [121]2018
→ Wnt/β-catenin↓ Chondrocyte ApoptosisGlcN increases β-catenin nuclear translocation, thus promoting chondrocyte proliferation.International Journal of Molecular MedicineMa et al. [49]2018
Green tea polyphenol┤ NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
L-theanine inhibits upregulation of MMPs, as well as inhibiting NF-κBNutrientsBai et al. [122]2020
Green tea catechins increase NF-κB inhibitors expressionAntioxidantsLuk et al. [123]2020
→PI3K/AKT↓ FOX-O1Epigallocatechin-3-gallate modulating AKT-FoxO1 via upregulating miR-486-5p.Archives of Biochemistry and BiophysicsChang et al. [124]2020
Jaceosidin┤ NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
Jaceosidin attenuates cartilage destruction by suppressing MMPs, ADAMTSs and the NFκB signaling pathway by blocking IκB degradation.Journal of Cellular and Molecular MedicineLee et al. [117]2019
Omega-3 PUFA┤ p38MAPK↓ Chondrocyte ApoptosisPUFA inactivates of p38MAPKInternational Journal of Molecular MedicineWang et al. [58]2016
→ PI3K/AKT
┤ NF-κB
↓ MMPsPUFA metabolite suppresses MMP-13 secretion by activating PI3K/AKT pathway directly, while inhibiting NF-κB pathway. Connective Tissue ResearchLu et al. [74]2020
OOP┤ NF-κB
┤ p38MAPK
↓ ADAMTS-4
↓ COX-2
↓ MMP
OOPs inhibited IL-1β-induced expression of inflammatory mediators through suppressing NF-κB and MAPK activation in chondrocytes.Food & FunctionFeng et al. [125]2017
→ Nrf2/HO-1
┤ NF-κB
↓ ADAMTS-4
↓ COX-2
↓ MMP
OOPs can activate Nrf-2 signaling and the blockage of NF-κB nuclear translocationCellsSerrelli et al. [126]2020
┤ NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
OOPs reduce the inflammatory and catabolic factors mediated by NF-κB (IL-1ß, IL-6, COX-2 and MMP-3AgingVarela-Eirín et al. [127]2020
┤ NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
Mechanistically, OOPs exhibited an anti-inflammatory effect by inactivating the PI3K/AKT/NF-κB pathway.Journal of Cellular PhysiologyChen et al. [128]2021
Quercitin┤ NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
Quercetin inhibits IL-1b and TNF-a production via TLR-4/NF-κB pathway.Journal of International Medical ResearchZhang et al. [129]2019
Resveratrol→Wnt/β-catenin↓ Chondrocyte ApoptosisRev increased osteoblastogenesis and bone formation through stimulation of Wnt signaling pathway.Journal of Cell PhysiologyAshrafizadeh et al. [47]2020
→Nrf2/HO-1↑ GPX1,3,4
↑ SOD1
↑ CAT
↑ GST
Res modulates the Nrf2 activation by inhibiting Keap1, Nrf2 gene expression, changing the upstream mediators of Nrf2, and potentiating the nuclear translocation of Nrf2.Biomedicine & PharmacotherapyFarkhondeh et al. [130]2020
→PI3K/AKT↓ FOX-O1Resveratrol may exert anti-OA effect by enhancing the self-limiting mechanism of inflammation through TLR4/Akt/FoxO1 axis.Drug Design, Development and TherapyXu et al. [131]2020
┤ NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
Resveratrol alleviates the interleukin-1β-induced chondrocytes injury through the NF-κB signaling pathway.Journal of Orthopaedic Surgery and ResearchYi et al. [132]2020
Sulforaphane→ Nrf2/HO-1↑ GPX1,3,4
↑ SOD1
↑ CAT
↑ GST
Sulforaphane ameliorates oxidative stress suppressing inflammatory cytokines and activating Keap1/Nrf2 pathway.Free Radical Biology and MedicineYang et al. [133]2020
Sulforaphane inhibits the production of inflammatory cytokines.PLoS ONEMoon et al. [134]2021
┤ JNK↓ ADAMTS-4
↓ COX-2
↓ MMP
Sulforaphane inhibits osteoclastogenesis by suppressing autophagy modulating JNK pathway.MoleculesLou et al. [135]2021
Wogonin┤ NF-κB↓ ADAMTS-4
↓ COX-2
↓ MMP
Wogonin downregulates NF-κB pathway and genes involved in inflammatory-response.Nature: Scientific ReportKhan et al. [136]2017
Abbreviations: ADAMTS = A disintegrin and metalloproteinase with thrombospondin motifs; AKT = Tyrosine kinase A; CAT = Catalase; COX-2 = Cyclooxygenase-2; ERK = Extracellular signal-regulated kinases; FOX-O1 = Forkhead box protein O1; GPX = glutathione peroxidase; GST = Glutatione S-transferasi; HIF-2α = Hypoxia-inducible factor; HO-1 = Heme oxygenase 1; MAPK = Mitogen-activated protein kinases; MMP = Matrix metalloproteinase; mTOR = mammalian target of rapamycin; NF-κB = Nuclear factor kappa-light-chain-enhancer of activated B cells; Nrf2 = Nuclear factor-erythroid factor-2; OOP = Olive Oil Polyphenols; PI3K = Fosfoinositide 3-chinasi; PUFA = Polyunsaturated Fatty Acids; SOD = Superoxide dismutase; TLR4 = Toll-like receptor; Wnt = Wingless-related integration site.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

de Sire, A.; Marotta, N.; Marinaro, C.; Curci, C.; Invernizzi, M.; Ammendolia, A. Role of Physical Exercise and Nutraceuticals in Modulating Molecular Pathways of Osteoarthritis. Int. J. Mol. Sci. 2021, 22, 5722. https://doi.org/10.3390/ijms22115722

AMA Style

de Sire A, Marotta N, Marinaro C, Curci C, Invernizzi M, Ammendolia A. Role of Physical Exercise and Nutraceuticals in Modulating Molecular Pathways of Osteoarthritis. International Journal of Molecular Sciences. 2021; 22(11):5722. https://doi.org/10.3390/ijms22115722

Chicago/Turabian Style

de Sire, Alessandro, Nicola Marotta, Cinzia Marinaro, Claudio Curci, Marco Invernizzi, and Antonio Ammendolia. 2021. "Role of Physical Exercise and Nutraceuticals in Modulating Molecular Pathways of Osteoarthritis" International Journal of Molecular Sciences 22, no. 11: 5722. https://doi.org/10.3390/ijms22115722

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