Next Article in Journal
Exome Sequencing for the Diagnostics of Osteogenesis Imperfecta in Six Russian Patients
Previous Article in Journal
A Tissue Engineered 3D Model of Cancer Cell Invasion for Human Head and Neck Squamous-Cell Carcinoma
Previous Article in Special Issue
The Role of Alarmins in the Pathogenesis of Rheumatoid Arthritis, Osteoarthritis, and Psoriasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Osteoarthritis: Insights into Diagnosis, Pathophysiology, Therapeutic Avenues, and the Potential of Natural Extracts

by
Chiara Coppola
1,†,
Marco Greco
2,†,
Anas Munir
1,
Debora Musarò
2,
Stefano Quarta
2,
Marika Massaro
3,
Maria Giulia Lionetto
2 and
Michele Maffia
4,*
1
Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Via Lecce-Arnesano, 73100 Lecce, Italy
2
Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
3
Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy
4
Department of Experimental Medicine, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Curr. Issues Mol. Biol. 2024, 46(5), 4063-4105; https://doi.org/10.3390/cimb46050251
Submission received: 28 February 2024 / Revised: 5 April 2024 / Accepted: 18 April 2024 / Published: 29 April 2024
(This article belongs to the Special Issue Molecular Research in Osteoarthritis and Osteoarticular Diseases)

Abstract

:
Osteoarthritis (OA) stands as a prevalent and progressively debilitating clinical condition globally, impacting joint structures and leading to their gradual deterioration through inflammatory mechanisms. While both non-modifiable and modifiable factors contribute to its onset, numerous aspects of OA pathophysiology remain elusive despite considerable research strides. Presently, diagnosis heavily relies on clinician expertise and meticulous differential diagnosis to exclude other joint-affecting conditions. Therapeutic approaches for OA predominantly focus on patient education for self-management alongside tailored exercise regimens, often complemented by various pharmacological interventions primarily targeting pain alleviation. However, pharmacological treatments typically exhibit short-term efficacy and local and/or systemic side effects, with prosthetic surgery being the ultimate resolution in severe cases. Thus, exploring the potential integration or substitution of conventional drug therapies with natural compounds and extracts emerges as a promising frontier in enhancing OA management. These alternatives offer improved safety profiles and possess the potential to target specific dysregulated pathways implicated in OA pathogenesis, thereby presenting a holistic approach to address the condition’s complexities.

1. Introduction

In recent decades, we have witnessed a continuous improvement in the overall quality of life (QoL) in most developed countries. While this has improved longevity and reduced mortality from infectious diseases, it has also allowed a range of non-infectious, chronic degenerative diseases to emerge, favored by several conditions and environmental factors, including hectic lifestyles, unhealthy nutrition, sedentary habits, and constant exposure to pervasive environmental pollution [1]. Consequently, for most developed countries today, these diseases represent the main cause of mortality and disability.
Prominent clinical conditions impacted by these trends include Parkinson’s disease (PD), osteoarthritis (OA), and type II diabetes mellitus (T2DM). Remarkably, since 1990, their prevalence has surged by 155.5%, 132.2%, and 129.7%, respectively. Presently, the global tally of diagnosed cases includes over 590 million individuals with OA, more than 530 million with T2DM, and exceeding 8.5 million with PD [2,3,4,5]. These conditions, which often occur concurrently in the same individual, present formidable challenges to healthcare systems and significantly compromise the QoL for those affected.
The term OA derives from the Greek words “ostheo-”, meaning “of the bone”, and “-arthritis”, which in turn is a combination of the two words “arthr-” and “-itis”, which stands, respectively, for “joint” and “inflammation” [6]. Encompassing a heterogeneous group of disorders affecting diarthrodial joints and sharing common biological features and clinical outcomes, OA represents one of the most common, invalidating medical conditions in adults. In the body, articular cartilage acts as a shock absorber for the ends of bones within a joint, thanks to its unique structural composition. It is made up of chondrocytes, which are cells that produce and maintain the turnover of the cartilaginous matrix, and an extracellular matrix (ECM) that is predominantly composed of collagen and proteoglycans [7]. This combination imbues the cartilage with a high-water content, granting it both mechanical resistance and durability. On the other hand, the sparse vascularization of the tissue and chondrocytes’ characteristically low metabolic activity limit the capacity of the cartilage to effectively regenerate after injury or natural degradation that occurs over time [8].
For a long time, OA was a mainly wear and tear process of the joint cartilage, and for this reason, the condition was denoted as “osteoarthrosis”, with the Greek term “-osis” indicating a degenerative process without inflammation. However, since the 1980s, it has become apparent that an inflammatory component plays a significant role in the pathogenic process. Consequently, the name of the disease has been revised to reflect this newfound understanding [9].
To date, two main forms of OA have been recognized, namely primary or idiopathic and secondary, as per the causes associated with its onset. While the etiology is complex, the risk factors vary with the forms of the disease. For primary OA, the primary contributors are age, sex, ethnicity, and genetics [10]. According to the World Health Organization, it typically develops between the late 40s and mid-50s, with about 73% of patients over the age of 55, and of these, 60% are female [11]. This can be attributed to natural aging processes, which cause a reduction in synovial fluid, and changes in its composition and quality. Moreover, in advanced age, the body has gone through considerable traumatic stress and wear, and the inflammatory processes that are triggered then lead to alterations in bone and cartilage, causing osteophytes [12]. The higher incidence of OA in females is believed to be primarily linked to hormonal factors. These factors include hormonal fluctuations during menstruation cycles and, notably, postmenopausal changes [13]. Estrogens play a crucial role in cartilage protection, inflammation modulation, and bone metabolism promotion [14,15,16]. Additionally, pregnancy, with its associated weight gain and prolonged hormonal fluctuations, weakens joints, particularly those in the lower part of the body and the spine [17,18]. Sex differences extend to joint alignment, with females generally exhibiting a higher quadriceps angle, lower arch height index, and a broader range of internal and external rotation in the hip joint. These factors result in prolonged stress on the knee and hip joints over time [19].
Secondary forms of OA, on the other hand, may be attributed to causative events capable of weakening the structure of the joint [10]. Among these, traumatic events at the level of articulation, sports activities or demanding jobs, conditions like overweight or obesity, metabolic diseases like diabetes or gout, joint malalignment, congenital deformity, body length, bone inequity, and reduced support to the structure due to weakness of ligaments or surrounding muscle tissue [20,21].
Investigating the pathogenesis of OA remains an ongoing challenge, with much yet to be explored and comprehended. Notably, the factors influencing the diverse timing of interindividual progression, the intricate dynamics of communication among cartilage and surrounding tissues, and the distinct roles played by molecules implicated in various forms of inflammation require further elucidation [22].
As of now, the therapeutic landscape for OA aims at mitigating pain symptoms, enhancing joint function, and, ideally, impeding or delaying their worsening. This typically involves the systemic or local administration of analgesic and/or anti-inflammatory molecules [23]. This conventional approach is often complemented by manual and instrumental physiotherapeutic interventions strategically designed to alleviate pain and fortify the muscles supporting the compromised joint [24]. Surgical prosthetic intervention, albeit considered a last resort, is the only definitive solution, yet it remains fraught with uncertainties concerning outcomes and recovery [25,26].
In recent years, there has been a growing acknowledgment of the significance of natural-type molecules, nutraceuticals, and various plant-based extracts. These substances have indeed shown the potential to act through mechanisms complementary to conventional drugs, offering lower side effects and, perhaps, more specific targeting of pathways fundamental to the pathophysiology of OA [27,28].
This article aims to report some of the most recent findings regarding alternative therapeutic regimes using natural extracts, correlating with the latest understanding of the pathogenesis mechanisms of OA.

2. Diagnosis

In the context of OA, pain represents a cardinal symptom, often manifesting as either persistent or intermittent discomfort, prompting people to seek medical attention. It results from inflammation and abnormal friction between the joint surfaces, and it is one of the warning bells that leads the physician to a diagnosis [29]. Individuals with OA may encounter pain while moving the affected joints or even at rest, and the severity varies with the stage of the disease. Additionally, joint stiffness, swelling, reduced flexibility, and, in advanced stages, joint deformity are common manifestations of OA [30]. For physicians, the diagnostic task relies exclusively on clinical assessments, and no laboratory test provides direct support, even if some circulating molecules can represent useful tools for differential diagnosis. In any case, an OA setting, given its inflammatory nature, may be associated with an increase in C-creative protein (CRP) and erythrocyte sedimentation rate (ESR) in serum [31,32]. On the other hand, the evaluation of additional parameters such as complete blood count (CBP), as well as the search for rheumatoid factor (RF), anti-cyclic citrullinated peptide (Anti-CCP), or antinuclear antibody (ANA) allows the exclusion of a rheumatoid arthritis condition [33,34,35,36].
The lack of clear-cut biomarkers underscores the need for a thorough assessment of symptoms, medical history, and physical examinations to achieve a precise diagnosis [37,38]. Furthermore, the need to distinguish OA from other clinical conditions, including inflammatory, infectious, or crystal deposition (e.g., gout) arthritis, as well as soft tissue injuries like bursitis, tendinitis, and meniscal tears, adds complexity to the diagnostic process [39,40,41,42]. Moreover, the uniqueness of the disease manifests in each joint, encompassing distinct onsets, progressions, and physical examination findings.
Numerous biomarkers have emerged in recent decades, with a focus on structural molecules associated with cartilage, bone, or synovium, specific to particular joints or referable to different ones. They are often detectable in circulating blood or urine, enabling noninvasive collection and correlation with cartilage, bone, or ECM-altered metabolism, as well as ongoing inflammatory pathways [43,44,45,46,47].
The Osteoarthritis Biomarkers Network is actively driving the research of novel biomarkers for OA, proposing a classification system based on the burden of disease, investigative, prognostic, efficacy of intervention, and diagnostic (BIPED) categories [48].
Given the chondrodegenerative nature of OA, circulating molecules reflective of cartilage turnover represent extremely promising diagnostic and prognostic indicators. Notably, epitopes of Fibulin-3 (Fib3-1, Fib3-2, and Fib3-3), aggrecanase-generated aggrecan (ARGS), cartilage oligomeric matrix protein (COMP), and C-telopeptide of type II collagen (CTX-II), along with metabolites like acylcarnitines, uric acids, cystine, and tyrosine, frequently exhibit altered levels in this condition [49,50,51,52,53,54,55].
Moreover, markers associated with low inflammation levels, such as TNF-α, IL-6, and IL-1β, are under extensive investigation and are proposed as discriminators between OA and other joint disorders characterized by a severe inflammatory component [56,57]. Additionally, small noncoding RNAs, particularly microRNAs (miRNAs), are emerging as useful tools in correlating their fluctuations with OA severity and in distinguishing them from other joint disorders [58,59,60].
Recently, Kolhe et al. (2017) demonstrated that well-defined pools of miRNAs can serve as sex-specific markers of OA. This is particularly important due to the higher incidence of the condition in women [61].
To date, however, none of these biomarkers has proven to be sufficiently discriminating, neither in the context of routine diagnosis nor in the evaluation of the disease progression.
Imaging serves as valuable assistance for physicians by confirming diagnoses and excluding alternative pathologies. In this regard, plain radiography emerges as a useful tool. While less commonly employed, alternative diagnostic methods like MRI and CT are utilized in the diagnosis of OA, and in many cases, MRI excels in detecting OA at earlier stages compared to conventional radiographs [62]. Additionally, ultrasonography can contribute to the diagnosis by identifying synovial inflammation, effusions, and any osteophyte formations associated with OA [63]. Ultrasound, characterized by its noninvasive, swift, and cost-effective nature, serves as an imaging technique for observing joint changes. However, a limitation lies in its inability to visualize bone conditions, restricting its diagnostic scope to soft tissues [64]. Furthermore, the data it produces may not fully align with the Kellgren and Lawrence (KL) radiological-based scale, established in 1957 by the eponymous physician duo and acknowledged by the World Health Organization (WHO) since 1961 as a classification system for the severity of OA [65,66].
The KL classification system, originally defined to assess conditions related to the knee, is a tool widely used in clinics, epidemiological studies, and research contexts [67,68,69]. After conducting a radiographic analysis, according to its guidelines, it is possible to assign a score ranging from 0 to 4 to the subject, reflecting the severity of OA, as outlined in Table 1 [70].
Despite its widespread adoption and ongoing revisions, the KL classification system has faced some criticism over time. Firstly, it relies on the presence or absence of osteophytes, which may not always be detectable. Furthermore, such formations are not clearly defined and leave considerable leeway for the subjectivity of the examiner. The scale also ignores other factors that affect OA, such as cartilage, synovial fluid, and soft tissue changes, as well as the patient’s symptoms [72,73,74]. Moreover, it is not compatible with other imaging modalities [73]. However, despite many efforts to develop more objective and reliable scales for OA diagnosis and evaluation [70], none of them have found the same widespread acceptance in the clinical and research community as the KL scale.

3. Pathogenesis

The axial skeleton represents the framework of the body, preventing the collapse of internal organs and offering protection. The joints, from the Latin “iuncus”, meaning “united”, interconnect the bones to allow their movement, also determining their direction and range when muscle tensions or external forces impact them. Of the different types of joints existing in our body, diarthrodial or synovial ones are lubricated by synovial fluid for smooth movement, exhibiting a complex architecture while sharing many fundamental elements.
The articulating ends of the bones are enveloped in a smooth, hyaline cartilage layer designed to minimize friction and offer cushioning, thus giving rise to a synovial cavity encapsulated by a sturdy capsule, serving to stabilize it [75]. At the interface between the bone and the hyaline cartilage, a chondro-osseous junction is present in the form of mineralized cartilage, bound to the bone by a lower cement line and, by the upper tidemark, to the articular soft cartilage [76,77]. The capsule comprises an outer layer of densely packed connective tissue, which is sparsely vascularized but richly innervated, firmly attached around the entire circumference of each bone articular end. On the inner side of the capsule lies the synovial membrane, endowed with secretory capabilities, responsible for producing synovial fluid [78]. Changes in the elements comprising joints are linked to the onset and progression of OA. While these alterations may initially impact just a single component, such as cartilage or bone, the interconnected nature of joint structures results in a condition that affects the entire joint, both physically and functionally [79]. In time, this causes a condition of persistent pain, disability, loss of function, and decreased QoL.
The pathophysiology of OA is still far from being fully understood despite some factors being well known. OA has long been linked primarily to the degeneration of articular cartilage, often stemming from factors such as trauma, overweight, metabolic disorders, and genetic predispositions. However, current understanding suggests that subchondral bone lesions play a pivotal role in the early stages of this disease, contributing to the development of ectopic bone and osteophytes [80,81]. It is important to highlight that the subchondral bone provides mechanical and trophic support to the joint cartilage, so its alteration can significantly impact the metabolic health of the joint cartilage [79].
In this context, the chronic low-grade inflammation of the synovial lining emerges as a central player in the pathophysiology of OA, with immunological mechanisms increasingly recognized as the main drivers in inflammation-induced tissue damage [82]. Moreover, neuroinflammation and central sensitization mechanisms are pivotal in initiating and perpetuating pain in the disease progression [83].
Articular cartilage is a tissue extremely resilient to mechanical stress and capable of withstanding loads far beyond normal body demands. It is composed of a matrix, the key components of which include type II collagen, hyaluronic acid, aggrecan, and various highly hydrated proteoglycans, secreted by a population of specialized cells called chondrocytes, which account for 1% of its total composition [84,85,86,87,88]. Despite its remarkable durability, however, articular cartilage exhibits a limited capacity for self-repair [6,89]. As aforementioned, cartilage lacks a robust blood vessel network, and the chondrocytes, which live in a mostly hypoxic environment, rely mainly on a nutrient diffusion process for sustaining their metabolism and their secretory activity [90,91].
The pericellular matrix, a specialized layer of ECM enveloping one to eight chondrocytes, forms the essential architecture of the chondrons, which represent the functional secreting units of the joint cartilage [92,93]. It preserves chondrocyte activity, shielding these cells from deleterious interactions with ECM components while regulating the flow of nutrients. The pericellular matrix consists of most of the elements of the ECM, with the addition of type VI and IX collagen, which greatly bolsters its resilience against external forces [94,95].
Chondrocytes, through interaction with their envelope via surface integrins acting as mechanical–chemical receptors, perceive mechanical stresses on the cartilage, thereby stimulating the deposition and turnover of matrix components [96]. Integrins play an important role in signal transduction, serving as mediators not only in mechanotransduction processes but also in processes of cell adhesion, migration, and inflammation response.
According to recent findings, a loss in the homeostasis of integrins signaling transduction is associated with OA onset [97,98,99]. During earlier stages of OA, cartilage undergoes several changes in its composition and structure. Physiologically, the structure and the composition of the ECM are handled by metalloproteinases (MMPs), adamalysins (ADAMs), and ADAM with thrombospondin motifs (ADAMTSs), whose activity is tightly regulated and balanced by tissue inhibitors of MMP (TIMPs) [100,101]. Physiologically, indeed, cartilage exists in a state of fluid balance between catabolism and anabolism; pathologically, this equilibrium is moved toward a greater MMP activity and, so, toward its catabolism. In the initial phases of OA, chondrocytes respond by increasing their synthesis of matrix, trying to restore the integrity of the compromised structure. The damage to the pericellular matrix, however, destroys their niche, causing a loss in their capability to efficiently perceive the state of the surrounding area [102,103].
Werb and coworkers (1989), in their pioneering in vitro work, observed that integrins could mediate, as a form of mechanical response, the increase in transcription and secretion of several MMPs, such as MMP-1, MMP-3, MMP-10, and MMP-13, with the latter having a pronounced activity in degrading type II collagen [104,105]. At the end of 1980, moreover, several studies found how the homeostasis of the cartilage was preserved thanks to the activity of the transforming growth factor beta (TGF-β), having a role in inhibiting the activity of the TIMPs [106].
TGF-β has also been shown to modulate the expression of integrins. The molecule has an important role in chondrocyte differentiation and maturation, as well as promoting cartilage synthesis. However, when present at high levels, it can have detrimental effects on cartilage integrity, compromising chondrocyte metabolic activity. Integrins themselves can respond to mechanical stimulation by inducing the release of TGF-β, but when this pathway is misruled, they contribute to ECM destruction [22,107].
One of the most deleterious consequences of the dysregulated activity of MMPs is the sustained activation of the chondrocyte pathway downstream of the α5β1 integrin, also known as the fibronectin receptor. This interaction occurs with a soluble fibronectin fragment generated from the proteolytic cleavage of the full-length protein [108,109]. Subsequently, this cascade triggers a significant activation of PKCδ, which in turn induces the activation of nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK), eliciting a complex cellular response [110,111,112]. The NF-κB pathway stands as a potent proinflammatory agent, exerting significant influence in the initiation of OA. Its activation can also be triggered by various proinflammatory cytokines, such as IL-1β, TNF-α, and IL-6, whose concentrations increase within the joint during inflammation, but it also perpetuates its activity through a positive feedback loop, driving chondrocytes to enhance their synthesis [113,114]. This cascade of events underscores the critical role of NF-κB in the perpetuation and exacerbation of OA pathology [115]. On the other hand, MAPK can suppress the synthesis of ECM components while simultaneously stimulating the synthesis and release of MMPs [116,117]. Together, the NF-κB and MAPK signaling pathways contribute to ECM degeneration and the progression of OA by generating reactive oxygen and nitrogen species (RONS), prostaglandin E2, MMPs, and ADAMTSs [118,119,120,121].
The gradual degradation experienced by cartilage initiates a calcification process, affecting both its superficial and deeper layers, eventually leading to the delamination and exposure of underlying bone over time [122]. This progression appears to be facilitated by the deposition of calcium crystals within chondrocytes and their surrounding regions, facilitated by the release of vesicles containing mineral salts. The discovery that these vesicles are enriched with microRNAs hints at a greater complexity underlying the process than previously understood [123]. From the bone marrow, new vessels and sensitive nervous terminations start to grow through the new fissures in the osteochondral junction, surrounded by novel bone [76]. This phenomenon also arises from the migration and excessive proliferation of new chondrocytes, which commence depositing new layers of type X collagen. However, unlike the typical physiological chondrogenic process, these layers are not replaced by type II collagen but instead undergo calcification (Figure 1) [124,125].
Recent findings have shed light on one of the mechanisms that could correlate hormone activity on the cartilage with the greater incidence of OA in the female sex. Wang and colleagues (2021) have observed a role for estrogen receptor α (ERα) in chondrocyte senescence, where the phenotype of these cells is influenced by the levels of this receptor. Utilizing RNA sequencing techniques, they observed differential transcription of the estrogen receptor-1 (ESR1) gene, encoding ERα, in chondrocytes between healthy individuals and OA patients [127]. This finding complements the observation of low ERα levels measured in OA-affected joints [128]. Furthermore, their study revealed that restoring ERα levels in chondrocytes to physiological levels reduces senescence, thereby ameliorating the pathological phenotype. Conversely, knockout of ERα exacerbates senescence, compromising the chondrocyte’s ability to efficiently respond to mechanical stress [127].

4. Treatment

While there is currently no cure for OA, treatment primarily focuses on mitigating modifiable risk factors before its onset and alleviating its symptoms once it occurs.
Proactive measures, such as weight loss or modification of conditions of postural or orthopedic abnormalities, stand out as an effective means to diminish the likelihood of developing OA in the lower limbs and spine joints [129,130,131]. Weight loss also represents the first line of treatment once OA manifests, especially in conditions affecting the knee [131]. However, during the acute pain phases of OA, patients often require a pharmacological approach to enhance their QoL. Effectively managing symptoms becomes pivotal, marking a shift toward targeted interventions aimed at minimizing discomfort and optimizing overall well-being.
Managing patients with OA demands a comprehensive approach, considering the progressive nature of the condition and its profound impact on patients’ QoL. While physiotherapy and rehabilitation are valuable components, they may not always suffice in addressing the debilitating pain associated with OA. Therefore, integrating pharmacological interventions such as nonsteroidal anti-inflammatory drugs (NSAIDs) and analgesics becomes essential [132,133,134]. Additionally, personalized treatment plans, overseen by medical specialists, may incorporate intra-articular therapies like steroids, platelet-rich plasma (PRP), and hyaluronates to augment symptom management [135,136,137].
Moreover, since the nature of OA-related pain is persistent and potentially distressing, it is essential to address its psychological impact. Collaborating with healthcare professionals like psychologists can provide valuable support alongside physicians and physical therapists, helping patients cope with pain and any associated depressive symptoms [138,139].
Ultimately, when conservative measures fail to provide adequate relief and functional improvement, surgical interventions may become necessary to address the specific joint pathology effectively.

4.1. Physical Treatments in OA

According to the latest and most authoritative guidelines, physical exercise stands out as the primary intervention in managing overt cases of OA, alongside education for self-management and physiotherapy [140,141]. Nevertheless, it is crucial to contextualize the therapy, considering that it may vary significantly depending on the specific joint affected by the degenerative process. Comorbidities also have to be considered to suggest to the individual not only the safest and most appropriate exercise but also the duration and intensity of the training sessions.
Physical exercise has demonstrated greater effectiveness in managing OA affecting the knee or hip compared to the hand [142]. There is no “one-size-fits-all” approach to exercise, as different activities suit different individuals based on their preferences, fitness levels, and health conditions. However, some popular forms of exercise, such as walking, using treadmills, or cycling, are noteworthy for their popular adoption and their positive reception by patients. Additionally, aquatic exercises serve as a valuable tool in OA management, strengthening the benefits of aerobic activity while significantly reducing the strain on the joints in a water-based environment [143]. Tai chi is highly recommended in individuals with knee or hip OA, as it has demonstrated effects on both physical and mental well-being [142]. Similarly, while yoga appears to offer significant benefits to patients, there is a dearth of literature recommending its use [142,144,145].
Finally, numerous studies have highlighted the utility, particularly in cases of knee OA, of strengthening the muscles that support the affected joint through targeted rehabilitation or training programs. Weakness in muscles such as the femoral quadriceps can lead to increased knee joint loading. However, the optimal approach to achieve this goal remains a subject of debate among medical societies [146].

4.2. Pharmacological Treatments in OA

4.2.1. Nonsteroidal Anti-Inflammatory Drugs

In the context of OA, the second-line treatment involves NSAIDs and analgesics. These compounds act by inhibiting prostaglandin-endoperoxide synthase, also known as cyclooxygenases (COXs), oxidoreductases facilitating the conversion of arachidonic acid into prostanoids [147]. Among the various isoforms of COXs, the inducible expression of COX-2 is closely associated with inflammatory cytokines such as IL-1b and TNF-a, in addition to oxidative stress [148]. Consistent with these findings, several studies have documented an elevated level of these inflammatory mediators, coupled with an increased amount of proinflammatory nitric oxide (NO) and a significant rise in the level of COX-2 within the cartilage of individuals affected by OA [149,150]. These observations underscore the interconnection between inflammatory processes, oxidative stress, and clinical conditions.
However, the use of NSAIDs comes with several drawbacks associated with the inhibition of prostaglandin synthesis. Non-selective COX inhibitors impact both COX-1 and COX-2 isoforms, affecting the gastric mucosa. Notably, COX-1 is crucial for protecting the stomach against ulcerations and bleeding [151]. Conversely, COX-2-specific inhibitors, designed to spare isoform 1 activity, are linked to an elevated risk of thrombosis [152].
In the body, while both COX-1 and COX-2 contribute to prostaglandin synthesis, COX-2 represents the primary source of prostacyclins, playing roles in inflammation and vasodilation [153,154]. Meanwhile, COX-1 catalyzes the synthesis of thromboxanes, exerting a potent vasoconstrictive effect [155]. The inhibition of COX-2 therefore shifts the balance toward increased vasoconstriction and prothrombotic activity [156].
Opting for NSAIDs in cream or gel formulation stands out as a preferred approach for managing OA, especially among the elderly. While these topicals may exhibit a slower absorption rate, their pharmacological activity is comparable to oral alternatives [157,158]. Their use is recommended by the main international guidelines for knee and hand OA treatment according to superior safety profile. The American College of Rheumatology strongly endorses the topical application of NSAIDs over oral consumption, particularly emphasizing this for individuals aged 75 years or older with knee OA. This recommendation holds particular significance for those with coexisting conditions and heightened risks of cardiovascular, gastrointestinal, or renal side effects, which are commonly observed in this age group [159].

4.2.2. Steroidal Anti-Inflammatory Drugs

Among the most powerful molecules capable of mitigating inflammatory responses in the human body are corticosteroids. Derived from cortisones, this class of drugs exerts its influence by downregulating the expression of numerous genes through interactions with their transcription factors [160,161]. However, prolonged use of corticosteroids is associated with significant side effects, including weight gain, swelling, hypertension, diabetes, and increased susceptibility to infections [162,163,164,165]. The literature contains poor data about the enteral use of such a class of molecules in OA contexts. The limited number of trials existing has shown to provide just slight benefits in pain relief in both hand and knee OA, although limited in time [166,167].
Another therapeutic strategy involves the direct administration of corticosteroid injections into the affected area. Initially, the pioneering work of Hollander and colleagues in 1951 was performed in knee arthritis rheumatoid [168], with Miller and coworkers replicating the treatment for OA subjects seven years later [169].
Since then, other joints affected by OA have also become sites of injection, and this form of therapy remains an area of active research [170,171,172,173]. These investigations aim to assess the duration of the anti-inflammatory and analgesic effects of corticosteroids, considering that the latest evidence indicates a relatively short duration, from a few weeks to months, necessitating careful consideration when determining the appropriate posology for treatment [166].
If a local use of corticosteroid keeps the drug effects mostly localized to the joint, reducing at the same time systemic side effects, some drawbacks in the procedure remain. Despite now being quite rare, infections can follow the procedure; in addition, some studies have associated the treatment with local side effects like necrosis, tendon weakening, and cartilage reduction [174,175,176,177]. On the other hand, despite being rare, some systemic responses to the drug may be observed, like headache, insomnia, short-term glucose levels increase, hypothalamic–pituitary–adrenal axis suppression, and iatrogenic Cushing syndrome [178,179,180].
According to Osteoarthritis Research Society International (OARSI), European League Against Rheumatism (EULAR), and Royal Australian College of General Practitioners (RACGP) guidelines, intra-articular corticosteroid use, together with physical exercise, is recommended for short-term pain reduction in knee OA, conditionally and limitedly in time [181], while other societies maintain a more cautious approach to the topic, like the American Academy of Orthopedic Surgeons (AAOS) [182].

4.2.3. Disease-Modifying OA Drugs

In recent years, with the attempt to relieve patients’ pain and block the progression of OA, efforts have been directed toward the research of innovative pharmacological strategies. Although the use of NSAIDs and corticosteroids provides some symptomatic relief, their benefits are modest, limited in time, and carry the risk of adverse events (AEs).
Several potential molecular targets have been identified thanks to the comprehension of the pathways involved in the condition’s onset and progression. These include matrix-degrading proteases, mechanisms of altered senescence of chondrocytes, cartilage repair mechanisms, bone remodeling processes, and low-grade inflammation mediators. Disease-modifying OA drugs (DMOADs) are a group of molecules capable of intervening in specific molecular mediators of these processes. Among the most promising targets are matrix metalloproteinases such as MMP-13 protease or ADAMTS-4 and -5 peptidase, growth factors like fibroblast growth factor-18 (FGF-18), bone morphogenetic protein (BMP-7), or TGF-β, cytokines, and small molecule like TNF-α or IL-1β [183,184,185,186,187,188,189,190,191,192].
Additional approaches to preserve cartilage health include targeting cellular senescence, a mechanism associated with stress response [193]. In OA, oxidative stress is associated with a premature joint cellular component aging process, affecting not only chondrocytes but also synovium fibroblasts, osteoblasts, osteoclasts, and musculoskeletal cells [194,195,196,197]. This gives origin to a senescence-associated secretory phenotype (SASP), resulting in a massive release of proinflammatory cytokines and proteases in the joint space [198].
Current research is focused on pursuing a pharmacological approach to the phenomenon by developing molecules with senolytic and/or senomorphic activity. Senolytic molecules can suppress anti-apoptotic or pro-senescence pathways, often upregulated in pathology. Among their targets are the anti-apoptotic PI3K/Akt pathway and B cell lymphoma family proteins Bcl-2, Bcl-XL, and Bcl-W, as well as pro-senescence proteins p15, p16, p21, and p53 [199,200,201,202,203,204]. On the other hand, senomorphics act by inhibiting cells’ SASP or neutralizing their biological effect; promising targets include AMPK signaling, IL-6 receptors, IL-8, and IL-1β, which in turn inhibit MMP-13 and ADAMTS5 production. Preliminary studies on senomorphic preparations capable of directly inhibiting matrix-degrading enzymes are also ongoing [205,206,207,208,209,210].
Finally, OA presents itself as an extremely painful condition. Despite the reduced innervation of the cartilage, the joint is rich in sensorial terminations, which, during the pathological degeneration of the tissue, start to perceive the effects of cytokines and proinflammatory stimuli [211,212]. The peripheral sensitization is followed by a central sensitization with the establishment of conditions of allodynia or hyperalgesia due to mechanical stresses. Increased levels of nerve growth factor (NGF) are commonly observed in synovial fluid, which is released by joint cells during OA progression and associated with peripheral nociceptor hyperactivation [213,214]. Consequently, targeting NGF is therefore nowadays considered a promising target to modulate OA patients’ perceived pain, with several trials aiming to inhibit its deleterious effects during joint degeneration [215,216,217].
Although DMOADs have undoubted theoretical application potential, their clinical use in the context of OA still seems far off. This is due to major challenges in development, including regulatory guidelines, current assessment by conventional radiography, and lack of patient stratification in clinical trials. It is indeed complex to translate data obtained from animal trials with artificially induced pathology to human models. Furthermore, both American and European pharmaceutical regulatory authorities require a demonstrated reduction in pain symptoms and joint space thinning for the approval of a drug for OA [218]. However, radiographic measurements used to assess this parameter can be challenging to standardize and evaluate in large trials [219]. In addition, there is no direct correlation between joint morphological changes and OA progression or severity. The need for biomarkers to assess the progress of trials more objectively on DMOADs is therefore deeply felt.

4.3. Regenerative Therapies in OA

Regenerative therapies are gaining increasing popularity in the field of orthopedic medicine for the treatment of OA and joint pathologies in general. Among these, some promising options are viscosupplementation with HA and Platelet-Rich Plasma (PRP) therapies. These therapies provide innovative and less-invasive solutions to enhance joint functionality and alleviate pain, enabling patients to regain a better QoL.

4.3.1. Hyaluronic Acid

In OA, a relatively recent therapeutic approach is represented by HA use as viscosupplementation, either through injection or enteral supplementation. HA is a high-molecular-weight (MW) molecule (6.5 kDA to 20 MDa) naturally synthesized by the body, composed of alternately repeating D-glucuronic acid and N-acetylglucosamine units [220]. It is physiologically present in soft connective tissue, cartilage, and synovial fluid, where it serves as a lubricant and antioxidant [221]. Additionally, HA plays a crucial role as a modulator of cytokine release, influencing cell proliferation and migration and reducing MMP activity [222].
In the context of OA, the onset and progression are associated with the alteration of joint HA and its degradation through depolymerization. This process leads to the formation of lower MW forms of HA, causing changes in the composition and mechanical properties of synovial fluid, as well as a reduction in cartilage renewal. Such processes are mainly related to increased hyaluronidase activity and RONS levels [223,224]. Moreover, while high-MW HA has a protective effect on the join, low-MW forms of the molecule appear to be proinflammatory [225,226].
The exogenous supply of HA results is unable to completely overlap the loss of the endogenous one but is still able to partially restore the functionality of the synovial fluid, stimulating matrix production and promoting an anti-inflammatory effect through NF-κB and MAPK signaling pathways inhibition by reducing TNFα, IL-1β, and IL-6 levels in the joint [227,228]. Moreover, being TNFα a stimulator of NO and metalloproteinase synthesis, its reduction further preserves the joint from OA-mediated degradation [229].
The Food and Drug Administration already approved the intra-articular use of HA for the therapy of knee OA in 2001, and several applications have been developed since then [230]. Currently, the treatment algorithm recommended by the European Society for Clinical and Economic Aspects of Osteoporosis and Osteoarthritis (ESCEO) advocates the use of intra-articular HA for managing knee OA in patients who continue to experience pain despite the use of NSAIDs. Indeed, given the lack of unequivocal and objectively comparative data regarding the efficacy of corticosteroids and HA injections in relieving OA pain, the latter option offers a superior safety profile [181]. Indeed, given the lack of unequivocal and objectively comparative data regarding the efficacy of corticosteroids and HA injections in relieving OA pain, the latter option offers a superior safety profile [181].
International and national societies guidelines, however, vary, with some, like the EULAR, suggesting the consideration of HA, emphasizing its potentially longer-lasting effects, while others recommend its use just in the second instance, when other therapeutical approaches have proven to be ineffective, like ESCEO or American College of Rheumatology (ACR) [182].

4.3.2. Platelet-Rich Plasma

PRP is a concentrated autologous mixture of platelets, growth factors, and bioactive components. It is obtained through the centrifugation of whole blood and is subsequently reinjected into the same donor, aiming to exploit the regenerative properties of platelets for therapeutic benefits [231]. Once activated by thrombin or collagen, it is capable of releasing molecules, such as cytokines and growth factors like IL-1b and TNF-α capable of reducing inflammatory responses, inhibiting the NF-κB pathway while also promoting mesenchymal stem cell proliferation and matrix deposition and inhibiting metalloproteinase activity, stimulating tissue healing [232,233,234]. PRP has also been shown in both in vivo and in vitro studies to promote, dose-dependently, chondrocyte proliferation and degradation of the damaged ones by stimulating autophagy, with a cascade effect on the increased synthesis of proteoglycan and collagen type II [235].
In 2020, Belk and coworkers published a meta-analysis spanning 18 studies comparing the benefits of PRP intra-articular injection with those provided by HA use. According to their observation, the use of PRP provides results comparable or even superior to those provided by HA treatments [236]. Also, Chen and colleagues, more recently, published a meta-analysis comparing the results of different literature productions involving more than 2700 patients divided into PRP and HA groups [237]. Its result showed that reported Western Ontario and McMaster Universities Arthritis Index (WOMAC) pain marks were higher for PRP-treated subjects, with comparable safety profiles. It is intriguing to consider that recent evidence about the combined use of PRP and HA injection seems promising in knee OA, with functional improvements and pain reduction still after 12 months from the treatment [238].
Due to the high amount of existing PRP formulations and uncertainties about their pharmacodynamics, these treatments have a high grade of heterogeneity. This makes it quite challenging to compare clinical trial results. For these reasons, the main international guidelines discourage the clinical use of PRP [182].

4.4. Surgical Approaches

In advanced stages of OA, the depletion of cartilage, joint space, and synovial fluid reaches critical levels, resulting in debilitating pain, functional impairment, and disability. Conservative measures, therefore, become inadequate to address the severity of symptoms, necessitating more invasive interventions, such as surgery.
One such intervention is arthroplasty, a procedure designed to partially or entirely replace, remodel, or realign the compromised joint. This approach, and in particular, total joint arthroplasty, is highly effective and safe for treating knee and hip OA and is also being used for spine, shoulder, ankle, and elbow joints [239]. Surgery is less commonly employed for conditions affecting the hands, where the side effects outweigh the benefits. In such cases, joint protection and splinting offer temporary relief and may postpone the need for surgical intervention [240,241].
Another surgical approach to address joint issues involves corrective osteotomy or arthrodesis. Corrective osteotomy entails reshaping the bones forming the joint to rectify deformities and redistribute the load on less damaged areas of cartilage [242,243,244]. On the other hand, arthrodesis involves immobilizing a mobile or semimobile joint by aligning the component bones and securing them with screws, plates, or bone grafts [245,246]. Osteotomy can alleviate pain and postpone the need for other surgical interventions, offering short-term relief. It may be considered in cases of early joint malalignment without significant cartilage damage or limited joint destruction [239]. It may be considered in cases of early joint malalignment without significant cartilage damage or limited joint destruction [239]. Arthrodesis, being a more radical type of approach, is generally considered when alternative surgical options are impractical or ineffective, particularly in the presence of severe osteoarticular damage [247].
A less-invasive surgical option for treating OA is arthroscopy, a procedure involving the insertion of surgical tools and a micro-camera into the joint capsule. While arthroscopy is commonly used for joints such as the spine, elbow, wrist, and hand, it also extends to larger joints like the ankle and hip. This approach is regarded as conservative in OA treatment, aiming to delay or prevent the need for more invasive procedures while preserving cartilage; for this reason, it is particularly favored in athletic or younger individuals [248].
In cases of spine OA, avoiding fusion surgeries is crucial to mitigate well-known side effects related to its loss of flexibility [249,250]. For elbow OA, treatment options may include joint surface replacement or removal of osteophytes [251]. Conversely, managing wrist and hand OA may involve denervation to alleviate symptoms rather than direct therapeutic intervention [252]. In larger joints, arthroscopy may facilitate the realignment of bone structures or correction of unilateral deformities [253,254]. Although these approaches vary widely, their collective goal is to alleviate pain and enhance joint function in OA patients.
Arthroscopic procedures can involve flushing the joint with saline solution to remove loose particles like cartilage or tissue fibers from the joint fluid, thereby reducing inflammation. Alternatively, a debridement approach can be employed to smooth rough cartilage surfaces and eliminate loose cartilage fragments [255].
Regardless of their invasiveness, surgical procedures inherently carry risks, which tend to increase with the complexity of the intervention. Even though joint replacements have been performed since the 1960s, the procedure cannot be considered entirely safe [256,257]. In the United States alone, it is projected that joint replacement surgeries will reach 3 million by 2030. The most common type of joint replacement procedures, namely knee and hip, have a mortality rate of 1%, with almost 5% of patients experiencing post-surgery complications [258]. Even less-invasive arthroscopic procedures present some side effects, like nerve damage, infections, and clot formation [259,260,261,262,263].
Additionally, implants typically have a lifespan estimated at 15–25 years and necessitate replacement after this period, a concern particularly meaningful for younger patients [264].
Finally, two potential issues may emerge post-surgery. The first is periprosthetic osteolysis, arising from an inflammatory response to wear debris generated by friction between prosthetic components. This reaction activates osteoclasts, leading to periprosthetic bone loss [264,265]. The second potential problem is aseptic loosening, characterized by the loss of implant stability due to disruption of the bond between bone and prosthesis or between prosthesis and cement [266,267]. Aseptic loosening, together with infection, now represents the first cause of short-term failure of early rejection of total joint arthroplasty [268,269,270]. Then, if the prosthesis remains stable and functional, the surgical intervention is typically confined to revising the bearing surfaces and potentially incorporating bone grafting. Alternatively, replacing the entire prosthesis may be considered. Following corrective revision surgery to address the underlying cause of wear, osteolytic areas have the potential to regress and re-ossify [271].
Presently, strategies to mitigate complications in prosthetic components involve developing durable, wear-resistant materials that generate smaller debris, while numerous efforts are constantly directed at optimizing component design and positioning aids to reduce mechanical stresses and micro-movements [272,273]. Furthermore, post-surgical administration of anti-inflammatory or anti-osteoclastic medications like bisphosphonates can inhibit the inflammatory response and bone resorption [274,275].
In perspective, an emerging approach alternative to joint replacement surgery could be represented by the implantation of engineered tissue scaffolds designed to facilitate osteochondral regeneration [276,277]. Recently, an international consortium of research centers and health care providers reported preliminary evaluations demonstrating promising interactions between some of their crafted structures and the surrounding matrix, as well as their encouraging potential in promoting cartilage and bone tissue regeneration [278]. Nevertheless, the durability and safety profile of these artificial structures over extended periods will require further rigorous evaluations before considering human applications.

5. Natural Extracts

While NSAIDs remain the front-line therapy for OA management, their safety profile is hardly ideal, based on results from multiple randomized controlled trials (RCTs) and meta-analyses over the years [279,280,281,282]. Briefly, the safety profile of each NSAID varies with its pharmacologic action and individual responses to the drug. The AEs associated with long-term NSAID therapy are gastrointestinal-like lesions and ulcers, cardiovascular-like stroke, myocardial infarction, renal-like edema, and acute kidney disease [283,284,285]. As most of the at-risk population of OA is aging adults and often presents with comorbidities, the risk of AEs increases, and therefore, long-term NSAID regimes become difficult to manage [133].
Nevertheless, OA therapy is essentially the management of pain as the condition itself is irreversible. For this reason, while the use of NSAIDs is inevitable, the pitfalls associated with them could be addressed using bioactive compounds extracted from medicinal plants, herbs, and food sources, collectively termed nutraceuticals [286]. Nutraceuticals are generally considered to be safe because they are obtained from common food sources and represent incredible promise owing to their potent anti-inflammatory and antioxidant properties [287]. A recent Australian study found that 35% of OA patients used nutritional supplements concurrently with conventional analgesics [288]. Aghamohammadi et al. (2020) ascertained that the use of nutraceuticals improved pain scores and consequently led to better physical function in OA patients [289]. Polyphenols are particularly implicated as one of the most important bioactive compounds because of their ROS-scavenging ability and the suppression of proinflammatory pathways, such as MAPK and NF-κB [290,291,292]. These properties have been tested in both in vitro and in vivo OA disease models with a suitable degree of success [293,294,295,296].
Unsurprisingly, therefore, these points make a suitable case for the use of nutraceuticals and plant extracts as an alternative therapy to traditional pharmaceutical agents in OA. A summary of their effectiveness in clinical trials is presented in Table 2.
This section will discuss plant extracts of particular importance and their anti-inflammatory roles from the perspective of OA management, including curcumin from turmeric, bromelain extracted from the pineapple plant, boswellic acid extracted from the Indian frankincense tree, devil’s claw and aescin from horse chestnut, Matricaria chamomilla, Glycine soja, Zingiber, and quercetin.

5.1. Curcumin

Being a hydrophobic polyphenol, curcumin, or diferuloylmethane, bears many virtues with other plant-derived polyphenols, particularly being antibacterial, antioxidant, and anti-inflammatory [301]. Derived from the plant Curcuma longa, curcumin in the form of powdered turmeric is an essential part of Asian cuisine as a spice and has been valued in traditional Ayurvedic medicine as a remedy to dress wounds and burns and even as an eye ointment [302]. Curcumin has also been tested for its anti-cancer potential in cervical, breast, lung, and pancreatic cancer [303,304,305,306].
In the context of OA, curcumin exerts its action through various mechanisms, as evidenced by multiple studies over the years [307,308,309]. The storm of secretion of inflammatory mediators in OA causes joint destruction, and hence, this represents an important therapeutic target. While curcumin on its own cannot induce apoptosis of synovial fibroblasts within safe doses, Shakibaei and colleagues (2005) found that it can protect chondrocytes from IL-1β-mediated alterations in vitro, hence indirectly exerting its anti-apoptotic effects [310,311]. Curcumin has an established ROS-scavenging activity through its excellent electron-transfer capability; however, its ability to independently perform this in OA models has not yet been reliably demonstrated [312,313]. Chen et al. (2023) showed that a combination of catalase and curcumin prevented oxidative stress by inducing the expression of ROS-scavenging enzymes through the NRF2/HO-1 signaling pathway [314]. Several studies have also explored curcumin-loaded nanoparticles to circumvent the issue of cytotoxicity, and in such a study, Crivelli and colleagues used silk fibroin nanoparticles to encapsulate curcumin and celecoxib to improve the ROS-scavenging ability of curcumin in an in vitro OA model [315]. By inhibiting matrix MMPs, which are involved in the degradation of the extracellular matrix, curcumin also has an anti-catabolic and chondroprotective role [308]. Indeed, Zhang et al. (2016) used curcumin and curcumin-encapsulated nanoparticles, which reduced the RNA expression of MMP 1, 3, and 13, IL-1β, and TNF-α while promoting the expression of CREB-binding protein (CBP)/p300-interacting transactivator with glutamic acid and aspartic acid-tail 2 (Cited2) in human primary chondrocytes [316].
Curcumin’s chondroprotective potential has also been demonstrated in vivo, particularly in mouse models, where it has been shown to decrease disease progression [316]. A 2018 study ascertained curcumin’s therapeutic effect in osteoarthritis to be a result of enhanced autophagy through the Akt/mTOR pathway [317]. Despite its many benefits, curcumin suffers from a major pitfall of poor absorption and solubility, which limits its clinical use. Perhaps this is the reason why only a few RCTs have been conducted over the years to test curcumin formulations in human subjects. A systematic review of RCTs using curcumin for osteoarthritis, conducted by Bannuru and colleagues (2018), revealed that the limited number of such trials was further compromised by their generally low study quality [318]. In such an RCT, it was ascertained that curcumin supplement was significantly better than placebo in improving pain scores for a period of three months in a cohort of OA patients [319]. However, in a comparative evaluation with ibuprofen, while curcumin was shown to have a better gastrointestinal tolerance, the pain scores were similar [297]. A novel encapsulation method used by Yabas and colleagues in their 2021 study claims to improve disease prognosis in a mouse model, addressing the issue of bioavailability [320].

5.2. Bromelain

Unlike curcumin, bromelain is not a polyphenol but a proteolytic enzyme obtained as an aqueous extract from the pineapple plant Ananas comosus [321]. However, like polyphenols, plant proteases also have beneficial antioxidant, anti-microbial, anti-hypertensive, and ACE-inhibitory properties [322,323,324,325]. Bromelain, which is perhaps the most clinically and industrially important protease, also has an array of additional attributes, like fibrinolytic, analgesic, and immunomodulatory activities [326,327,328].
The enzyme was first used as an anti-inflammatory compound in 1964, directed against patients with rheumatoid and osteoarthritis [329]. In a 2006 pilot study, bromelain on its own was determined to be inefficacious in improving WOMAC scores in moderate to severe OA [330]. When compared with an NSAID like diclofenac, a similar result was achieved by Kasemsuk et al. [331]. Later clinical studies incorporated other extracts like curcumin and Harpagophytum porocumbens (HP) with bromelain with suitable pain scores [332]. Italiano and colleagues (2019) assessed the QoL in osteoarthritis patients to find that food supplements containing bromelain and Boswellia serrata improved QoL scores [333]. However, in another clinical trial that combined bromelain with enzymes like trypsin and rutoside trihydrate, the researchers found no significant difference in the pain scores when compared with diclofenac [334].
The anti-inflammatory actions of bromelain from the context of osteoarthritis were explained by a 2021 study, in which the enzyme extract is shown to suppress glycosaminoglycan (GAG) and hence counteract the degradative effects of IL-1β and MMPs [335]. Brochard et al. (2021) demonstrated that bromelain has a similar effect on lipopolysaccharide-induced inflammation, albeit in a combinatorial form with curcumin and Boswellia serrata [336]. Similar effects were observed by Quarta and colleagues (2022) in an in vitro model of inflammation [337]. These mixed results suggest that further studies are needed to ascertain the mechanisms of action of bromelain, particularly on its conditions of use.

5.3. Boswellia serrata

Boswellia serrata is a tree that grows in arid, mountainous regions of India, North Africa, and the Middle East and yields a rubbery oleoresin from its bark, which is called Boswellia Gum Resin Extract (BSE) [338]. For simplicity, in this review, the names of the tree and the extract are used interchangeably. The Boswellia extract is chemically composed of terpenes and the associated terpene acids, namely β-boswellic acid, acetyl-β-boswellic acid, 3-O-acetyl-11-keto-β-boswellic acid, and acetyl-11-keto-β-boswellic acid [339].
Boswellia extract has shown immense potential, primarily due to its anti-inflammatory role, in furthering alternative therapy for diseases like inflammatory bowel disease, asthma, peritumoral brain edema, and osteoarthritis [340,341,342,343]. Kimmatkar and colleagues, in their 2003 RCT, found that when compared with placebo controls, Boswellia extract was significant in decreasing knee pain and swelling [344]. This was also reflected in later studies, either in novel formulations or in combination with other extracts [345,346,347,348]. Neither of these studies, however, compared the efficacy of the extract with any common NSAID; hence, the results cannot be considered conclusive.
Kulkarni et al. showed that Boswellia extract encapsulated in solid lipid nanoparticles reduced proinflammatory cytokines when compared with a non-encapsulated extract [349]. The researchers also found that even though Boswellia extract did not significantly improve pain scores, subjects did not need to resort to a common NSAID during the trial. Similar conclusions were drawn by Henrotin and colleagues in a separate study, specifically from the context of hand osteoarthritis, where NSAID use decreased by 64% over a period of three months [350].
Besides clinical trials, several in vitro and in vivo studies suggest that Boswellia extracts, and more specifically, boswellic acids, inhibit cytokines and enzymes associated with inflammation like COX-1 and Cathepsin-G (catG) [351,352,353]. Even though the concentrations of the bioactive components in Boswellia extract necessary to perform the inhibitory actions have been found to be sub-par, a prevailing theory is that their accumulation might be in lipophilic extra- or intracellular components [339,354].

5.4. Harpagophytum procumbens

The complex extract from HP, or devil’s claw, contains phenolic acids and glycosides, triterpenes, phytosterols, iridoid glucosides like harpagoside, and various flavonoids. Various in vitro studies have testified to the anti-inflammatory and analgesic characteristics of the components of this extract by counteracting the production of cytokines [355,356,357]. Of note is a 2017 study, which evidenced that the major active component of HP extract, namely harpagoside, suppresses IL-6 production in an in vitro model of OA chondrocytes [358]. The same group had earlier postulated that the anti-inflammatory activity of harpagoside is due to its inhibition of the NF-κB pathway [359]. Mariano and colleagues, however, argued that the chondroprotection in in vitro models is due to all the various individual components in the HP extract [360].
More recently, Farpour et al. studied the differences in WOMAC and Visual Analog Scale (VAS) scores of cohorts taking HP tablets and a common NSAID. There were no discernible differences in the pain scores, and the researchers did not compare the long-term gastrointestinal impacts among the two cohorts [361]. A larger, multicenter study conducted in Poland investigated the supplemental efficacy of a multicomponent gel containing HP extract with conventional OA therapy, with suitable pain scores and improved functional capacity [362]. Despite promising results, the long-term safety of HP extract supplements has not been determined and warrants further investigation [363].

5.5. Aescin

Like Boswellia serrata extract, aescin is also a terpenoid composed of two forms, α-escin and β-escin [364]. Aescin itself is extracted from the fruit and the seeds of Aesculus hippocastanum, commonly known as horse chestnut. Aescin has been shown to be effective against edema, inflammation, and various tumors [365,366,367,368]. Aescin exerts its anti-inflammatory role by promoting the uptake of the Glucocorticoid receptor through the cell membrane, which then inhibits the release of TNF-α, IL-1β, and IL-6 [369]. Much like HP extract, it can also act through the inhibition of NF-κB [370].
In a large prospective study conducted in Russia and Ukraine, researchers found that replacing the NSAID diclofenac with a topical cream formulation of aescin helped alleviate pain in OA patients of various clinical stages. However, this study did not have control groups, and the pain score questionnaires were obtained from the physicians rather than the patients themselves [371]. In a later trial, Zeng and colleagues investigated the impact of sodium aescinate, which is a commercial form of aescin, on OA patients while comparing it with conventional NSAID therapy. The researchers found that there was a statistically significant difference in pain alleviation and joint functioning in the intervention group as compared to the control [372]. A similar effect was observed earlier by Maghsoudi et al., albeit in an in vitro synoviocyte model [373].
While studies specifically directed against OA are sparse, the gastroprotective ability of aescin demonstrated in other cellular and patient-specific models is a lucrative prospect for its long-term use [374,375].

5.6. Matricaria chamomilla

Matricaria chamomilla, usually called chamomile, is a well-known medicinal plant of the Asteraceae family. Native to southern and eastern Europe and northern and western Asia, today it is widely distributed throughout the world [376,377]. The chemical compounds of this plant include apigenin, apigenin-7-O-glucoside (APG), caffeic acid, chlorogenic acid, luteolin, and luteolin-7-O-glucoside, terpene bisabolol farnesene, chamazulene, flavonoids (including apigenin, quercetin, patuletin, and luteolin), and coumarin [378,379,380,381,382].
Pharmacological investigations have reported several biological activities of M. chamomilla, and its possible use in various fields, including medicine, has been investigated. [383]. Several in vivo studies have demonstrated its therapeutic effect on a wide range of pathologies, including nervous diseases, diabetes, metabolic disorders, cardiovascular diseases, and allergies [384,385,386,387,388].
The plant has also been shown to relieve pain, heal wounds, and act as a protective agent for kidneys and liver, as well as gastrointestinal and reproductive systems [389,390,391]. Additionally, M. chamomile has anti-inflammatory and antioxidant activities and strong antiplatelet and anticarcinogenic properties. It can heal skin lesions and is beneficial for anxiety disorders [167,392,393,394,395,396,397].
In vitro, chamomile has been shown to inhibit free radical level formation following H2O2 treatments in human skin fibroblasts, as well as TNF-α production [398,399].
On the other hand, in vivo studies have also shown its capability to improve joint function and reduce knee and back pain [400]. The component that would appear to play a crucial role in chamomile’s anti-inflammatory effects is apigenin. Shoara et al. (2015) showed a significant beneficial effect of a traditional topical formulation of chamomile flower oil on analgesic use in patients with knee OA [401]. Additionally, chamomile oil showed some beneficial effects also on patients’ pain, stiffness, and physical activity. It has been observed that apigenin can have a strong inhibitory effect on prostaglandin E2 levels [402].
Finally, chamomile has been shown to interfere with the COX-2 pathway with a mechanism similar to those exerted by NSAIDs [403]. In addition to chamomile flavonoids and essential oils being able to penetrate the skin layers [404], chamomile has also shown anti-inflammatory and analgesic effects when applied topically [405].

5.7. Glycine soja

Glycine soja (GS), also known as wild soybean, is an annual climbing herb of the legume family (Fabaceae) [406]. It is native to East Asia, the Russian Far East, eastern China, the Korean peninsula, and Japan [407]. Considered the progenitor of cultivated soybean, GS has been used in China for more than 2000 years and is considered an excellent source of soy-derived drugs [408].
GS contains a wide range of compounds, including saponins and isoflavones (e.g., daidzein, 6-hydroxy-daidzein, daidzein glycosides, genistein, genistein glycosides, glycitein, and glycitein glycosides) [409]. Health benefits associated with soy polyphenols are attributed to phenolic acids, flavonoids, and anthocyanins [410]. Soy isoflavones are of particular interest in the pharmaceutical industry. GS shows various biologically relevant effects, improving blood lipid profile and reducing hepatic steatosis and adipocyte size in mice exposed to a high-fat diet [411].
Yun Mi Lee and collaborators studied both in vitro and in vivo the effects of GS leaves and stems (GSLSs) on OA, focusing on inflammation and ECM degradation [406]. In particular, the anti-inflammatory effects of GSLSs were investigated in SW1353 human chondrocytes stimulated with IL-1β, and significant reductions in the levels of the inflammatory mediators PGE2, IL-1β, IL-6, and TNF-α were shown following the treatments [412].
GSLSs also inhibit the expression of inflammatory cytokines and matrix metalloproteinases, with a protective role in collagen type II degradation, as observed in IL-1β-stimulated chondrocytes [406,413]. The chondro-protective role of GSLSs is due to the inhibition of NF-κB activation [414]. Collectively, GSLSs significantly reduce OA-associated joint pain, also lowering serum levels of proinflammatory mediators, cytokines, and matrix metalloproteinases. Thus, GSLSs represent a useful therapeutic candidate for OA.

5.8. Zingiber officinale Roscoe

Zingiber officinale Roscoe, commonly known as ginger, belongs to the Zingiberaceae family and has been used as a spice and herbal remedy for years. Its rhizome is a particularly rich source of bioactive substances used in Ayurvedic and Chinese medicine [415]. The pharmacological properties of ginger are related to numerous active phytocompounds belonging to the phenols and terpenes. The rhizomes of ginger plants comprise two different types of compounds. The first is the non-volatile oleoresin, the source of ginger’s pungent taste, and the second is the volatile essential oils [416]. Oleoresin comprises the main physiologically active substances of this spice, such as gingerols, shogaols, paradols, and zingerone [417]. One of the main components of ginger is 6-gingerol, which has anti-inflammatory and analgesic properties [418,419].
Preclinical studies have shown that the phytochemical compounds of ginger, including 6-shogaol, zingerone, and cedrol, are effective anti-rheumatic agents, as they alter signaling pathways involved in OA pathophysiology [420].
It has been shown that gingerols, shogaols, paradols, and other polyphenols present in ginger, together with sesquiterpenes, inhibit TNF-α, IL-1β, IL-2, IL-4, IL-6, IL-17, PGE 2, and COX-2 in human synoviocytes; this modulates the activation of NF-κB and the degradation of its inhibitor IkB-α [421,422,423,424,425,426]. Additionally, many studies highlight the antioxidant effects of ginger [427]. Its phenolic constituent, 6-gingerol, inhibited LPS-induced iNOS expression and the production of NO and other reactive nitrogen species in macrophages [428]. Therefore, gingerols and their derivatives may represent an alternative to NSAIDs without serious gastrointestinal or renal side effects. Naderi and coworkers concluded that ginger may be recommended as an appropriate supplement for patients with OA, as it is useful for reducing pain, stiffness, and inflammation in patients with OA [429].

5.9. Quercetin

Quercetin, a flavonoid widely distributed in fruits, herbs, and vegetables, has demonstrated a wide range of beneficial health properties. These include anti-inflammatory, immunomodulatory, and antioxidant effects, as well as contingent anti-arthritic and joint-protective properties in inflammatory joint diseases [300,430,431]. Recently, several studies have highlighted the therapeutic potential of quercetin in OA, effectively inhibiting inflammation and apoptosis of chondrocytes and thus preventing disease progression [432,433]. Quercetin has been identified as a potent senolytic drug capable of inducing apoptosis of senescent cells [434].
Understanding the molecular mechanisms involved in the action of quercetin is crucial to developing new targeted therapeutic strategies for the management of OA and maximizing its clinical potential in inflammatory joint diseases.
Quercetin has also been shown to inhibit endoplasmic reticulum stress-related cartilage degeneration, a key process in the pathogenesis of OA. This was demonstrated through the activation of the SIRT1/AMPK signaling pathway, which led to the prevention of OA progression in rat models [435]. Li and coworkers have also demonstrated in vivo the ability of quercetin to suppress the IRAK1/NLRP3 signaling pathway, reducing the levels of proinflammatory cytokines in OA [436]. Furthermore, quercetin demonstrates significant chondroprotective effects, reducing cartilage degradation and apoptosis of chondrocytes and promoting the synthesis of glycosaminoglycans to enhance the repair of damaged cartilage [432]. Wang and colleagues suggested that these effects are associated with the molecule’s ability to inhibit the p38 MAPK and ADAMTSs signaling pathway, thereby reducing relevant inflammatory factors, and promoting the expression of type II collagen to promote cartilage regeneration [437]. Finally, quercetin has been shown to inhibit MMP activity in OA [438].

6. Challenges in Natural Molecules Quality Control and Standardization

With the increasing demand for natural extracts and nutraceuticals for health-related applications, more attention is being paid to quality assurance and standardization of preparations. Despite the importance of the sector, also underlined by the World Health Organization’s Strategy on Traditional Medicine 2014–2023, discrepancies between regulatory bodies and manufacturers regarding the amount of quality testing required for dietary supplements represent a significant challenge in the industry [439]. Botanical extracts and mixtures pose unique challenges in detecting identification errors and contamination, both biological and chemical in nature. The lack of analytical methods and reference standards for the numerous bioactive ingredients present in dietary supplements is a significant scientific challenge. There is still no universal agreement on the acceptance of a single officially approved analysis method, as there are divergent opinions on who should be responsible for developing standards and analytical methods [440].
To ensure the safety of botanical dietary supplements, it is essential to implement stringent quality control and assurance measures throughout the entire production process. This includes sourcing botanical materials from reliable suppliers, with careful authentication of plant species through macroscopic and microscopic botanical examinations.
During the production of nutraceuticals, the use of solvents, additives, and purification techniques can be influenced by the presence of contaminants such as pesticides, herbicides, and heavy metals, which are known to cause serious adverse effects, including liver and kidney toxicity and even carcinogenicity [441,442]. Therefore, the safety of such products has become a priority for regulatory authorities. Although the use of pesticides is necessary to preserve the quality of medicinal herbs, it is crucial to adhere to the WHO guidelines regarding the presence of environmental contaminants in the final herbal products [443]. It is crucial to define and implement rigorous production procedures, ensuring standardization and quality control throughout all stages of the process [442].
After processing, botanical dietary supplements must be carefully analyzed to identify and remove any hazardous contaminants, such as pesticides, herbicides, and heavy metals, and undergo chromatographic tests to exclude the presence of unintended pharmaceutical contaminations. Chemical standardization, based on the concentration of active ingredients, and biological standardization, through in vitro and in vivo assays, are crucial to ensure the safety and reproducibility of botanical dietary supplements, providing consumers with products containing consistent levels of active ingredients and predictable pharmacological effects [440]. However, the uniform and coherent regulation of such products still poses a challenge, as global standards governing their production, sale, and marketing are lacking, highlighting the need for international consensus on how to define and regulate this category of products [444,445].

7. Future Research Directions

The beneficial effects of natural molecules are numerous, offering promising options for complementing established treatment approaches. As widely discussed in the former section of the article, many of these molecules show beneficial effects in addressing OA, targeting pathways implicated in joint degradation and underlying pathological mechanisms. Inflammation and oxidative stress, recognized as paramount elements in OA contexts, represent an important target to slow tissue degeneration and improve patients’ QoL [446]. The listed natural extracts, which represent only the most important among an incredibly wide range of molecules used in traditional medicine to counteract OA, converge to alleviate these pathological elements through pleiotropic mechanisms [447,448]. Their integration in clinical and classical therapeutical regimes could therefore reduce the dosage and frequency of use of molecules such as NSAIDs, as well as their AEs.
Regarding natural molecules, it is critical to recognize that merely identifying those with significant beneficial effects for a particular disorder is not sufficient. Equally important is the evaluation of optimal dosage, formulation, and absorption efficiency in the body [449]. This factor could partially account for the significant variability observed in clinical trial outcomes or, in some cases, the absence of conclusive results altogether. The lack of objective methods to accurately determine disease status and measure treatment-related improvements, excluding patients’ reported outcomes, contributes to the problem. Two further elements could provide a strong improvement in OA management and treatment. A better comprehension of OA early-stage processes, considering the heterogeneity of its manifestations and progression in the population, is required. It is also important to abandon “one-size-fits-all” therapeutical approaches to others that are more tailored to patients.
Tailoring therapies to patients may mean modulating the number of drugs to be used and the number of daily administrations. Reaching the innermost layers of the joint may present difficulties, however [450]. So, the use of a targeted and slow-release pharmacological approach may be considered. Formulations such as microemulsions, liposomes, sequessomes, solid lipid nanoparticles, or nanostructured lipid carriers have been studied in recent years with great interest. To date, however, only one phase II clinical trial based on the use of diclofenac in the form of nanoemulsion cream results [451].
Similarly promising but extremely cutting-edge could be the use of plant extracellular vesicles (PEVs) in the therapy of OA. In contrast to their animal counterparts, mammalian cell-derived extracellular vesicles (MEVs), and synthetic carriers, PEVs have the advantage of being non-immunogenic. These molecules can be isolated through numerous methods from a variety of edible plants, retaining many molecules with potent biological activities such as nucleic acids, lipids, and metabolites within them [452]. The anti-inflammatory and immunomodulatory potential demonstrated is certainly attractive and is suitable for use in OA therapy. However, it will still take a long time before safety studies open them up for human use and for the technology to overcome the many bioproduction and plant bioengineering challenges that currently limit large-scale production.

8. Conclusions

OA is a progressive condition associated with the destruction of the joint cartilage, and no resolutive therapies are clinically available nowadays. The main problem, however, is represented by the disability following the joint loss of function and the increasing and constant pain deriving from the process. The use of painkillers has a limited and temporary effect, acting just on OA symptoms and not on the causes generating the suffering. In the last decades, several approaches have been developed to reduce the degeneration process of the cartilage as well as the pain by acting directly inside the joint when directly injected into it. Also, in this case, the benefits appear limited, and clinical trials have not been able to prove their efficacy and safety up until now. In such a context, several natural molecules have been used for a very long time for their beneficial properties as anti-inflammatories and antioxidants.
OA is a progressive condition associated with several causative factors, destroying joint cartilage. Therapeutic strategies are directed against the palliative management of the disease alone, and no definitive cure exists. As the disability followed by the joint loss of function leads to severe pain, painkillers are routinely prescribed and have a limited and temporary impact while causing several side effects. Moreover, several other approaches have also been adapted, for instance, the injection of strengthening factors like HA or PRP, with limited clinical success. Natural plant extracts and nutraceuticals have been hailed as potent alternative therapies for OA because of their intrinsic anti-inflammatory and antioxidant properties. Recent evidence has shown them to be effective at alleviating pain through the inhibition of proinflammatory pathways like the NF-κB one. Moreover, they also offer a direct effect in promoting cartilage protection by mitigating MMP activity and promotion mechanisms of autophagy, favoring the removal of damaged chondrons. At the state of the art, more focused studies and trials are, however, required to consolidate evidence in their favor and in perspective for recommending clinical guidelines for their use.

Funding

This research was funded by (i) the Italian Ministry of Research, thanks to the PRIN 2022 project (MUR) prot. 2022E7FZEJ “The double face of hypoxia in health and disease”, assigned to Rita Paroni, Department of Health Sciences, University of Milan, Milan, Italy, and to the innovative Ph.D. position made available to AM; and (ii) the Apulia region, thanks to the post-doc position of the POC PUGLIA FESR ESF 2014/2020 entitled “PaRTiRe—Parkinson’s research” through the “Riparti project” made available to M.G.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

All the authors would like to express their gratitude to Antonio Danieli, whose work contributes daily to the functioning of our laboratory. M.G. also wants to express his gratitude to Luca Laudisa, Virginia Manca and Alessandra Perrone, who, with professionalism and much patience, supported him during the writing process.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Di Renzo, L.; Gualtieri, P.; De Lorenzo, A. Diet, Nutrition and Chronic Degenerative Diseases. Nutrients 2021, 13, 1372. [Google Scholar] [CrossRef] [PubMed]
  2. Steinmetz, J.D.; Culbreth, G.T.; Haile, L.M.; Rafferty, Q.; Lo, J.; Fukutaki, K.G.; Cruz, J.A.; Smith, A.E.; Vollset, S.E.; Brooks, P.M.; et al. Global, Regional, and National Burden of Osteoarthritis, 1990–2020 and Projections to 2050: A Systematic Analysis for the Global Burden of Disease Study 2021. Lancet Rheumatol. 2023, 5, e508–e522. [Google Scholar] [CrossRef] [PubMed]
  3. Lin, X.; Xu, Y.; Pan, X.; Xu, J.; Ding, Y.; Sun, X.; Song, X.; Ren, Y.; Shan, P.-F. Global, Regional, and National Burden and Trend of Diabetes in 195 Countries and Territories: An Analysis from 1990 to 2025. Sci. Rep. 2020, 10, 14790. [Google Scholar] [CrossRef] [PubMed]
  4. Ou, Z.; Pan, J.; Tang, S.; Duan, D.; Yu, D.; Nong, H.; Wang, Z. Global Trends in the Incidence, Prevalence, and Years Lived with Disability of Parkinson’s Disease in 204 Countries/Territories From 1990 to 2019. Front. Public Health 2021, 9, 776847. [Google Scholar] [CrossRef] [PubMed]
  5. Greco, M.; Munir, A.; Musarò, D.; Coppola, C.; Maffia, M. Restoring Autophagic Function: A Case for Type 2 Diabetes Mellitus Drug Repurposing in Parkinson’s Disease. Front. Neurosci. 2023, 17, 1244022. [Google Scholar] [CrossRef] [PubMed]
  6. 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] [PubMed]
  7. Abramoff, B.; Caldera, F.E. Osteoarthritis: Pathology, Diagnosis, and Treatment Options. Med. Clin. N. Am. 2020, 104, 293–311. [Google Scholar] [CrossRef] [PubMed]
  8. Tuckermann, J.; Adams, R.H. The Endothelium–Bone Axis in Development, Homeostasis and Bone and Joint Disease. Nat. Rev. Rheumatol. 2021, 17, 608–620. [Google Scholar] [CrossRef] [PubMed]
  9. Tonutti, A.; Granata, V.; Marrella, V.; Sobacchi, C.; Ragusa, R.; Sconza, C.; Rani, N.; Di Matteo, B.; Ceribelli, A. The Role of WNT and IL-1 Signaling in Osteoarthritis: Therapeutic Implications for Platelet-Rich Plasma Therapy. Front. Aging 2023, 4, 1201019. [Google Scholar] [CrossRef]
  10. Taruc-Uy, R.L.; Lynch, S.A. Diagnosis and Treatment of Osteoarthritis. Prim. Care Clin. Off. Pract. 2013, 40, 821–836. [Google Scholar] [CrossRef]
  11. GBD 2019: Global Burden of 369 Diseases and Injuries in 204 Countries and Territories, 1990–2019: A Systematic Analysis for the Global Burden of Disease Study 2019. Available online: https://vizhub.healthdata.org/gbd-results/ (accessed on 7 February 2024).
  12. Shane Anderson, A.; Loeser, R.F. Why Is Osteoarthritis an Age-Related Disease? Best. Pract. Res. Clin. Rheumatol. 2010, 24, 15–26. [Google Scholar] [CrossRef] [PubMed]
  13. Hussain, S.; Wang, Y.; Giles, G.G.; Graves, S.; Wluka, A.E.; Cicuttini, F.M. Female Reproductive and Hormonal Factors and Incidence of Primary Total Knee Arthroplasty Due to Osteoarthritis. Osteoarthr. Cartil. 2018, 26, S206. [Google Scholar] [CrossRef]
  14. Tang, J.; Liu, T.; Wen, X.; Zhou, Z.; Yan, J.; Gao, J.; Zuo, J. Estrogen-Related Receptors: Novel Potential Regulators of Osteoarthritis Pathogenesis. Mol. Med. 2021, 27, 5. [Google Scholar] [CrossRef]
  15. Martín-Millán, M.; Castañeda, S. Estrogens, Osteoarthritis and Inflammation. Jt. Bone Spine 2013, 80, 368–373. [Google Scholar] [CrossRef]
  16. Khosla, S.; Oursler, M.J.; Monroe, D.G. Estrogen and the Skeleton. Trends Endocrinol. Metab. 2012, 23, 576–581. [Google Scholar] [CrossRef] [PubMed]
  17. Reese, M.E.; Casey, E. Hormonal Influence on the Neuromusculoskeletal System in Pregnancy. In Musculoskeletal Health in Pregnancy and Postpartum; Springer International Publishing: Cham, Switzerland, 2015; pp. 19–39. [Google Scholar]
  18. Bliddal, M.; Pottegård, A.; Kirkegaard, H.; Olsen, J.; Jørgensen, J.S.; Sørensen, T.I.A.; Dreyer, L.; Nohr, E.A. Association of Pre-Pregnancy Body Mass Index, Pregnancy-Related Weight Changes, and Parity with the Risk of Developing Degenerative Musculoskeletal Conditions. Arthritis Rheumatol. 2016, 68, 1156–1164. [Google Scholar] [CrossRef]
  19. Mitani, Y. Gender-Related Differences in Lower Limb Alignment, Range of Joint Motion, and the Incidence of Sports Injuries in Japanese University Athletes. J. Phys. Ther. Sci. 2017, 29, 12–15. [Google Scholar] [CrossRef]
  20. Grainger, A.J.; Resnik, C.S. Arthritis. In Musculoskeletal Diseases 2021–2024: Diagnostic Imaging; Hodler, J., Kubik-Huch, R.A., von Schulthess, G.K., Eds.; Springer: Cham, Switzerland, 2021; pp. 149–168. [Google Scholar]
  21. Felson, D.T. Identifying Different Osteoarthritis Phenotypes through Epidemiology. Osteoarthr. Cartil. 2010, 18, 601–604. [Google Scholar] [CrossRef] [PubMed]
  22. Xia, B.; Chen, D.; Zhang, J.; Hu, S.; Jin, H.; Tong, P. Osteoarthritis Pathogenesis: A Review of Molecular Mechanisms. Calcif. Tissue Int. 2014, 95, 495–505. [Google Scholar] [CrossRef]
  23. Maniar, K.H.; Jones, I.A.; Gopalakrishna, R.; Vangsness, C.T. Lowering Side Effects of NSAID Usage in Osteoarthritis: Recent Attempts at Minimizing Dosage. Expert. Opin. Pharmacother. 2018, 19, 93–102. [Google Scholar] [CrossRef]
  24. van Doormaal, M.C.M.; Meerhoff, G.A.; Vliet Vlieland, T.P.M.; Peter, W.F. A Clinical Practice Guideline for Physical Therapy in Patients with Hip or Knee Osteoarthritis. Musculoskelet. Care 2020, 18, 575–595. [Google Scholar] [CrossRef] [PubMed]
  25. Liddle, A.D.; Pegg, E.C.; Pandit, H. Knee Replacement for Osteoarthritis. Maturitas 2013, 75, 131–136. [Google Scholar] [CrossRef] [PubMed]
  26. Lohmander, L.S.; Peltonen, M.; Andersson-Assarsson, J.C.; Maglio, C.; Sjöholm, K.; Taube, M.; Jacobson, P.; Svensson, P.A.; Carlsson, L.M.S.; Ahlin, S. Bariatric Surgery, Osteoarthritis and Arthroplasty of the Hip and Knee in Swedish Obese Subjects—Up to 31 Years Follow-up of a Controlled Intervention Study. Osteoarthr. Cartil. 2023, 31, 636–646. [Google Scholar] [CrossRef] [PubMed]
  27. Grigore, A.; Vulturescu, V. Natural Approach in Osteoarthritis Therapy. Recent Adv. Inflamm. Allergy Drug Discov. 2022, 16, 26–31. [Google Scholar] [CrossRef] [PubMed]
  28. Lee, Y.T.; Yunus, M.H.M.; Ugusman, A.; Yazid, M.D. Natural Compounds Affecting Inflammatory Pathways of Osteoarthritis. Antioxidants 2022, 11, 1722. [Google Scholar] [CrossRef] [PubMed]
  29. Hunter, D.J.; McDougall, J.J.; Keefe, F.J. The Symptoms of Osteoarthritis and the Genesis of Pain. Rheum. Dis. Clin. N. Am. 2008, 34, 623–643. [Google Scholar] [CrossRef] [PubMed]
  30. Fu, K.; Robbins, S.R.; McDougall, J.J. Osteoarthritis: The Genesis of Pain. Rheumatology 2018, 57, iv43–iv50. [Google Scholar] [CrossRef]
  31. Association, C.O. Diagnosis and Treatment of Osteoarthritis. Orthop. Surg. 2010, 2, 1–6. [Google Scholar] [CrossRef]
  32. Hanada, M.; Takahashi, M.; Furuhashi, H.; Koyama, H.; Matsuyama, Y. Elevated Erythrocyte Sedimentation Rate and High-Sensitivity C-Reactive Protein in Osteoarthritis of the Knee: Relationship with Clinical Findings and Radiographic Severity. Ann. Clin. Biochem. Int. J. Lab. Med. 2016, 53, 548–553. [Google Scholar] [CrossRef] [PubMed]
  33. Walker, C.; Faustino, A.; Lanas, A. Monitoring Complete Blood Counts and Haemoglobin Levels in Osteoarthritis Patients: Results from a European Survey Investigating Primary Care Physician Behaviours and Understanding. Open Rheumatol. J. 2014, 8, 110–115. [Google Scholar] [CrossRef]
  34. Mekic, M.; Hadzigrahic, E. Anti-Cyclic Citrullinated Peptide Antibody as a Predictor of Rheumathoid Arthritis Complications. Med. Arch. 2020, 74, 183. [Google Scholar] [CrossRef]
  35. Mohammed, A.; Alshamarri, T.; Adeyeye, T.; Lazariu, V.; McNutt, L.-A.; Carpenter, D.O. A Comparison of Risk Factors for Osteo- and Rheumatoid Arthritis Using NHANES Data. Prev. Med. Rep. 2020, 20, 101242. [Google Scholar] [CrossRef] [PubMed]
  36. De Rycke, L. Rheumatoid Factor and Anticitrullinated Protein Antibodies in Rheumatoid Arthritis: Diagnostic Value, Associations with Radiological Progression Rate, and Extra-Articular Manifestations. Ann. Rheum. Dis. 2004, 63, 1587–1593. [Google Scholar] [CrossRef]
  37. Katz, J.N.; Arant, K.R.; Loeser, R.F. Diagnosis and Treatment of Hip and Knee Osteoarthritis. JAMA 2021, 325, 568. [Google Scholar] [CrossRef] [PubMed]
  38. Zhang, Z.; Huang, C.; Jiang, Q.; Zheng, Y.; Liu, Y.; Liu, S.; Chen, Y.; Mei, Y.; Ding, C.; Chen, M.; et al. Guidelines for the Diagnosis and Treatment of Osteoarthritis in China (2019 Edition). Ann. Transl. Med. 2020, 8, 1213. [Google Scholar] [CrossRef]
  39. Sen, R.; Hurley, J.A. Osteoarthritis; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  40. Yokose, C.; Chen, M.; Berhanu, A.; Pillinger, M.H.; Krasnokutsky, S. Gout and Osteoarthritis: Associations, Pathophysiology, and Therapeutic Implications. Curr. Rheumatol. Rep. 2016, 18, 65. [Google Scholar] [CrossRef] [PubMed]
  41. Oliviero, F.; Bindoli, S.; Scanu, A.; Feist, E.; Doria, A.; Galozzi, P.; Sfriso, P. Autoinflammatory Mechanisms in Crystal-Induced Arthritis. Front. Med. 2020, 7, 166. [Google Scholar] [CrossRef]
  42. Ivory, D.; Velázquez, C.R. The Forgotten Crystal Arthritis: Calcium Pyrophosphate Deposition. Mo. Med. 2012, 109, 64–68. [Google Scholar]
  43. Haartmans, M.J.J.; Emanuel, K.S.; Tuijthof, G.J.M.; Heeren, R.M.A.; Emans, P.J.; Cillero-Pastor, B. Mass Spectrometry-Based Biomarkers for Knee Osteoarthritis: A Systematic Review. Expert. Rev. Proteom. 2021, 18, 693–706. [Google Scholar] [CrossRef]
  44. Felekkis, K.; Pieri, M.; Papaneophytou, C. Exploring the Feasibility of Circulating MiRNAs as Diagnostic and Prognostic Biomarkers in Osteoarthritis: Challenges and Opportunities. Int. J. Mol. Sci. 2023, 24, 13144. [Google Scholar] [CrossRef]
  45. Kraus, V.B.; Karsdal, M.A. Osteoarthritis: Current Molecular Biomarkers and the Way Forward. Calcif. Tissue Int. 2021, 109, 329–338. [Google Scholar] [CrossRef] [PubMed]
  46. Rocha, F.A.C.; Ali, S.A. Soluble Biomarkers in Osteoarthritis in 2022: Year in Review. Osteoarthr. Cartil. 2023, 31, 167–176. [Google Scholar] [CrossRef] [PubMed]
  47. Nagy, E.; Nagy-Finna, C.; Popoviciu, H.-V.; Kovács, B. Soluble Biomarkers of Osteoporosis and Osteoarthritis, from Pathway Mapping to Clinical Trials: An Update. Clin. Interv. Aging 2020, 15, 501–518. [Google Scholar] [CrossRef]
  48. Bauer, D.C.; Hunter, D.J.; Abramson, S.B.; Attur, M.; Corr, M.; Felson, D.; Heinegård, D.; Jordan, J.M.; Kepler, T.B.; Lane, N.E.; et al. Classification of Osteoarthritis Biomarkers: A Proposed Approach. Osteoarthr. Cartil. 2006, 14, 723–727. [Google Scholar] [CrossRef]
  49. Runhaar, J.; Sanchez, C.; Taralla, S.; Henrotin, Y.; Bierma-Zeinstra, S.M.A. Fibulin-3 Fragments Are Prognostic Biomarkers of Osteoarthritis Incidence in Overweight and Obese Women. Osteoarthr. Cartil. 2016, 24, 672–678. [Google Scholar] [CrossRef]
  50. Henrotin, Y.; Gharbi, M.; Mazzucchelli, G.; Dubuc, J.; De Pauw, E.; Deberg, M. Fibulin 3 Peptides Fib3-1 and Fib3-2 Are Potential Biomarkers of Osteoarthritis. Arthritis Rheum. 2012, 64, 2260–2267. [Google Scholar] [CrossRef] [PubMed]
  51. Larsson, S.; Lohmander, L.S.; Struglics, A. Biological Variation of Human Aggrecan ARGS Neoepitope in Synovial Fluid and Serum in Early-Stage Knee Osteoarthritis and after Knee Injury. Osteoarthr. Cartil. Open 2022, 4, 100307. [Google Scholar] [CrossRef]
  52. Verma, P.; Dalal, K. Serum Cartilage Oligomeric Matrix Protein (COMP) in Knee Osteoarthritis: A Novel Diagnostic and Prognostic Biomarker. J. Orthop. Res. 2013, 31, 999–1006. [Google Scholar] [CrossRef]
  53. Tootsi, K.; Kals, J.; Zilmer, M.; Paapstel, K.; Ottas, A.; Märtson, A. Medium- and Long-chain Acylcarnitines Are Associated with Osteoarthritis Severity and Arterial Stiffness in End-stage Osteoarthritis Patients: A Case-control Study. Int. J. Rheum. Dis. 2018, 21, 1211–1218. [Google Scholar] [CrossRef]
  54. Park, Y.M.; Kim, S.J.; Lee, K.J.; Yang, S.S.; Min, B.-H.; Yoon, H.C. Detection of CTX-II in Serum and Urine to Diagnose Osteoarthritis by Using a Fluoro-Microbeads Guiding Chip. Biosens. Bioelectron. 2015, 67, 192–199. [Google Scholar] [CrossRef]
  55. Sasaki, E.; Yamamoto, H.; Asari, T.; Matsuta, R.; Ota, S.; Kimura, Y.; Sasaki, S.; Ishibashi, K.; Yamamoto, Y.; Kami, K.; et al. Metabolomics with Severity of Radiographic Knee Osteoarthritis and Early Phase Synovitis in Middle-Aged Women from the Iwaki Health Promotion Project: A Cross-Sectional Study. Arthritis Res. Ther. 2022, 24, 145. [Google Scholar] [CrossRef] [PubMed]
  56. Cuéllar, V.G.; Cuéllar, J.M.; Kirsch, T.; Strauss, E.J. Correlation of Synovial Fluid Biomarkers with Cartilage Pathology and Associated Outcomes in Knee Arthroscopy. Arthrosc. J. Arthrosc. Relat. Surg. 2016, 32, 475–485. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, Z.-W.; Chen, L.; Hao, X.-R.; Qu, Z.-A.; Huang, S.-B.; Ma, X.-J.; Wang, J.-C.; Wang, W.-M. Elevated Levels of Interleukin-1β, Interleukin-6, Tumor Necrosis Factor-α and Vascular Endothelial Growth Factor in Patients with Knee Articular Cartilage Injury. World J. Clin. Cases 2019, 7, 1262–1269. [Google Scholar] [CrossRef] [PubMed]
  58. Guo, X.; Wei, S.; Xu, F.; Cai, X.; Wang, H.; Ding, R. MicroRNA-532-5p Is Implicated in the Regulation of Osteoporosis by Forkhead Box O1 and Osteoblast Differentiation. BMC Musculoskelet. Disord. 2020, 21, 296. [Google Scholar] [CrossRef]
  59. Stanczyk, J.; Pedrioli, D.M.L.; Brentano, F.; Sanchez-Pernaute, O.; Kolling, C.; Gay, R.E.; Detmar, M.; Gay, S.; Kyburz, D. Altered Expression of MicroRNA in Synovial Fibroblasts and Synovial Tissue in Rheumatoid Arthritis. Arthritis Rheum. 2008, 58, 1001–1009. [Google Scholar] [CrossRef] [PubMed]
  60. Pertusa, C.; Tarín, J.J.; Cano, A.; García-Pérez, M.Á.; Mifsut, D. Serum MicroRNAs in Osteoporotic Fracture and Osteoarthritis: A Genetic and Functional Study. Sci. Rep. 2021, 11, 19372. [Google Scholar] [CrossRef] [PubMed]
  61. Kolhe, R.; Hunter, M.; Liu, S.; Jadeja, R.N.; Pundkar, C.; Mondal, A.K.; Mendhe, B.; Drewry, M.; Rojiani, M.V.; Liu, Y.; et al. Gender-Specific Differential Expression of Exosomal MiRNA in Synovial Fluid of Patients with Osteoarthritis. Sci. Rep. 2017, 7, 2029. [Google Scholar] [CrossRef]
  62. Li, Q.; Amano, K.; Link, T.M.; Ma, C.B. Advanced Imaging in Osteoarthritis. Sports Health A Multidiscip. Approach 2016, 8, 418–428. [Google Scholar] [CrossRef] [PubMed]
  63. Sanchez-Lopez, E.; Coras, R.; Torres, A.; Lane, N.E.; Guma, M. Synovial Inflammation in Osteoarthritis Progression. Nat. Rev. Rheumatol. 2022, 18, 258–275. [Google Scholar] [CrossRef]
  64. Kaeley, G.S.; Bakewell, C.; Deodhar, A. The Importance of Ultrasound in Identifying and Differentiating Patients with Early Inflammatory Arthritis: A Narrative Review. Arthritis Res. Ther. 2020, 22, 1. [Google Scholar] [CrossRef]
  65. Kellgren, J.H.; Lawrence, J.S. Radiological Assessment of Osteo-Arthrosis. Ann. Rheum. Dis. 1957, 16, 494–502. [Google Scholar] [CrossRef] [PubMed]
  66. Sangha, O. Epidemiology of Rheumatic Diseases. Rheumatology 2000, 39, 3–12. [Google Scholar] [CrossRef] [PubMed]
  67. Yoon, J.S.; Yon, C.-J.; Lee, D.; Lee, J.J.; Kang, C.H.; Kang, S.-B.; Lee, N.-K.; Chang, C.B. Assessment of a Novel Deep Learning-Based Software Developed for Automatic Feature Extraction and Grading of Radiographic Knee Osteoarthritis. BMC Musculoskelet. Disord. 2023, 24, 869. [Google Scholar] [CrossRef] [PubMed]
  68. Antony, J.; McGuinness, K.; Moran, K.; O’Connor, N.E. Automatic Detection of Knee Joints and Quantification of Knee Osteoarthritis Severity Using Convolutional Neural Networks. In Machine Learning and Data Mining in Pattern Recognition. MLDM 2017. Lecture Notes in Computer Science; Perner, P., Ed.; Springer: Cham, Switzerland, 2017; pp. 376–390. ISBN 978-3-319-62415-0. [Google Scholar]
  69. Cueva, J.H.; Castillo, D.; Espinós-Morató, H.; Durán, D.; Díaz, P.; Lakshminarayanan, V. Detection and Classification of Knee Osteoarthritis. Diagnostics 2022, 12, 2362. [Google Scholar] [CrossRef] [PubMed]
  70. Kohn, M.D.; Sassoon, A.A.; Fernando, N.D. Classifications in Brief: Kellgren-Lawrence Classification of Osteoarthritis. Clin. Orthop. Relat. Res. 2016, 474, 1886–1893. [Google Scholar] [CrossRef] [PubMed]
  71. Yanagisawa, R. The Atlas of Standard Radiographs of Arthritis. Rheumatology 2005, 44, iv43–iv72. [Google Scholar] [CrossRef]
  72. Kondal, S.; Kulkarni, V.; Gaikwad, A.; Kharat, A.; Pant, A. Automatic Grading of Knee Osteoarthritis on the Kellgren-Lawrence Scale from Radiographs Using Convolutional Neural Networks. In Advances in Deep Learning, Artificial Intelligence and Robotics. Lecture Notes in Networks and Systems; Springer: Cham, Switzerland, 2022; pp. 163–173. [Google Scholar]
  73. Park, H.-J.; Kim, S.S.; Lee, S.-Y.; Park, N.-H.; Park, J.-Y.; Choi, Y.-J.; Jeon, H.-J. A Practical MRI Grading System for Osteoarthritis of the Knee: Association with Kellgren–Lawrence Radiographic Scores. Eur. J. Radiol. 2013, 82, 112–117. [Google Scholar] [CrossRef] [PubMed]
  74. Niinimäki, E.; Paloneva, J.; Pölönen, I.; Heinonen, A.; Äyrämö, S. Validation of Knee KL-Classifying Deep Neural Network with Finnish Patient Data. In Computational Sciences and Artificial Intelligence in Industry; Springer: Cham, Switzerland, 2022; pp. 177–188. [Google Scholar]
  75. Field, R.E.; Blakey, C.; Malagelada, F. Anatomy: Capsule and Synovium. In Hip Joint Restoration; Springer: New York, NY, USA, 2017; pp. 27–33. [Google Scholar]
  76. Wang, W.; Ye, R.; Xie, W.; Zhang, Y.; An, S.; Li, Y.; Zhou, Y. Roles of the Calcified Cartilage Layer and Its Tissue Engineering Reconstruction in Osteoarthritis Treatment. Front. Bioeng. Biotechnol. 2022, 10, 911281. [Google Scholar] [CrossRef] [PubMed]
  77. Fawns, H.T.; Landells, J.W. Histochemical Studies of Rheumatic Conditions: I. Observations on the Fine Structures of the Matrix of Normal Bone and Cartilage. Ann. Rheum. Dis. 1953, 12, 105–113. [Google Scholar] [CrossRef]
  78. Ralphs, J.R.; Benjamin, M. The Joint Capsule: Structure, Composition, Ageing and Disease. J. Anat. 1994, 184 Pt 3, 503–509. [Google Scholar]
  79. Coaccioli, S.; Sarzi-Puttini, P.; Zis, P.; Rinonapoli, G.; Varrassi, G. Osteoarthritis: New Insight on Its Pathophysiology. J. Clin. Med. 2022, 11, 6013. [Google Scholar] [CrossRef] [PubMed]
  80. Burr, D.B.; Gallant, M.A. Bone Remodelling in Osteoarthritis. Nat. Rev. Rheumatol. 2012, 8, 665–673. [Google Scholar] [CrossRef] [PubMed]
  81. Hu, Y.; Chen, X.; Wang, S.; Jing, Y.; Su, J. Subchondral Bone Microenvironment in Osteoarthritis and Pain. Bone Res. 2021, 9, 20. [Google Scholar] [CrossRef] [PubMed]
  82. Woodell-May, J.E.; Sommerfeld, S.D. Role of Inflammation and the Immune System in the Progression of Osteoarthritis. J. Orthop. Res. 2020, 38, 253–257. [Google Scholar] [CrossRef] [PubMed]
  83. Ohashi, Y.; Uchida, K.; Fukushima, K.; Inoue, G.; Takaso, M. Mechanisms of Peripheral and Central Sensitization in Osteoarthritis Pain. Cureus 2023, 15, e35331. [Google Scholar] [CrossRef] [PubMed]
  84. Wu, Z.; Korntner, S.; Mullen, A.; Zeugolis, D. Collagen Type II: From Biosynthesis to Advanced Biomaterials for Cartilage Engineering. Biomater. Biosyst. 2021, 4, 100030. [Google Scholar] [CrossRef] [PubMed]
  85. Roughley, P.J.; Mort, J.S. The Role of Aggrecan in Normal and Osteoarthritic Cartilage. J. Exp. Orthop. 2014, 1, 8. [Google Scholar] [CrossRef] [PubMed]
  86. Chen, H.; Tan, X.-N.; Hu, S.; Liu, R.-Q.; Peng, L.-H.; Li, Y.-M.; Wu, P. Molecular Mechanisms of Chondrocyte Proliferation and Differentiation. Front. Cell Dev. Biol. 2021, 9, 664168. [Google Scholar] [CrossRef]
  87. Lin, W.; Liu, Z.; Kampf, N.; Klein, J. The Role of Hyaluronic Acid in Cartilage Boundary Lubrication. Cells 2020, 9, 1606. [Google Scholar] [CrossRef]
  88. Alcaide-Ruggiero, L.; Cugat, R.; Domínguez, J.M. Proteoglycans in Articular Cartilage and Their Contribution to Chondral Injury and Repair Mechanisms. Int. J. Mol. Sci. 2023, 24, 10824. [Google Scholar] [CrossRef]
  89. Mobasheri, A.; Batt, M. An Update on the Pathophysiology of Osteoarthritis. Ann. Phys. Rehabil. Med. 2016, 59, 333–339. [Google Scholar] [CrossRef]
  90. 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] [PubMed]
  91. Carter, D.R.; Beaupré, G.S.; Wong, M.; Smith, R.L.; Andriacchi, T.P.; Schurman, D.J. The Mechanobiology of Articular Cartilage Development and Degeneration. Clin. Orthop. Relat. Res. 2004, 427, S69–S77. [Google Scholar] [CrossRef] [PubMed]
  92. Poole, C.A. Review. Articular Cartilage Chondrons: Form, Function and Failure. J. Anat. 1997, 191, 1–13. [Google Scholar] [CrossRef]
  93. Amr, M.; Mallah, A.; Yasmeen, S.; Van Wie, B.; Gozen, A.; Mendenhall, J.; Abu-Lail, N.I. From Chondrocytes to Chondrons, Maintenance of Phenotype and Matrix Production in a Composite 3D Hydrogel Scaffold. Gels 2022, 8, 90. [Google Scholar] [CrossRef] [PubMed]
  94. Dubey, N.K.; Deng, W.-P. 20—Polymeric Gels for Cartilage Tissue Engineering. In Polymeric Gels; Kunal, P., Indranil, B., Eds.; Woodhead Publishing: Cambridge, UK, 2018; pp. 505–525. [Google Scholar]
  95. Alexopoulos, L.G.; Haider, M.A.; Vail, T.P.; Guilak, F. Alterations in the Mechanical Properties of the Human Chondrocyte Pericellular Matrix with Osteoarthritis. J. Biomech. Eng. 2003, 125, 323–333. [Google Scholar] [CrossRef]
  96. Dieterle, M.P.; Husari, A.; Rolauffs, B.; Steinberg, T.; Tomakidi, P. Integrins, Cadherins and Channels in Cartilage Mechanotransduction: Perspectives for Future Regeneration Strategies. Expert. Rev. Mol. Med. 2021, 23, e14. [Google Scholar] [CrossRef]
  97. Song, F.; Mao, X.; Dai, J.; Shan, B.; Zhou, Z.; Kang, Y. Integrin AVβ3 Signaling in the Progression of Osteoarthritis Induced by Excessive Mechanical Stress. Inflammation 2023, 46, 739–751. [Google Scholar] [CrossRef]
  98. Tang, X.; Muhammad, H.; McLean, C.; Miotla-Zarebska, J.; Fleming, J.; Didangelos, A.; Önnerfjord, P.; Leask, A.; Saklatvala, J.; Vincent, T.L. Connective Tissue Growth Factor Contributes to Joint Homeostasis and Osteoarthritis Severity by Controlling the Matrix Sequestration and Activation of Latent TGFβ. Ann. Rheum. Dis. 2018, 77, 1372–1380. [Google Scholar] [CrossRef]
  99. Huck, L.; Pontier, S.M.; Zuo, D.M.; Muller, W.J. Β1-Integrin Is Dispensable for the Induction of ErbB2 Mammary Tumors but Plays a Critical Role in the Metastatic Phase of Tumor Progression. Proc. Natl. Acad. Sci. USA 2010, 107, 15559–15564. [Google Scholar] [CrossRef]
  100. 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]
  101. Yamamoto, K.; Wilkinson, D.; Bou-Gharios, G. Targeting Dysregulation of Metalloproteinase Activity in Osteoarthritis. Calcif. Tissue Int. 2021, 109, 277–290. [Google Scholar] [CrossRef] [PubMed]
  102. 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]
  103. Peng, Z.; Sun, H.; Bunpetch, V.; Koh, Y.; Wen, Y.; Wu, D.; Ouyang, H. The Regulation of Cartilage Extracellular Matrix Homeostasis in Joint Cartilage Degeneration and Regeneration. Biomaterials 2021, 268, 120555. [Google Scholar] [CrossRef]
  104. Werb, Z.; Tremble, P.M.; Behrendtsen, O.; Crowley, E.; Damsky, C.H. Signal Transduction through the Fibronectin Receptor Induces Collagenase and Stromelysin Gene Expression. J. Cell Biol. 1989, 109, 877–889. [Google Scholar] [CrossRef]
  105. Yang, C.-Y.; Chanalaris, A.; Troeberg, L. ADAMTS and ADAM Metalloproteinases in Osteoarthritis—Looking beyond the ‘Usual Suspects’. Osteoarthr. Cartil. 2017, 25, 1000–1009. [Google Scholar] [CrossRef] [PubMed]
  106. Sporn, M.B. Transforming Growth Factor—β. JAMA 1989, 262, 938. [Google Scholar] [CrossRef] [PubMed]
  107. Finnson, K.W.; Chi, Y.; Bou-Gharios, G.; Leask, A.; Philip, A. TGF-Beta Signaling in Cartilage Homeostasis and Osteoarthritis. Front. Biosci. 2012, S4, 251. [Google Scholar] [CrossRef]
  108. Schaffner, F.; Ray, A.M.; Dontenwill, M. Integrin A5β1, the Fibronectin Receptor, as a Pertinent Therapeutic Target in Solid Tumors. Cancers 2013, 5, 27–47. [Google Scholar] [CrossRef]
  109. Ramage, L. Integrins and Extracellular Matrix in Mechanotransduction. Cell Health Cytoskelet. 2011, 2012, 1–9. [Google Scholar] [CrossRef]
  110. Pang, C.; Wen, L.; Lu, X.; Luo, S.; Qin, H.; Zhang, X.; Zhu, B.; Luo, S. Ruboxistaurin Maintains the Bone Mass of Subchondral Bone for Blunting Osteoarthritis Progression by Inhibition of Osteoclastogenesis and Bone Resorption Activity. Biomed. Pharmacother. 2020, 131, 110650. [Google Scholar] [CrossRef] [PubMed]
  111. Pang, C.; Wen, L.; Qin, H.; Zhu, B.; Lu, X.; Luo, S. Sotrastaurin, a PKC Inhibitor, Attenuates RANKL-induced Bone Resorption and Attenuates Osteochondral Pathologies Associated with the Development of OA. J. Cell Mol. Med. 2020, 24, 8452–8465. [Google Scholar] [CrossRef] [PubMed]
  112. Jin, H.; Jiang, S.; Wang, R.; Zhang, Y.; Dong, J.; Li, Y. Mechanistic Insight Into the Roles of Integrins in Osteoarthritis. Front. Cell Dev. Biol. 2021, 9, 693484. [Google Scholar] [CrossRef] [PubMed]
  113. Jimi, E.; Huang, F.; Nakatomi, C. NF-ΚB Signaling Regulates Physiological and Pathological Chondrogenesis. Int. J. Mol. Sci. 2019, 20, 6275. [Google Scholar] [CrossRef] [PubMed]
  114. Yu, H.; Lin, L.; Zhang, Z.; Zhang, H.; Hu, H. Targeting NF-ΚB Pathway for the Therapy of Diseases: Mechanism and Clinical Study. Signal Transduct. Target. Ther. 2020, 5, 209. [Google Scholar] [CrossRef] [PubMed]
  115. Choi, M.C.; Jo, J.; Park, J.; Kang, H.K.; Park, Y. Park NF-B Signaling Pathways in Osteoarthritic Cartilage Destruction. Cells 2019, 8, 734. [Google Scholar] [CrossRef] [PubMed]
  116. Xu, F.; Zhao, L.-J.; Liao, T.; Li, Z.-C.; Wang, L.-L.; Lin, P.-Y.; Jiang, R.; Wei, Q.-J. Ononin Ameliorates Inflammation and Cartilage Degradation in Rat Chondrocytes with IL-1β-Induced Osteoarthritis by Downregulating the MAPK and NF-ΚB Pathways. BMC Complement. Med. Ther. 2022, 22, 25. [Google Scholar] [CrossRef] [PubMed]
  117. Zhang, H.; Li, S.; Lu, J.; Jin, J.; Zhu, G.; Wang, L.; Yan, Y.; He, L.; Wang, B.; Wang, X.; et al. α-Cyperone (CYP) down-Regulates NF-ΚB and MAPKs Signaling, Attenuating Inflammation and Extracellular Matrix Degradation in Chondrocytes, to Ameliorate Osteoarthritis in Mice. Aging 2021, 13, 17690–17706. [Google Scholar] [CrossRef] [PubMed]
  118. Tu, J.; Li, W.; Zhang, Y.; Wu, X.; Song, Y.; Kang, L.; Liu, W.; Wang, K.; Li, S.; Hua, W.; et al. Simvastatin Inhibits IL-1β-Induced Apoptosis and Extracellular Matrix Degradation by Suppressing the NF-KB and MAPK Pathways in Nucleus Pulposus Cells. Inflammation 2017, 40, 725–734. [Google Scholar] [CrossRef]
  119. Huang, B.-P.; Lin, C.-H.; Chen, H.-M.; Lin, J.-T.; Cheng, Y.-F.; Kao, S.-H. AMPK Activation Inhibits Expression of Proinflammatory Mediators Through Downregulation of PI3K/P38 MAPK and NF-ΚB Signaling in Murine Macrophages. DNA Cell Biol. 2015, 34, 133–141. [Google Scholar] [CrossRef]
  120. Scotece, M.; Conde, J.; Abella, V.; López, V.; Francisco, V.; Ruiz, C.; Campos, V.; Lago, F.; Gomez, R.; Pino, J.; et al. Oleocanthal Inhibits Catabolic and Inflammatory Mediators in LPS-Activated Human Primary Osteoarthritis (OA) Chondrocytes Through MAPKs/NF-ΚB Pathways. Cell. Physiol. Biochem. 2018, 49, 2414–2426. [Google Scholar] [CrossRef] [PubMed]
  121. Thummuri, D.; Jeengar, M.K.; Shrivastava, S.; Nemani, H.; Ramavat, R.N.; Chaudhari, P.; Naidu, V.G.M. Thymoquinone Prevents RANKL-Induced Osteoclastogenesis Activation and Osteolysis in an In Vivo Model of Inflammation by Suppressing NF-KB and MAPK Signalling. Pharmacol. Res. 2015, 99, 63–73. [Google Scholar] [CrossRef] [PubMed]
  122. Jiang, S.; Zhang, C.; Lu, Y.; Yuan, F. The Molecular Mechanism Research of Cartilage Calcification Induced by Osteoarthritis. Bioengineered 2022, 13, 13082–13088. [Google Scholar] [CrossRef] [PubMed]
  123. Liu, Q.; Wang, R.; Hou, S.; He, F.; Ma, Y.; Ye, T.; Yu, S.; Chen, H.; Wang, H.; Zhang, M. Chondrocyte-Derived Exosomes Promote Cartilage Calcification in Temporomandibular Joint Osteoarthritis. Arthritis Res. Ther. 2022, 24, 44. [Google Scholar] [CrossRef] [PubMed]
  124. Shen, G. The Role of Type X Collagen in Facilitating and Regulating Endochondral Ossification of Articular Cartilage. Orthod. Craniofac Res. 2005, 8, 11–17. [Google Scholar] [CrossRef] [PubMed]
  125. Alcaide-Ruggiero, L.; Molina-Hernández, V.; Granados, M.M.; Domínguez, J.M. Main and Minor Types of Collagens in the Articular Cartilage: The Role of Collagens in Repair Tissue Evaluation in Chondral Defects. Int. J. Mol. Sci. 2021, 22, 13329. [Google Scholar] [CrossRef] [PubMed]
  126. Krasnokutsky, S.; Attur, M.; Palmer, G.; Samuels, J.; Abramson, S.B. Current Concepts in the Pathogenesis of Osteoarthritis. Osteoarthr. Cartil. 2008, 16, S1–S3. [Google Scholar] [CrossRef]
  127. Wang, N.; Zhang, X.; Rothrauff, B.B.; Fritch, M.R.; Chang, A.; He, Y.; Yeung, M.; Liu, S.; Lipa, K.E.; Lei, G.; et al. Novel Role of Estrogen Receptor-α on Regulating Chondrocyte Phenotype and Response to Mechanical Loading. Osteoarthr. Cartil. 2022, 30, 302–314. [Google Scholar] [CrossRef] [PubMed]
  128. Guo, Y.; Tian, L.; Du, X.; Deng, Z. MiR-203 Regulates Estrogen Receptor α and Cartilage Degradation in IL-1β-Stimulated Chondrocytes. J. Bone Miner. Metab. 2020, 38, 346–356. [Google Scholar] [CrossRef]
  129. AKALTUN, M.S.; KOÇYİĞİT, B.F. Assessment of Foot Posture and Related Factors in Patients with Knee Osteoarthritis. Arch. Rheumatol. 2021, 36, 267–273. [Google Scholar] [CrossRef]
  130. Fu, S.; Duan, T.; Hou, M.; Yang, F.; Chai, Y.; Chen, Y.; Liu, B.; Ma, Y.; Liu, A.; Wang, X.; et al. Postural Balance in Individuals with Knee Osteoarthritis During Stand-to-Sit Task. Front. Hum. Neurosci. 2021, 15, 760960. [Google Scholar] [CrossRef] [PubMed]
  131. Lim, Y.Z.; Wong, J.; Hussain, S.M.; Estee, M.M.; Zolio, L.; Page, M.J.; Harrison, C.L.; Wluka, A.E.; Wang, Y.; Cicuttini, F.M. Recommendations for Weight Management in Osteoarthritis: A Systematic Review of Clinical Practice Guidelines. Osteoarthr. Cartil. Open 2022, 4, 100298. [Google Scholar] [CrossRef] [PubMed]
  132. Biederman, R.E. Pharmacology in Rehabilitation: Nonsteroidal Anti-Inflammatory Agents. J. Orthop. Sports Phys. Ther. 2005, 35, 356–367. [Google Scholar] [CrossRef] [PubMed]
  133. Magni, A.; Agostoni, P.; Bonezzi, C.; Massazza, G.; Menè, P.; Savarino, V.; Fornasari, D. Management of Osteoarthritis: Expert Opinion on NSAIDs. Pain. Ther. 2021, 10, 783–808. [Google Scholar] [CrossRef] [PubMed]
  134. Weng, Q.; Goh, S.-L.; Wu, J.; Persson, M.S.M.; Wei, J.; Sarmanova, A.; Li, X.; Hall, M.; Doherty, M.; Jiang, T.; et al. Comparative Efficacy of Exercise Therapy and Oral Non-Steroidal Anti-Inflammatory Drugs and Paracetamol for Knee or Hip Osteoarthritis: A Network Meta-Analysis of Randomised Controlled Trials. Br. J. Sports Med. 2023, 57, 990–996. [Google Scholar] [CrossRef] [PubMed]
  135. Mohamadi, A.; Chan, J.J.; Claessen, F.M.A.P.; Ring, D.; Chen, N.C. Corticosteroid Injections Give Small and Transient Pain Relief in Rotator Cuff Tendinosis: A Meta-Analysis. Clin. Orthop. Relat. Res. 2017, 475, 232–243. [Google Scholar] [CrossRef] [PubMed]
  136. Agostini, F.; Ferrillo, M.; Bernetti, A.; Finamore, N.; Mangone, M.; Giudice, A.; Paoloni, M.; de Sire, A. Hyaluronic Acid Injections for Pain Relief and Functional Improvement in Patients with Temporomandibular Disorders: An Umbrella Review of Systematic Reviews. J. Oral. Rehabil. 2023, 50, 1518–1534. [Google Scholar] [CrossRef] [PubMed]
  137. Barman, A.; Mishra, A.; Maiti, R.; Sahoo, J.; Thakur, K.B.; Sasidharan, S.K. Can Platelet-Rich Plasma Injections Provide Better Pain Relief and Functional Outcomes in Persons with Common Shoulder Diseases: A Meta-Analysis of Randomized Controlled Trials. Clin. Shoulder Elb. 2022, 25, 73–89. [Google Scholar] [CrossRef]
  138. Stubbs, B.; Aluko, Y.; Myint, P.K.; Smith, T.O. Prevalence of Depressive Symptoms and Anxiety in Osteoarthritis: A Systematic Review and Meta-Analysis. Age Ageing 2016, 45, 228–235. [Google Scholar] [CrossRef]
  139. Wang, S.-T.; Ni, G.-X. Depression in Osteoarthritis: Current Understanding. Neuropsychiatr. Dis. Treat. 2022, 18, 375–389. [Google Scholar] [CrossRef]
  140. Uritani, D.; Koda, H.; Sugita, S. Effects of Self-Management Education Programmes on Self-Efficacy for Osteoarthritis of the Knee: A Systematic Review of Randomised Controlled Trials. BMC Musculoskelet. Disord. 2021, 22, 515. [Google Scholar] [CrossRef]
  141. Diener, I. Physiotherapy Support for Self-Management of Persisting Musculoskeletal Pain Disorders. S. Afr. J. Physiother. 2021, 77, 1564. [Google Scholar] [CrossRef] [PubMed]
  142. 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, 72, 220–233. [Google Scholar] [CrossRef] [PubMed]
  143. Song, J.-A.; Oh, J.W. Effects of Aquatic Exercises for Patients with Osteoarthritis: Systematic Review with Meta-Analysis. Healthcare 2022, 10, 560. [Google Scholar] [CrossRef]
  144. Guo, G.; Wu, B.; Xie, S.; Xu, J.; Zhou, X.; Wu, G.; Lu, P. Effectiveness and Safety of Tai Chi for Chronic Pain of Knee Osteoarthritis. Medicine 2022, 101, e28497. [Google Scholar] [CrossRef] [PubMed]
  145. Lauche, R.; Hunter, D.J.; Adams, J.; Cramer, H. Yoga for Osteoarthritis: A Systematic Review and Meta-Analysis. Curr. Rheumatol. Rep. 2019, 21, 47. [Google Scholar] [CrossRef] [PubMed]
  146. Rocha, T.C.; Ramos, P.d.S.; Dias, A.G.; Martins, E.A. The Effects of Physical Exercise on Pain Management in Patients with Knee Osteoarthritis: A Systematic Review with Metanalysis. Rev. Bras. Ortop. 2020, 55, 509–517. [Google Scholar] [CrossRef] [PubMed]
  147. Rouzer, C.A.; Marnett, L.J. Cyclooxygenases: Structural and Functional Insights. J. Lipid Res. 2009, 50, S29–S34. [Google Scholar] [CrossRef] [PubMed]
  148. Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative Stress, Inflammation, and Cancer: How Are They Linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef]
  149. Amin, A.R.; Attur, M.; Patel, R.N.; Thakker, G.D.; Marshall, P.J.; Rediske, J.; Stuchin, S.A.; Patel, I.R.; Abramson, S.B. Superinduction of Cyclooxygenase-2 Activity in Human Osteoarthritis-Affected Cartilage. Influence of Nitric Oxide. J. Clin. Investig. 1997, 99, 1231–1237. [Google Scholar] [CrossRef]
  150. Li, W.; Hu, S.; Chen, X.; Shi, J. The Antioxidant Resveratrol Protects against Chondrocyte Apoptosis by Regulating the COX-2/NF-ΚB Pathway in Created Temporomandibular Osteoarthritis. Biomed. Res. Int. 2021, 2021, 9978651. [Google Scholar] [CrossRef] [PubMed]
  151. Takeuchi, K.; Amagase, K. Roles of Cyclooxygenase, Prostaglandin E2 and EP Receptors in Mucosal Protection and Ulcer Healing in the Gastrointestinal Tract. Curr. Pharm. Des. 2018, 24, 2002–2011. [Google Scholar] [CrossRef] [PubMed]
  152. Flower, R.J. The Development of COX2 Inhibitors. Nat. Rev. Drug Discov. 2003, 2, 179–191. [Google Scholar] [CrossRef] [PubMed]
  153. Ricciotti, E.; Yu, Y.; Grosser, T.; FitzGerald, G.A. COX-2, the Dominant Source of Prostacyclin. Proc. Natl. Acad. Sci. USA 2013, 110, E183. [Google Scholar] [CrossRef] [PubMed]
  154. Morita, I. Distinct Functions of COX-1 and COX-2. Prostaglandins Other Lipid Mediat. 2002, 68–69, 165–175. [Google Scholar] [CrossRef] [PubMed]
  155. Dragani, A.; Pascale, S.; Recchiuti, A.; Mattoscio, D.; Lattanzio, S.; Petrucci, G.; Mucci, L.; Ferrante, E.; Habib, A.; Ranelletti, F.O.; et al. The Contribution of Cyclooxygenase-1 and -2 to Persistent Thromboxane Biosynthesis in Aspirin-Treated Essential Thrombocythemia: Implications for Antiplatelet Therapy. Blood 2010, 115, 1054–1061. [Google Scholar] [CrossRef] [PubMed]
  156. Spektor, G.; Fuster, V. Drug Insight: Cyclo-Oxygenase 2 Inhibitors and Cardiovascular Risk—Where Are We Now? Nat. Clin. Pract. Cardiovasc. Med. 2005, 2, 290–300. [Google Scholar] [CrossRef]
  157. Pennick, G.; Robinson-Miller, A.; Cush, I. Topical NSAIDs for Acute Local Pain Relief: In Vitro Characterization of Drug Delivery Profiles into and through Human Skin. Drug Dev. Ind. Pharm. 2021, 47, 908–918. [Google Scholar] [CrossRef] [PubMed]
  158. Klinge, S.A.; Sawyer, G.A. Effectiveness and Safety of Topical versus Oral Nonsteroidal Anti-Inflammatory Drugs: A Comprehensive Review. Phys. Sportsmed. 2013, 41, 64–74. [Google Scholar] [CrossRef]
  159. 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, S18–S21. [Google Scholar] [CrossRef]
  160. Cruz-Topete, D.; Cidlowski, J.A. Glucocorticoids: Molecular Mechanisms of Action. In Immunopharmacology and Inflammation; Springer International Publishing: Cham, Switzerland, 2018; pp. 249–266. [Google Scholar]
  161. Barnes, P.J. Corticosteroid Effects on Cell Signalling. Eur. Respir. J. 2006, 27, 413–426. [Google Scholar] [CrossRef] [PubMed]
  162. Rice, J.B.; White, A.G.; Scarpati, L.M.; Wan, G.; Nelson, W.W. Long-Term Systemic Corticosteroid Exposure: A Systematic Literature Review. Clin. Ther. 2017, 39, 2216–2229. [Google Scholar] [CrossRef] [PubMed]
  163. Li, J.-X.; Cummins, C.L. Fresh Insights into Glucocorticoid-Induced Diabetes Mellitus and New Therapeutic Directions. Nat. Rev. Endocrinol. 2022, 18, 540–557. [Google Scholar] [CrossRef] [PubMed]
  164. Grennan, D.; Wang, S. Steroid Side Effects. JAMA 2019, 322, 282. [Google Scholar] [CrossRef]
  165. Conklin, A.I.; Hong, J. Obesity Prevention in Corticosteroid-treated Patients: Use and Effectiveness of Strategies for Weight Management. Clin. Obes. 2019, 9, e12312. [Google Scholar] [CrossRef] [PubMed]
  166. Estee, M.M.; Cicuttini, F.M.; Page, M.J.; Butala, A.D.; Wluka, A.E.; Hussain, S.M.; Wang, Y. Efficacy of Corticosteroids for Hand Osteoarthritis—A Systematic Review and Meta-Analysis of Randomized Controlled Trials. BMC Musculoskelet. Disord. 2022, 23, 665. [Google Scholar] [CrossRef] [PubMed]
  167. Wenham, C.Y.J.; Hensor, E.M.A.; Grainger, A.J.; Hodgson, R.; Balamoody, S.; Dore, C.J.; Emery, P.; Conaghan, P.G. A Randomized, Double-Blind, Placebo-Controlled Trial of Low-Dose Oral Prednisolone for Treating Painful Hand Osteoarthritis. Rheumatology 2012, 51, 2286–2294. [Google Scholar] [CrossRef]
  168. Hollander, J.L. Intra-Articular Hydrocortisone in Arthritis and Allied Conditions; a Summary of Two Years’ Clinical Experience. J. Bone Jt. Surg. Am. 1953, 35, 983–990. [Google Scholar] [CrossRef]
  169. Miller, J.H.; White, J.; Norton, T.H. The value of intra-articular injections in osteoarthritis of the knee. J. Bone Jt. Surg. Br. 1958, 40, 636–643. [Google Scholar] [CrossRef]
  170. Kroon, F.P.B.; Rubio, R.; Schoones, J.W.; Kloppenburg, M. Intra-Articular Therapies in the Treatment of Hand Osteoarthritis: A Systematic Literature Review. Drugs Aging 2016, 33, 119–133. [Google Scholar] [CrossRef]
  171. Parker, E.B.; Hering, K.A.; Chiodo, C.P.; Smith, J.T.; Bluman, E.M.; Martin, E.A. Intraarticular Injections in the Foot and Ankle: Medication Selection Patterns and Perceived Risk Of Chondrotoxicity. Foot Ankle Orthop. 2023, 8, 24730114231216990. [Google Scholar] [CrossRef] [PubMed]
  172. Metzger, C.M.; Farooq, H.; Merrell, G.A.; Kaplan, F.T.D.; Greenberg, J.A.; Crosby, N.E.; Peck, K.M.; Hoyer, R.W. Efficacy of a Single, Image-Guided Corticosteroid Injection for Glenohumeral Arthritis. J. Shoulder Elb. Surg. 2021, 30, 1128–1134. [Google Scholar] [CrossRef]
  173. Zhong, H.-M.; Zhao, G.-F.; Lin, T.; Zhang, X.-X.; Li, X.-Y.; Lin, J.-F.; Zhao, S.-Q.; Pan, Z.-J. Intra-Articular Steroid Injection for Patients with Hip Osteoarthritis: A Systematic Review and Meta-Analysis. Biomed. Res. Int. 2020, 2020, 6320154. [Google Scholar] [CrossRef] [PubMed]
  174. McAlindon, T.E.; LaValley, M.P.; Harvey, W.F.; Price, L.L.; Driban, J.B.; Zhang, M.; Ward, R.J. Effect of Intra-Articular Triamcinolone vs Saline on Knee Cartilage Volume and Pain in Patients with Knee Osteoarthritis. JAMA 2017, 317, 1967. [Google Scholar] [CrossRef] [PubMed]
  175. Pelletier, J.-P.; Raynauld, J.-P.; Abram, F.; Dorais, M.; Paiement, P.; Martel-Pelletier, J. Intra-Articular Corticosteroid Knee Injection Induces a Reduction in Meniscal Thickness with No Treatment Effect on Cartilage Volume: A Case–Control Study. Sci. Rep. 2020, 10, 13789. [Google Scholar] [CrossRef]
  176. Guermazi, A.; Neogi, T.; Katz, J.N.; Kwoh, C.K.; Conaghan, P.G.; Felson, D.T.; Roemer, F.W. Intra-Articular Corticosteroid Injections for the Treatment of Hip and Knee Osteoarthritis-Related Pain: Considerations and Controversies with a Focus on Imaging— Radiology Scientific Expert Panel. Radiology 2020, 297, 503–512. [Google Scholar] [CrossRef]
  177. Aaron, R.K.; Voisinet, A.; Racine, J.; Ali, Y.; Feller, E.R. Corticosteroid-associated Avascular Necrosis: Dose Relationships and Early Diagnosis. Ann. N. Y. Acad. Sci. 2011, 1240, 38–46. [Google Scholar] [CrossRef]
  178. McCormick, B.P.; Sequeira, S.B.; Hasenauer, M.D.; McKinstry, R.P.; Boucher, H.R. Cushing’s Syndrome Is Associated with Early Medical- and Surgical-Related Complications Following Total Joint Arthroplasty: A National Database Study. J. Arthroplast. 2023, 38, 2568–2572. [Google Scholar] [CrossRef] [PubMed]
  179. Russell, S.J.; Sala, R.; Conaghan, P.G.; Habib, G.; Vo, Q.; Manning, R.; Kivitz, A.; Davis, Y.; Lufkin, J.; Johnson, J.R.; et al. Triamcinolone Acetonide Extended-Release in Patients with Osteoarthritis and Type 2 Diabetes: A Randomized, Phase 2 Study. Rheumatology 2018, 57, 2235–2241. [Google Scholar] [CrossRef] [PubMed]
  180. Mader, R.; Lavi, I.; Luboshitzky, R. Evaluation of the Pituitary–Adrenal Axis Function Following Single Intraarticular Injection of Methylprednisolone. Arthritis Rheum. 2005, 52, 924–928. [Google Scholar] [CrossRef]
  181. 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] [PubMed]
  182. Pavone, V.; Vescio, A.; Turchetta, M.; Giardina, S.M.C.; Culmone, A.; Testa, G. Injection-Based Management of Osteoarthritis of the Knee: A Systematic Review of Guidelines. Front. Pharmacol. 2021, 12, 661805. [Google Scholar] [CrossRef] [PubMed]
  183. Bendele, A.M.; Neelagiri, M.; Neelagiri, V.; Sucholeiki, I. Development of a Selective Matrix Metalloproteinase 13 (MMP-13) Inhibitor for the Treatment of Osteoarthritis. Eur. J. Med. Chem. 2021, 224, 113666. [Google Scholar] [CrossRef] [PubMed]
  184. Ding, Y.; O’Keefe, H.; DeLorey, J.L.; Israel, D.I.; Messer, J.A.; Chiu, C.H.; Skinner, S.R.; Matico, R.E.; Murray-Thompson, M.F.; Li, F.; et al. Discovery of Potent and Selective Inhibitors for ADAMTS-4 through DNA-Encoded Library Technology (ELT). ACS Med. Chem. Lett. 2015, 6, 888–893. [Google Scholar] [CrossRef] [PubMed]
  185. Verma, P.; Dalal, K. ADAMTS-4 and ADAMTS-5: Key Enzymes in Osteoarthritis. J. Cell Biochem. 2011, 112, 3507–3514. [Google Scholar] [CrossRef] [PubMed]
  186. Siebuhr, A.S.; Werkmann, D.; Bay-Jensen, A.-C.; Thudium, C.S.; Karsdal, M.A.; Serruys, B.; Ladel, C.; Michaelis, M.; Lindemann, S. The Anti-ADAMTS-5 Nanobody® M6495 Protects Cartilage Degradation Ex Vivo. Int. J. Mol. Sci. 2020, 21, 5992. [Google Scholar] [CrossRef] [PubMed]
  187. Briat, A.; Jacques, C.; Malige, M.; Sudre, L.; Nourissat, G.; Auzeloux, P.; Guehring, H.; Cachin, F.; Berenbaum, F.; Miot-Noirault, E. 99mTc-NTP 15-5 Is a Companion Radiotracer for Assessing Joint Functional Response to Sprifermin (RhFGF-18) in a Murine Osteoarthritis Model. Sci. Rep. 2022, 12, 8146. [Google Scholar] [CrossRef] [PubMed]
  188. Ladel, C. SP0089 Pre-Clinical Proof for Dmoad Activity of FGF-18 (Sprifermin). Ann. Rheum. Dis. 2013, 72, A21. [Google Scholar] [CrossRef]
  189. Moretti, A.; Paoletta, M.; Liguori, S.; Ilardi, W.; Snichelotto, F.; Toro, G.; Gimigliano, F.; Iolascon, G. The Rationale for the Intra-Articular Administration of Clodronate in Osteoarthritis. Int. J. Mol. Sci. 2021, 22, 2693. [Google Scholar] [CrossRef]
  190. Goldring, M.B. Anticytokine Therapy for Osteoarthritis. Expert. Opin. Biol. Ther. 2001, 1, 817–829. [Google Scholar] [CrossRef]
  191. Cho, J.; Kim, T.; Park, Y.; Shin, J.; Kang, S.; Lee, B. InvossaTM(Tissuegene-C) in Patients with Osteoarthritis: A Phase III Trial. Osteoarthr. Cartil. 2016, 24, S190. [Google Scholar] [CrossRef]
  192. Hunter, D.J.; Pike, M.C.; Jonas, B.L.; Kissin, E.; Krop, J.; McAlindon, T. Phase 1 Safety and Tolerability Study of BMP-7 in Symptomatic Knee Osteoarthritis. BMC Musculoskelet. Disord. 2010, 11, 232. [Google Scholar] [CrossRef] [PubMed]
  193. Gasek, N.S.; Kuchel, G.A.; Kirkland, J.L.; Xu, M. Strategies for Targeting Senescent Cells in Human Disease. Nat. Aging 2021, 1, 870–879. [Google Scholar] [CrossRef]
  194. Yagi, M.; Endo, K.; Komori, K.; Sekiya, I. Comparison of the Effects of Oxidative and Inflammatory Stresses on Rat Chondrocyte Senescence. Sci. Rep. 2023, 13, 7697. [Google Scholar] [CrossRef] [PubMed]
  195. Farr, J.N.; Fraser, D.G.; Wang, H.; Jaehn, K.; Ogrodnik, M.B.; Weivoda, M.M.; Drake, M.T.; Tchkonia, T.; LeBrasseur, N.K.; Kirkland, J.L.; et al. Identification of Senescent Cells in the Bone Microenvironment. J. Bone Miner. Res. 2016, 31, 1920–1929. [Google Scholar] [CrossRef]
  196. Sousa-Victor, P.; Gutarra, S.; García-Prat, L.; Rodriguez-Ubreva, J.; Ortet, L.; Ruiz-Bonilla, V.; Jardí, M.; Ballestar, E.; González, S.; Serrano, A.L.; et al. Geriatric Muscle Stem Cells Switch Reversible Quiescence into Senescence. Nature 2014, 506, 316–321. [Google Scholar] [CrossRef]
  197. Fang, H.; Huang, L.; Welch, I.; Norley, C.; Holdsworth, D.W.; Beier, F.; Cai, D. Early Changes of Articular Cartilage and Subchondral Bone in The DMM Mouse Model of Osteoarthritis. Sci. Rep. 2018, 8, 2855. [Google Scholar] [CrossRef]
  198. Cuollo, L.; Antonangeli, F.; Santoni, A.; Soriani, A. The Senescence-Associated Secretory Phenotype (SASP) in the Challenging Future of Cancer Therapy and Age-Related Diseases. Biology 2020, 9, 485. [Google Scholar] [CrossRef]
  199. O’Reilly, L.A.; Huang, D.C.; Strasser, A. The Cell Death Inhibitor Bcl-2 and Its Homologues Influence Control of Cell Cycle Entry. EMBO J. 1996, 15, 6979–6990. [Google Scholar] [CrossRef] [PubMed]
  200. Liu, S.; Liu, S.; Wang, X.; Zhou, J.; Cao, Y.; Wang, F.; Duan, E. The PI3K-Akt Pathway Inhibits Senescence and Promotes Self-renewal of Human Skin-derived Precursors In Vitro. Aging Cell 2011, 10, 661–674. [Google Scholar] [CrossRef]
  201. Velletri, T.; Huang, Y.; Wang, Y.; Li, Q.; Hu, M.; Xie, N.; Yang, Q.; Chen, X.; Chen, Q.; Shou, P.; et al. Loss of P53 in Mesenchymal Stem Cells Promotes Alteration of Bone Remodeling through Negative Regulation of Osteoprotegerin. Cell Death Differ. 2021, 28, 156–169. [Google Scholar] [CrossRef]
  202. Zhu, Y.; Tchkonia, T.; Fuhrmann-Stroissnigg, H.; Dai, H.M.; Ling, Y.Y.; Stout, M.B.; Pirtskhalava, T.; Giorgadze, N.; Johnson, K.O.; Giles, C.B.; et al. Identification of a Novel Senolytic Agent, Navitoclax, Targeting the Bcl-2 Family of Anti-apoptotic Factors. Aging Cell 2016, 15, 428–435. [Google Scholar] [CrossRef] [PubMed]
  203. Georget, M.; Defois, A.; Guiho, R.; Bon, N.; Allain, S.; Boyer, C.; Halgand, B.; Waast, D.; Grimandi, G.; Fouasson-Chailloux, A.; et al. Development of a DNA Damage-Induced Senescence Model in Osteoarthritic Chondrocytes. Aging 2023, 15, 8576–8593. [Google Scholar] [CrossRef] [PubMed]
  204. Malaise, O.; Tachikart, Y.; Constantinides, M.; Mumme, M.; Ferreira-Lopez, R.; Noack, S.; Krettek, C.; Noël, D.; Wang, J.; Jorgensen, C.; et al. Mesenchymal Stem Cell Senescence Alleviates Their Intrinsic and Seno-Suppressive Paracrine Properties Contributing to Osteoarthritis Development. Aging 2019, 11, 9128–9146. [Google Scholar] [CrossRef] [PubMed]
  205. Zhang, X.-X.; He, S.-H.; Liang, X.; Li, W.; Li, T.-F.; Li, D.-F. Aging, Cell Senescence, the Pathogenesis and Targeted Therapies of Osteoarthritis. Front. Pharmacol. 2021, 12, 728100. [Google Scholar] [CrossRef] [PubMed]
  206. Yun, K.; Im, S.-H. Transcriptional Regulation of MMP13 by Lef1 in Chondrocytes. Biochem. Biophys. Res. Commun. 2007, 364, 1009–1014. [Google Scholar] [CrossRef] [PubMed]
  207. Wang, Y.; Zhao, H.; Jia, S.; Wang, Q.; Yao, W.; Yang, Y.; Bai, L. Senomorphic Agent Pterostilbene Ameliorates Osteoarthritis through the PI3K/AKT/NF-ΚB Axis: An In Vitro and In Vivo Study. Am. J. Transl. Res. 2022, 14, 5243–5262. [Google Scholar] [PubMed]
  208. Yang, X.-D.; Corvalan, J.R.F.; Wang, P.; Roy, C.M.-N.; Davis, C.G. Fully Human Anti-Interleukin-8 Monoclonal Antibodies: Potential Therapeutics for the Treatment of Inflammatory Disease States. J. Leukoc. Biol. 1999, 66, 401–410. [Google Scholar] [CrossRef] [PubMed]
  209. Wiegertjes, R.; van de Loo, F.A.J.; Blaney Davidson, E.N. A Roadmap to Target Interleukin-6 in Osteoarthritis. Rheumatology 2020, 59, 2681–2694. [Google Scholar] [CrossRef]
  210. Li, J.; Zhang, B.; Liu, W.-X.; Lu, K.; Pan, H.; Wang, T.; Oh, C.; Yi, D.; Huang, J.; Zhao, L.; et al. Metformin Limits Osteoarthritis Development and Progression through Activation of AMPK Signalling. Ann. Rheum. Dis. 2020, 79, 635–645. [Google Scholar] [CrossRef]
  211. Miller, R.J.; Jung, H.; Bhangoo, S.K.; White, F.A. Cytokine and Chemokine Regulation of Sensory Neuron Function. In Sensory Nerves; Springer: Berlin/Heidelberg, Germany, 2009; pp. 417–449. [Google Scholar]
  212. Salvador, A.F.; de Lima, K.A.; Kipnis, J. Neuromodulation by the Immune System: A Focus on Cytokines. Nat. Rev. Immunol. 2021, 21, 526–541. [Google Scholar] [CrossRef] [PubMed]
  213. Julius, D.; Basbaum, A.I. Molecular Mechanisms of Nociception. Nature 2001, 413, 203–210. [Google Scholar] [CrossRef] [PubMed]
  214. Tazawa, R.; Kenmoku, T.; Uchida, K.; Arendt-Nielsen, L.; Nagura, N.; Nakawaki, M.; Matsumoto, T.; Inoue, G.; Takeuchi, H.; Jimbo, T.; et al. Increased Nerve Growth Factor Expression in the Synovial Tissues of Patients with Rotator Cuff Tears. Mol. Pain. 2021, 17, 174480692110212. [Google Scholar] [CrossRef] [PubMed]
  215. Dakin, P.; DiMartino, S.J.; Gao, H.; Maloney, J.; Kivitz, A.J.; Schnitzer, T.J.; Stahl, N.; Yancopoulos, G.D.; Geba, G.P. The Efficacy, Tolerability, and Joint Safety of Fasinumab in Osteoarthritis Pain: A Phase IIb/III Double-Blind, Placebo-Controlled, Randomized Clinical Trial. Arthritis Rheumatol. 2019, 71, 1824–1834. [Google Scholar] [CrossRef]
  216. Sanga, P.; Katz, N.; Polverejan, E.; Wang, S.; Kelly, K.M.; Haeussler, J.; Thipphawong, J. Efficacy, Safety, and Tolerability of Fulranumab, an Anti-Nerve Growth Factor Antibody, in the Treatment of Patients with Moderate to Severe Osteoarthritis Pain. Pain 2013, 154, 1910–1919. [Google Scholar] [CrossRef] [PubMed]
  217. Nencini, S.; Ringuet, M.; Kim, D.-H.; Chen, Y.-J.; Greenhill, C.; Ivanusic, J.J. Mechanisms of Nerve Growth Factor Signaling in Bone Nociceptors and in an Animal Model of Inflammatory Bone Pain. Mol. Pain. 2017, 13, 174480691769701. [Google Scholar] [CrossRef] [PubMed]
  218. Oo, W.M.; Hunter, D.J. Disease Modification in Osteoarthritis: Are We There Yet? Clin. Exp. Rheumatol. 2019, 37 (Suppl. S1), 135–140. [Google Scholar] [PubMed]
  219. Oo, W.M.; Yu, S.P.-C.; Daniel, M.S.; Hunter, D.J. Disease-Modifying Drugs in Osteoarthritis: Current Understanding and Future Therapeutics. Expert. Opin. Emerg. Drugs 2018, 23, 331–347. [Google Scholar] [CrossRef]
  220. Gupta, R.C.; Lall, R.; Srivastava, A.; Sinha, A. Hyaluronic Acid: Molecular Mechanisms and Therapeutic Trajectory. Front. Vet. Sci. 2019, 6, 192. [Google Scholar] [CrossRef]
  221. Kogan, G.; Šoltés, L.; Stern, R.; Schiller, J.; Mendichi, R. Hyaluronic Acid: Its Function and Degradation in In Vivo Systems. In Studies in Natural Products Chemistry; Elsevier: Amsterdam, The Netherlands, 2008; pp. 789–882. [Google Scholar]
  222. Hemmati-Sadeghi, S.; Ringe, J.; Dehne, T.; Haag, R.; Sittinger, M. Hyaluronic Acid Influence on Normal and Osteoarthritic Tissue-Engineered Cartilage. Int. J. Mol. Sci. 2018, 19, 1519. [Google Scholar] [CrossRef]
  223. Lanza, V.; Greco, V.; Bocchieri, E.; Sciuto, S.; Inturri, R.; Messina, L.; Vaccaro, S.; Bellia, F.; Rizzarelli, E. Synergistic Effect of L-Carnosine and Hyaluronic Acid in Their Covalent Conjugates on the Antioxidant Abilities and the Mutual Defense against Enzymatic Degradation. Antioxidants 2022, 11, 664. [Google Scholar] [CrossRef] [PubMed]
  224. Žádníková, P.; Šínová, R.; Pavlík, V.; Šimek, M.; Šafránková, B.; Hermannová, M.; Nešporová, K.; Velebný, V. The Degradation of Hyaluronan in the Skin. Biomolecules 2022, 12, 251. [Google Scholar] [CrossRef] [PubMed]
  225. Rayahin, J.E.; Buhrman, J.S.; Zhang, Y.; Koh, T.J.; Gemeinhart, R.A. High and Low Molecular Weight Hyaluronic Acid Differentially Influence Macrophage Activation. ACS Biomater. Sci. Eng. 2015, 1, 481–493. [Google Scholar] [CrossRef]
  226. Hu, L.; Nomura, S.; Sato, Y.; Takagi, K.; Ishii, T.; Honma, Y.; Watanabe, K.; Mizukami, Y.; Muto, J. Anti-Inflammatory Effects of Differential Molecular Weight Hyaluronic Acids on UVB-Induced Calprotectin-Mediated Keratinocyte Inflammation. J. Dermatol. Sci. 2022, 107, 24–31. [Google Scholar] [CrossRef]
  227. Luan, X.; Cong, Z.; Anastassiades, T.P.; Gao, Y. N-Butyrylated Hyaluronic Acid Achieves Anti-Inflammatory Effects In Vitro and in Adjuvant-Induced Immune Activation in Rats. Molecules 2022, 27, 3267. [Google Scholar] [CrossRef] [PubMed]
  228. Bowman, S.; Awad, M.E.; Hamrick, M.W.; Hunter, M.; Fulzele, S. Recent Advances in Hyaluronic Acid Based Therapy for Osteoarthritis. Clin. Transl. Med. 2018, 7, e6. [Google Scholar] [CrossRef] [PubMed]
  229. Chang, C.; Hsu, Y.; Chen, Y.; Lin, F.; Sadhasivam, S.; Loo, S.; Savitha, S. Anti-inflammatory Effects of Hydrophilic and Lipophilic Statins with Hyaluronic Acid against LPS-induced Inflammation in Porcine Articular Chondrocytes. J. Orthop. Res. 2014, 32, 557–565. [Google Scholar] [CrossRef]
  230. Lo, G.H.; LaValley, M.; McAlindon, T.; Felson, D.T. Intra-Articular Hyaluronic Acid in Treatment of Knee Osteoarthritis. JAMA 2003, 290, 3115. [Google Scholar] [CrossRef] [PubMed]
  231. Pavlovic, V.; Ciric, M.; Jovanovic, V.; Trandafilovic, M.; Stojanovic, P. Platelet-Rich Fibrin: Basics of Biological Actions and Protocol Modifications. Open Med. 2021, 16, 446–454. [Google Scholar] [CrossRef]
  232. Zhao, H.; Zhu, W.; Mao, W.; Shen, C. Platelet-Rich Plasma Inhibits Adriamycin-Induced Inflammation via Blocking the NF-ΚB Pathway in Articular Chondrocytes. Mol. Med. 2021, 27, 66. [Google Scholar] [CrossRef]
  233. van Buul, G.M.; Koevoet, W.L.M.; Kops, N.; Bos, P.K.; Verhaar, J.A.N.; Weinans, H.; Bernsen, M.R.; van Osch, G.J.V.M. Platelet-Rich Plasma Releasate Inhibits Inflammatory Processes in Osteoarthritic Chondrocytes. Am. J. Sports Med. 2011, 39, 2362–2370. [Google Scholar] [CrossRef] [PubMed]
  234. Xin, F.; Wang, H.; Yuan, F.; Ding, Y.; Pabelick, C. Platelet-Rich Plasma Combined with Alendronate Reduces Pain and Inflammation in Induced Osteoarthritis in Rats by Inhibiting the Nuclear Factor-Kappa B Signaling Pathway. Biomed. Res. Int. 2020, 2020, 8070295. [Google Scholar] [CrossRef] [PubMed]
  235. Asjid, R.; Faisal, T.; Qamar, K.; Khan, S.A.; Khalil, A.; Zia, M.S. Platelet-Rich Plasma-Induced Inhibition of Chondrocyte Apoptosis Directly Affects Cartilage Thickness in Osteoarthritis. Cureus 2019, 11, e6050. [Google Scholar] [CrossRef]
  236. Belk, J.W.; Kraeutler, M.J.; Houck, D.A.; Goodrich, J.A.; Dragoo, J.L.; McCarty, E.C. Platelet-Rich Plasma Versus Hyaluronic Acid for Knee Osteoarthritis: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Am. J. Sports Med. 2021, 49, 249–260. [Google Scholar] [CrossRef] [PubMed]
  237. Chen, L.; Jin, S.; Yao, Y.; He, S.; He, J. Comparison of Clinical Efficiency between Intra-Articular Injection of Platelet-Rich Plasma and Hyaluronic Acid for Osteoarthritis: A Meta-Analysis of Randomized Controlled Trials. Ther. Adv. Musculoskelet. Dis. 2023, 15, 1759720X2311570. [Google Scholar] [CrossRef]
  238. Karasavvidis, T.; Totlis, T.; Gilat, R.; Cole, B.J. Platelet-Rich Plasma Combined with Hyaluronic Acid Improves Pain and Function Compared with Hyaluronic Acid Alone in Knee Osteoarthritis: A Systematic Review and Meta-Analysis. Arthrosc. J. Arthrosc. Relat. Surg. 2021, 37, 1277–1287.e1. [Google Scholar] [CrossRef] [PubMed]
  239. Proffen, B.; Vavken, P.; Dorotka, R. Surgical Management of Osteoarthritis. Wien. Med. Wochenschr. 2013, 163, 243–250. [Google Scholar] [CrossRef] [PubMed]
  240. Madry, H. Surgical Therapy in Osteoarthritis. Osteoarthr. Cartil. 2022, 30, 1019–1034. [Google Scholar] [CrossRef] [PubMed]
  241. Deveza, L.A.; Hunter, D.J.; Wajon, A.; Bennell, K.L.; Vicenzino, B.; Hodges, P.; Eyles, J.P.; Jongs, R.; Riordan, E.A.; Duong, V.; et al. Efficacy of Combined Conservative Therapies on Clinical Outcomes in Patients with Thumb Base Osteoarthritis: Protocol for a Randomised, Controlled Trial (COMBO). BMJ Open 2017, 7, e014498. [Google Scholar] [CrossRef]
  242. Kerzner, B.; Fortier, L.M.; Swindell, H.W.; McCormick, J.R.; Kasson, L.B.; Hevesi, M.; LaPrade, R.F.; Mandelbaum, B.R.; Chahla, J. An Update on the Use of Orthobiologics Combined with Corrective Osteotomies for Osteoarthritis: Osteotomy Site and Intra-Articular Efficacy. Oper. Tech. Sports Med. 2022, 30, 150933. [Google Scholar] [CrossRef]
  243. Sabzevari, S.; Ebrahimpour, A.; Roudi, M.K.; Kachooei, A.R. High Tibial Osteotomy: A Systematic Review and Current Concept. Arch. Bone Jt. Surg. 2016, 4, 204–212. [Google Scholar] [PubMed]
  244. Peng, H.; Ou, A.; Huang, X.; Wang, C.; Wang, L.; Yu, T.; Zhang, Y.; Zhang, Y. Osteotomy Around the Knee: The Surgical Treatment of Osteoarthritis. Orthop. Surg. 2021, 13, 1465–1473. [Google Scholar] [CrossRef] [PubMed]
  245. Komura, S.; Hirakawa, A.; Hirose, H.; Akiyama, H. Minimally Invasive Arthroscopy-Assisted Arthrodesis for Thumb Carpometacarpal Osteoarthritis. Arch. Orthop. Trauma. Surg. 2023, 144, 967–974. [Google Scholar] [CrossRef] [PubMed]
  246. Herrera-Pérez, M.; Valderrabano, V.; Godoy-Santos, A.L.; de César Netto, C.; González-Martín, D.; Tejero, S. Ankle Osteoarthritis: Comprehensive Review and Treatment Algorithm Proposal. EFORT Open Rev. 2022, 7, 448–459. [Google Scholar] [CrossRef] [PubMed]
  247. Brumat, P.; Kunšič, O.; Novak, S.; Slokar, U.; Pšenica, J.; Topolovec, M.; Mihalič, R.; Trebše, R. The Surgical Treatment of Osteoarthritis. Life 2022, 12, 982. [Google Scholar] [CrossRef] [PubMed]
  248. Briggs, K.K.; Bolia, I.K. Hip Arthroscopy: An Evidence-Based Approach. Lancet 2018, 391, 2189–2190. [Google Scholar] [CrossRef] [PubMed]
  249. Cho, S.-M.; Kim, S.-H.; Ha, S.-K.; Kim, S.-D.; Lim, D.-J.; Cha, J.; Kim, B.-J. Paraspinal Muscle Changes after Single-Level Posterior Lumbar Fusion: Volumetric Analyses and Literature Review. BMC Musculoskelet. Disord. 2020, 21, 73. [Google Scholar] [CrossRef]
  250. Okuda, S.; Yamashita, T.; Matsumoto, T.; Nagamoto, Y.; Sugiura, T.; Takahashi, Y.; Maeno, T.; Iwasaki, M. Adjacent Segment Disease After Posterior Lumbar Interbody Fusion: A Case Series of 1000 Patients. Glob. Spine J. 2018, 8, 722–727. [Google Scholar] [CrossRef]
  251. Martinez-Catalan, N.; Sanchez-Sotelo, J. Primary Elbow Osteoarthritis: Evaluation and Management. J. Clin. Orthop. Trauma. 2021, 19, 67–74. [Google Scholar] [CrossRef]
  252. Zhu, S.L.; Chin, B.; Sarraj, M.; Wang, E.; Dunn, E.E.; McRae, M.C. Denervation as a Treatment for Arthritis of the Hands: A Systematic Review of the Current Literature. HAND 2023, 18, 183–191. [Google Scholar] [CrossRef]
  253. Lee, D.H.; Kim, S.J.; Kim, S.A.; Ju, G. Past, Present, and Future of Cartilage Restoration: From Localized Defect to Arthritis. Knee Surg. Relat. Res. 2022, 34, 1. [Google Scholar] [CrossRef] [PubMed]
  254. Tischer, T.; Paul, J.; Pape, D.; Hirschmann, M.T.; Imhoff, A.B.; Hinterwimmer, S.; Feucht, M.J. The Impact of Osseous Malalignment and Realignment Procedures in Knee Ligament Surgery: A Systematic Review of the Clinical Evidence. Orthop. J. Sports Med. 2017, 5, 232596711769728. [Google Scholar] [CrossRef] [PubMed]
  255. Rönn, K.; Reischl, N.; Gautier, E.; Jacobi, M. Current Surgical Treatment of Knee Osteoarthritis. Arthritis 2011, 2011, 454873. [Google Scholar] [CrossRef] [PubMed]
  256. Charnley, J. Anchorage Of The Femoral Head Prosthesis To The Shaft Of The Femur. J. Bone Jt. Surg. Br. 1960, 42, 28–30. [Google Scholar] [CrossRef] [PubMed]
  257. Charnley, J. Arthroplasty Of The Hip: A New Operation. Lancet 1961, 277, 1129–1132. [Google Scholar] [CrossRef]
  258. Katz, J.N.; Earp, B.E.; Gomoll, A.H. Surgical Management of Osteoarthritis. Arthritis Care Res. 2010, 62, 1220–1228. [Google Scholar] [CrossRef]
  259. van Adrichem, R.A.; Nelissen, R.G.H.H.; Schipper, I.B.; Rosendaal, F.R.; Cannegieter, S.C. Risk of Venous Thrombosis after Arthroscopy of the Knee: Results from a Large Population-based Case–Control Study. J. Thromb. Haemost. 2015, 13, 1441–1448. [Google Scholar] [CrossRef] [PubMed]
  260. Chavalparit, P.; Chuaychoosakoon, C.; Parinyakhup, W.; Boonriong, T. Deep Vein Thrombosis Following Arthroscopic Meniscal Root Repair: A Case Report. Int. J. Surg. Case Rep. 2021, 85, 106193. [Google Scholar] [CrossRef]
  261. Papavasiliou, A.V.; Bardakos, N.V. Complications of Arthroscopic Surgery of the Hip. Bone Jt. Res. 2012, 1, 131–144. [Google Scholar] [CrossRef]
  262. Friberger Pajalic, K.; Turkiewicz, A.; Englund, M. Update on the Risks of Complications after Knee Arthroscopy. BMC Musculoskelet. Disord. 2018, 19, 179. [Google Scholar] [CrossRef]
  263. Shin, J.J.; Popchak, A.J.; Musahl, V.; Irrgang, J.J.; Lin, A. Complications After Arthroscopic Shoulder Surgery: A Review of the American Board of Orthopaedic Surgery Database. JAAOS Glob. Res. Rev. 2018, 2, e093. [Google Scholar] [CrossRef] [PubMed]
  264. Goodman, S.B.; Gallo, J. Periprosthetic Osteolysis: Mechanisms, Prevention and Treatment. J. Clin. Med. 2019, 8, 2091. [Google Scholar] [CrossRef]
  265. Zhang, R.; Lin, J.; Chen, F.; Chen, M. Worldwide Trends of Research on Periprosthetic Osteolysis: A Bibliometric Study Based on VOSviewer. Indian J. Orthop. 2021, 55, 1326–1334. [Google Scholar] [CrossRef]
  266. Evans, J.T.; Walker, R.W.; Evans, J.P.; Blom, A.W.; Sayers, A.; Whitehouse, M.R. How Long Does a Knee Replacement Last? A Systematic Review and Meta-Analysis of Case Series and National Registry Reports with More than 15 Years of Follow-Up. Lancet 2019, 393, 655–663. [Google Scholar] [CrossRef]
  267. Jones, M.D.; Buckle, C.L. How Does Aseptic Loosening Occur and How Can We Prevent It? Orthop. Trauma. 2020, 34, 146–152. [Google Scholar] [CrossRef]
  268. Sharkey, P.F.; Lichstein, P.M.; Shen, C.; Tokarski, A.T.; Parvizi, J. Why Are Total Knee Arthroplasties Failing Today—Has Anything Changed After 10 Years? J. Arthroplast. 2014, 29, 1774–1778. [Google Scholar] [CrossRef] [PubMed]
  269. Siddiqi, A.; Kamath, A.F. Aseptic Loosening—A US Perspective. In Essentials of Cemented Knee Arthroplasty; Springer: Berlin/Heidelberg, Germany, 2022; pp. 587–601. [Google Scholar]
  270. Feng, X.; Gu, J.; Zhou, Y. Primary Total Hip Arthroplasty Failure: Aseptic Loosening Remains the Most Common Cause of Revision. Am. J. Transl. Res. 2022, 14, 7080–7089. [Google Scholar] [PubMed]
  271. Sheth, N.P.; Rozell, J.C.; Paprosky, W.G. Evaluation and Treatment of Patients with Acetabular Osteolysis After Total Hip Arthroplasty. J. Am. Acad. Orthop. Surg. 2019, 27, e258–e267. [Google Scholar] [CrossRef]
  272. Kulkarni, P.G.; Paudel, N.; Magar, S.; Santilli, M.F.; Kashyap, S.; Baranwal, A.K.; Zamboni, P.; Vasavada, P.; Katiyar, A.; Singh, A.V. Overcoming Challenges and Innovations in Orthopedic Prosthesis Design: An Interdisciplinary Perspective. Biomed. Mater. Devices 2023, 2, 58–69. [Google Scholar] [CrossRef]
  273. Shah, R.; Gashi, B.; Hoque, S.; Marian, M.; Rosenkranz, A. Enhancing Mechanical and Biomedical Properties of Protheses—Surface and Material Design. Surf. Interfaces 2021, 27, 101498. [Google Scholar] [CrossRef]
  274. Neogi, T.; Li, S.; Peloquin, C.; Misra, D.; Zhang, Y. Effect of Bisphosphonates on Knee Replacement Surgery. Ann. Rheum. Dis. 2018, 77, 92–97. [Google Scholar] [CrossRef] [PubMed]
  275. McDonald, C.L.; Lemme, N.J.; Testa, E.J.; Aaron, R.; Hartnett, D.A.; Cohen, E.M. Bisphosphonates in Total Joint Arthroplasty: A Review of Their Use and Complications. Arthroplast. Today 2022, 14, 133–139. [Google Scholar] [CrossRef] [PubMed]
  276. Fu, J.-N.; Wang, X.; Yang, M.; Chen, Y.-R.; Zhang, J.-Y.; Deng, R.-H.; Zhang, Z.-N.; Yu, J.-K.; Yuan, F.-Z. Scaffold-Based Tissue Engineering Strategies for Osteochondral Repair. Front. Bioeng. Biotechnol. 2022, 9, 812383. [Google Scholar] [CrossRef] [PubMed]
  277. Deng, C.; Zhou, Q.; Zhang, M.; Li, T.; Chen, H.; Xu, C.; Feng, Q.; Wang, X.; Yin, F.; Cheng, Y.; et al. Bioceramic Scaffolds with Antioxidative Functions for ROS Scavenging and Osteochondral Regeneration. Adv. Sci. 2022, 9, 2105727. [Google Scholar] [CrossRef] [PubMed]
  278. Donate, R.; Tamaddon, M.; Ribeiro, V.; Monzón, M.; Oliveira, J.M.; Liu, C. Translation through Collaboration: Practice Applied in BAMOS Project in In Vivo Testing of Innovative Osteochondral Scaffolds. Biomater. Transl. 2022, 3, 102–104. [Google Scholar] [PubMed]
  279. van Walsem, A.; Pandhi, S.; Nixon, R.M.; Guyot, P.; Karabis, A.; Moore, R.A. Relative Benefit-Risk Comparing Diclofenac to Other Traditional Non-Steroidal Anti-Inflammatory Drugs and Cyclooxygenase-2 Inhibitors in Patients with Osteoarthritis or Rheumatoid Arthritis: A Network Meta-Analysis. Arthritis Res. Ther. 2015, 17, 66. [Google Scholar] [CrossRef] [PubMed]
  280. Baigent, C.; Bhala, N.; Emberson, J.; Merhi, A.; Abramson, S.; Arber, N.; Baron, J.A.; Bombardier, C.; Cannon, C.; Farkouh, M.E.; et al. Vascular and Upper Gastrointestinal Effects of Non-Steroidal Anti-Inflammatory Drugs: Meta-Analyses of Individual Participant Data from Randomised Trials. Lancet 2013, 382, 769–779. [Google Scholar] [CrossRef] [PubMed]
  281. Zhang, X.; Donnan, P.T.; Bell, S.; Guthrie, B. Non-Steroidal Anti-Inflammatory Drug Induced Acute Kidney Injury in the Community Dwelling General Population and People with Chronic Kidney Disease: Systematic Review and Meta-Analysis. BMC Nephrol. 2017, 18, 256. [Google Scholar] [CrossRef]
  282. Curtis, E.; Fuggle, N.; Shaw, S.; Spooner, L.; Ntani, G.; Parsons, C.; Corp, N.; Honvo, G.; Baird, J.; Maggi, S.; et al. Safety of Cyclooxygenase-2 Inhibitors in Osteoarthritis: Outcomes of a Systematic Review and Meta-Analysis. Drugs Aging 2019, 36, 25–44. [Google Scholar] [CrossRef]
  283. Lanas, A.; Tornero, J.; Zamorano, J.L. Assessment of Gastrointestinal and Cardiovascular Risk in Patients with Osteoarthritis Who Require NSAIDs: The LOGICA Study. Ann. Rheum. Dis. 2010, 69, 1453–1458. [Google Scholar] [CrossRef]
  284. Bally, M.; Dendukuri, N.; Rich, B.; Nadeau, L.; Helin-Salmivaara, A.; Garbe, E.; Brophy, J.M. Risk of Acute Myocardial Infarction with NSAIDs in Real World Use: Bayesian Meta-Analysis of Individual Patient Data. BMJ 2017, 357, 1909. [Google Scholar] [CrossRef] [PubMed]
  285. Ungprasert, P.; Cheungpasitporn, W.; Crowson, C.S.; Matteson, E.L. Individual Non-Steroidal Anti-Inflammatory Drugs and Risk of Acute Kidney Injury: A Systematic Review and Meta-Analysis of Observational Studies. Eur. J. Intern. Med. 2015, 26, 285–291. [Google Scholar] [CrossRef] [PubMed]
  286. Puri, V.; Nagpal, M.; Singh, I.; Singh, M.; Dhingra, G.A.; Huanbutta, K.; Dheer, D.; Sharma, A.; Sangnim, T. A Comprehensive Review on Nutraceuticals: Therapy Support and Formulation Challenges. Nutrients 2022, 14, 4637. [Google Scholar] [CrossRef] [PubMed]
  287. Gonçalves, R.F.S.; Martins, J.T.; Duarte, C.M.M.; Vicente, A.A.; Pinheiro, A.C. Advances in Nutraceutical Delivery Systems: From Formulation Design for Bioavailability Enhancement to Efficacy and Safety Evaluation. Trends Food Sci. Technol. 2018, 78, 270–291. [Google Scholar] [CrossRef]
  288. Wang, Z.; Jones, G.; Blizzard, L.; Aitken, D.; Zhou, Z.; Wang, M.; Balogun, S.; Cicuttini, F.; Antony, B. Prevalence and Correlates of the Use of Complementary and Alternative Medicines among Older Adults with Joint Pain. Int. J. Rheum. Dis. 2023, 26, 1760–1769. [Google Scholar] [CrossRef] [PubMed]
  289. 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, 20892. [Google Scholar] [CrossRef] [PubMed]
  290. Pacifico, S.; Piccolella, S.; Nocera, P.; Tranquillo, E.; Dal Poggetto, F.; Catauro, M. New Insights into Phenol and Polyphenol Composition of Stevia Rebaudiana Leaves. J. Pharm. Biomed. Anal. 2019, 163, 45–57. [Google Scholar] [CrossRef]
  291. Sandoval-Acuña, C.; Ferreira, J.; Speisky, H. Polyphenols and Mitochondria: An Update on Their Increasingly Emerging ROS-Scavenging Independent Actions. Arch. Biochem. Biophys. 2014, 559, 75–90. [Google Scholar] [CrossRef]
  292. Khan, H.; Ullah, H.; Castilho, P.C.M.F.; Gomila, A.S.; D’Onofrio, G.; Filosa, R.; Wang, F.; Nabavi, S.M.; Daglia, M.; Silva, A.S.; et al. Targeting NF-ΚB Signaling Pathway in Cancer by Dietary Polyphenols. Crit. Rev. Food Sci. Nutr. 2020, 60, 2790–2800. [Google Scholar] [CrossRef]
  293. Sharma, S.; Sahu, D.; Das, H.R.; Sharma, D. Amelioration of Collagen-Induced Arthritis by Salix Nigra Bark Extract via Suppression of pro-Inflammatory Cytokines and Oxidative Stress. Food Chem. Toxicol. 2011, 49, 3395–3406. [Google Scholar] [CrossRef]
  294. Blain, E.J.; Ali, A.Y.; Duance, V.C. Boswellia Frereana (Frankincense) Suppresses Cytokine-Induced Matrix Metalloproteinase Expression and Production of pro-Inflammatory Molecules in Articular Cartilage. Phytother. Res. 2010, 24, 905–912. [Google Scholar] [CrossRef] [PubMed]
  295. Mülek, M.; Seefried, L.; Genest, F.; Högger, P. Distribution of Constituents and Metabolites of Maritime Pine Bark Extract (Pycnogenol®) into Serum, Blood Cells, and Synovial Fluid of Patients with Severe Osteoarthritis: A Randomized Controlled Trial. Nutrients 2017, 9, 443. [Google Scholar] [CrossRef] [PubMed]
  296. Henrotin, Y.; Clutterbuck, A.L.; Allaway, D.; Lodwig, E.M.; Harris, P.; Mathy-Hartert, M.; Shakibaei, M.; Mobasheri, A. Biological Actions of Curcumin on Articular Chondrocytes. Osteoarthr. Cartil. 2010, 18, 141–149. [Google Scholar] [CrossRef] [PubMed]
  297. Kuptniratsaikul, V.; Dajpratham, P.; Taechaarpornkul, W.; Buntragulpoontawee, M.; Lukkanapichonchut, P.; Chootip, C.; Saengsuwan, J.; Tantayakom, K.; Laongpech, S. Efficacy and Safety of Curcuma domestica Extracts Compared with Ibuprofen in Patients with Knee Osteoarthritis: A Multicenter Study. Clin. Interv. Aging 2014, 9, 451–458. [Google Scholar] [CrossRef] [PubMed]
  298. Henrotin, Y.; Malaise, M.; Wittoek, R.; de Vlam, K.; Brasseur, J.-P.; Luyten, F.P.; Jiangang, Q.; Van den Berghe, M.; Uhoda, R.; Bentin, J.; et al. Bio-Optimized Curcuma Longa Extract Is Efficient on Knee Osteoarthritis Pain: A Double-Blind Multicenter Randomized Placebo Controlled Three-Arm Study. Arthritis Res. Ther. 2019, 21, 179. [Google Scholar] [CrossRef] [PubMed]
  299. Henrotin, Y.; Gharbi, M.; Dierckxsens, Y.; Priem, F.; Marty, M.; Seidel, L.; Albert, A.; Heuse, E.; Bonnet, V.; Castermans, C. Decrease of a Specific Biomarker of Collagen Degradation in Osteoarthritis, Coll2-1, by Treatment with Highly Bioavailable Curcumin during an Exploratory Clinical Trial. BMC Complement. Altern. Med. 2014, 14, 159. [Google Scholar] [CrossRef] [PubMed]
  300. Javadi, F.; Ahmadzadeh, A.; Eghtesadi, S.; Aryaeian, N.; Zabihiyeganeh, M.; Rahimi Foroushani, A.; Jazayeri, S. The Effect of Quercetin on Inflammatory Factors and Clinical Symptoms in Women with Rheumatoid Arthritis: A Double-Blind, Randomized Controlled Trial. J. Am. Coll. Nutr. 2017, 36, 9–15. [Google Scholar] [CrossRef] [PubMed]
  301. Liczbiński, P.; Michałowicz, J.; Bukowska, B. Molecular Mechanism of Curcumin Action in Signaling Pathways: Review of the Latest Research. Phytother. Res. 2020, 34, 1992–2005. [Google Scholar] [CrossRef] [PubMed]
  302. Hatcher, H.; Planalp, R.; Cho, J.; Torti, F.M.; Torti, S.V. Curcumin: From Ancient Medicine to Current Clinical Trials. Cell. Mol. Life Sci. 2008, 65, 1631–1652. [Google Scholar] [CrossRef]
  303. Zaman, M.S.; Chauhan, N.; Yallapu, M.M.; Gara, R.K.; Maher, D.M.; Kumari, S.; Sikander, M.; Khan, S.; Zafar, N.; Jaggi, M.; et al. Curcumin Nanoformulation for Cervical Cancer Treatment. Sci. Rep. 2016, 6, 20051. [Google Scholar] [CrossRef]
  304. Hu, S.; Xu, Y.; Meng, L.; Huang, L.; Sun, H. Curcumin Inhibits Proliferation and Promotes Apoptosis of Breast Cancer Cells. Exp. Ther. Med. 2018, 16, 1266–1272. [Google Scholar] [CrossRef] [PubMed]
  305. Mehta, H.J.; Patel, V.; Sadikot, R.T. Curcumin and Lung Cancer—A Review. Target. Oncol. 2014, 9, 295–310. [Google Scholar] [CrossRef] [PubMed]
  306. Sun, X.D.; Liu, X.E.; Huang, D.S. Curcumin Reverses the Epithelial-Mesenchymal Transition of Pancreatic Cancer Cells by Inhibiting the Hedgehog Signaling Pathway. Oncol. Rep. 2013, 29, 2401–2407. [Google Scholar] [CrossRef]
  307. Clutterbuck, A.L.; Mobasheri, A.; Shakibaei, M.; Allaway, D.; Harris, P. Interleukin-1β–Induced Extracellular Matrix Degradation and Glycosaminoglycan Release Is Inhibited by Curcumin in an Explant Model of Cartilage Inflammation. Ann. N. Y. Acad. Sci. 2009, 1171, 428–435. [Google Scholar] [CrossRef] [PubMed]
  308. Csaki, C.; Mobasheri, A.; Shakibaei, M. Synergistic Chondroprotective Effects of Curcumin and Resveratrol in Human Articular Chondrocytes: Inhibition of IL-1β-Induced NF-ΚB-Mediated Inflammation and Apoptosis. Arthritis Res. Ther. 2009, 11, R165. [Google Scholar] [CrossRef] [PubMed]
  309. Buhrmann, C.; Mobasheri, A.; Matis, U.; Shakibaei, M. Curcumin Mediated Suppression of Nuclear Factor-ΚB Promotes Chondrogenic Differentiation of Mesenchymal Stem Cells in a High-Density Co-Culture Microenvironment. Arthritis Res. Ther. 2010, 12, R127. [Google Scholar] [CrossRef] [PubMed]
  310. Shakibaei, M.; Schulze-Tanzil, G.; John, T.; Mobasheri, A. Curcumin Protects Human Chondrocytes from IL-L1beta-Induced Inhibition of Collagen Type II and Beta1-Integrin Expression and Activation of Caspase-3: An Immunomorphological Study. Ann. Anat. 2005, 187, 487–497. [Google Scholar] [CrossRef] [PubMed]
  311. Kloesch, B.; Becker, T.; Dietersdorfer, E.; Kiener, H.; Steiner, G. Anti-Inflammatory and Apoptotic Effects of the Polyphenol Curcumin on Human Fibroblast-like Synoviocytes. Int. Immunopharmacol. 2013, 15, 400–405. [Google Scholar] [CrossRef] [PubMed]
  312. Anjomshoa, S.; Namazian, M.; Noorbala, M.R. Is Curcumin a Good Scavenger of Reactive Oxygen Species? A Computational Investigation. Theor. Chem. Acc. 2017, 136, 103. [Google Scholar] [CrossRef]
  313. Barzegar, A.; Moosavi-Movahedi, A.A. Intracellular ROS Protection Efficiency and Free Radical-Scavenging Activity of Curcumin. PLoS ONE 2011, 6, e26012. [Google Scholar] [CrossRef]
  314. Chen, B.; He, Q.; Chen, C.; Lin, Y.; Xiao, J.; Pan, Z.; Li, M.; Li, S.; Yang, J.; Wang, F.C.; et al. Combination of Curcumin and Catalase Protects against Chondrocyte Injury and Knee Osteoarthritis Progression by Suppressing Oxidative Stress. Biomed. Pharmacother. 2023, 168, 115751. [Google Scholar] [CrossRef]
  315. Crivelli, B.; Bari, E.; Perteghella, S.; Catenacci, L.; Sorrenti, M.; Mocchi, M.; Faragò, S.; Tripodo, G.; Prina-Mello, A.; Torre, M.L. Silk Fibroin Nanoparticles for Celecoxib and Curcumin Delivery: ROS-Scavenging and Anti-Inflammatory Activities in an In Vitro Model of Osteoarthritis. Eur. J. Pharm. Biopharm. 2019, 137, 37–45. [Google Scholar] [CrossRef]
  316. 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]
  317. Zhang, G.; Cao, J.; Yang, E.; Liang, B.; Ding, J.; Liang, J.; Xu, J. Curcumin Improves Age-Related and Surgically Induced Osteoarthritis by Promoting Autophagy in Mice. Biosci. Rep. 2018, 38, 20171691. [Google Scholar] [CrossRef]
  318. Bannuru, R.R.; Osani, M.C.; Al-Eid, F.; Wang, C. Efficacy of Curcumin and Boswellia for Knee Osteoarthritis: Systematic Review and Meta-Analysis. Semin. Arthritis Rheum. 2018, 48, 416–429. [Google Scholar] [CrossRef] [PubMed]
  319. Haroyan, A.; Mukuchyan, V.; Mkrtchyan, N.; Minasyan, N.; Gasparyan, S.; Sargsyan, A.; Narimanyan, M.; Hovhannisyan, A. Efficacy and Safety of Curcumin and Its Combination with Boswellic Acid in Osteoarthritis: A Comparative, Randomized, Double-Blind, Placebo-Controlled Study. BMC Complement. Altern. Med. 2018, 18, 7. [Google Scholar] [CrossRef] [PubMed]
  320. Yabas, M.; Orhan, C.; Er, B.; Tuzcu, M.; Durmus, A.S.; Ozercan, I.H.; Sahin, N.; Bhanuse, P.; Morde, A.A.; Padigaru, M.; et al. A Next Generation Formulation of Curcumin Ameliorates Experimentally Induced Osteoarthritis in Rats via Regulation of Inflammatory Mediators. Front. Immunol. 2021, 12, 609629. [Google Scholar] [CrossRef]
  321. Mazorra-Manzano, M.A.; Ramírez-Suarez, J.C.; Yada, R.Y. Plant Proteases for Bioactive Peptides Release: A Review. Crit. Rev. Food Sci. Nutr. 2018, 58, 2147–2163. [Google Scholar] [CrossRef]
  322. Lafarga, T.; Gallagher, E.; Aluko, R.E.; Auty, M.A.E.; Hayes, M. Addition of an Enzymatic Hydrolysate of Bovine Globulins to Bread and Determination of Hypotensive Effects in Spontaneously Hypertensive Rats. J. Agric. Food Chem. 2016, 64, 1741–1750. [Google Scholar] [CrossRef] [PubMed]
  323. Zarei, M.; Ebrahimpour, A.; Abdul-Hamid, A.; Anwar, F.; Saari, N. Production of Defatted Palm Kernel Cake Protein Hydrolysate as a Valuable Source of Natural Antioxidants. Int. J. Mol. Sci. 2012, 13, 8097–8111. [Google Scholar] [CrossRef]
  324. Memarpoor-Yazdi, M.; Asoodeh, A.; Chamani, J.K. A Novel Antioxidant and Antimicrobial Peptide from Hen Egg White Lysozyme Hydrolysates. J. Funct. Foods 2012, 4, 278–286. [Google Scholar] [CrossRef]
  325. Gajanan, P.G.; Elavarasan, K.; Shamasundar, B.A. Bioactive and Functional Properties of Protein Hydrolysates from Fish Frame Processing Waste Using Plant Proteases. Environ. Sci. Pollut. Res. 2016, 23, 24901–24911. [Google Scholar] [CrossRef]
  326. Zhi, N.N.; Zong, K.; Jia, X.Y.; Wang, L.; Liang, J. Effect of High Pressure Processing on Fibrinolytic Activity of Fruit Bromelain In Vivo. J. Food Process Eng. 2019, 42, e13146. [Google Scholar] [CrossRef]
  327. Muhammad, Z.A.; Ahmad, T. Therapeutic Uses of Pineapple-Extracted Bromelain in Surgical Care—A Review. JPMA J. Pak. Med. Assoc. 2017, 67, 121–125. [Google Scholar] [PubMed]
  328. Rathnavelu, V.; Alitheen, N.B.; Sohila, S.; Kanagesan, S.; Ramesh, R. Potential Role of Bromelain in Clinical and Therapeutic Applications. Biomed. Rep. 2016, 5, 283. [Google Scholar] [CrossRef] [PubMed]
  329. Brien, S.; Lewith, G.; Walker, A.; Hicks, S.M.; Middleton, D. Bromelain as a Treatment for Osteoarthritis: A Review of Clinical Studies. Evid.-Based Complement. Altern. Med. 2004, 1, 251–257. [Google Scholar] [CrossRef] [PubMed]
  330. Brien, S.; Lewith, G.; Walker, A.F.; Middleton, R.; Prescott, P.; Bundy, R. Bromelain as an Adjunctive Treatment for Moderate-to-Severe Osteoarthritis of the Knee: A Randomized Placebo-Controlled Pilot Study. QJM Int. J. Med. 2006, 99, 841–850. [Google Scholar] [CrossRef] [PubMed]
  331. Kasemsuk, T.; Saengpetch, N.; Sibmooh, N.; Unchern, S. Improved WOMAC Score Following 16-Week Treatment with Bromelain for Knee Osteoarthritis. Clin. Rheumatol. 2016, 35, 2531–2540. [Google Scholar] [CrossRef]
  332. Conrozier, T.; Mathieu, P.; Bonjean, M.; Marc, J.; Renevier, J.; Balblanc, J. A Complex of Three Natural Anti-Inflammatory Agents Provides Relief of Osteoarthritis Pain. Altern. Ther. Health Med. 2014, 20 (Suppl. S1), 32–37. [Google Scholar]
  333. Italiano, G.; Raimondo, M.; Giannetti, G.; Gargiulo, A. Benefits of a Food Supplement Containing Boswellia Serrata and Bromelain for Improving the Quality of Life in Patients with Osteoarthritis: A Pilot Study. J. Altern. Complement. Med. 2020, 26, 123–129. [Google Scholar] [CrossRef]
  334. Jayachandran, S.; Khobre, P. Efficacy of Bromelain along with Trypsin, Rutoside Trihydrate Enzymes and Diclofenac Sodium Combination Therapy for the Treatment of TMJ Osteoarthritis—A Randomised Clinical Trial. J. Clin. Diagn. Res. 2017, 11, ZC09–ZC11. [Google Scholar] [CrossRef] [PubMed]
  335. Pothacharoen, P.; Chaiwongsa, R.; Chanmee, T.; Insuan, O.; Wongwichai, T.; Janchai, P.; Vaithanomsat, P. Bromelain Extract Exerts Antiarthritic Effects via Chondroprotection and the Suppression of TNF-α–Induced NF-ΚB and MAPK Signaling. Plants 2021, 10, 2273. [Google Scholar] [CrossRef] [PubMed]
  336. Brochard, S.; Pontin, J.; Bernay, B.; Boumediene, K.; Conrozier, T.; Baugé, C. The Benefit of Combining Curcumin, Bromelain and Harpagophytum to Reduce Inflammation in Osteoarthritic Synovial Cells. BMC Complement. Med. Ther. 2021, 21, 261. [Google Scholar] [CrossRef] [PubMed]
  337. Quarta, S.; Santarpino, G.; Carluccio, M.A.; Calabriso, N.; Scoditti, E.; Siculella, L.; Damiano, F.; Maffia, M.; Verri, T.; De Caterina, R.; et al. Analysis of the Anti-Inflammatory and Anti-Osteoarthritic Potential of Flonat Fast®, a Combination of Harpagophytum Procumbens DC. Ex Meisn., Boswellia Serrata Roxb., Curcuma Longa L., Bromelain and Escin (Aesculus hippocastanum), Evaluated in In Vitro Mo. Pharmaceuticals 2022, 15, 1263. [Google Scholar] [CrossRef] [PubMed]
  338. Ammon, H.P.T. Modulation of the Immune System by Boswellia Serrata Extracts and Boswellic Acids. Phytomedicine 2010, 17, 862–867. [Google Scholar] [CrossRef] [PubMed]
  339. Abdel-Tawab, M.; Werz, O.; Schubert-Zsilavecz, M. Boswellia Serrata: An Overall Assessment of In Vitro, Preclinical, Pharmacokinetic and Clinical Data. Clin. Pharmacokinet. 2011, 50, 349–369. [Google Scholar] [CrossRef] [PubMed]
  340. Catanzaro, D.; Rancan, S.; Orso, G.; Dall’acqua, S.; Brun, P.; Giron, M.C.; Carrara, M.; Castagliuolo, I.; Ragazzi, E.; Caparrotta, L.; et al. Boswellia Serrata Preserves Intestinal Epithelial Barrier from Oxidative and Inflammatory Damage. PLoS ONE 2015, 10, e0125375. [Google Scholar] [CrossRef] [PubMed]
  341. Gupta, I.; Gupta, V.; Parihar, A.; Gupta, S.; Lüdtke, R.; Safayhi, H.; Ammon, H.P. Effects of Boswellia Serrata Gum Resin in Patients with Bronchial Asthma: Results of a Double-Blind, Placebo-Controlled, 6-Week Clinical Study. Eur. J. Med. Res. 1998, 3, 511–514. [Google Scholar] [PubMed]
  342. Streffer, J.R.; Bitzer, M.; Schabet, M.; Dichgans, J.; Weller, M. Response of Radiochemotherapy-Associated Cerebral Edema to a Phytotherapeutic Agent, H15. Neurology 2001, 56, 1219–1221. [Google Scholar] [CrossRef]
  343. Sengupta, K.; Alluri, K.V.; Satish, A.R.; Mishra, S.; Golakoti, T.; Sarma, K.V.S.; Dey, D.; Raychaudhuri, S.P. A Double Blind, Randomized, Placebo Controlled Study of the Efficacy and Safety of 5-Loxin® for Treatment of Osteoarthritis of the Knee. Arthritis Res. Ther. 2008, 10, R85. [Google Scholar] [CrossRef]
  344. Kimmatkar, N.; Thawani, V.; Hingorani, L.; Khiyani, R. Efficacy and Tolerability of Boswellia Serrata Extract in Treatment of Osteoarthritis of Knee—A Randomized Double Blind Placebo Controlled Trial. Phytomedicine 2003, 10, 3–7. [Google Scholar] [CrossRef] [PubMed]
  345. Karlapudi, V.; Sunkara, K.B.; Konda, P.R.; Sarma, K.V.; Rokkam, M.P. Efficacy and Safety of Aflapin®, a Novel Boswellia Serrata Extract, in the Treatment of Osteoarthritis of the Knee: A Short-Term 30-Day Randomized, Double-Blind, Placebo-Controlled Clinical Study. J. Am. Nutr. Assoc. 2023, 42, 159–168. [Google Scholar] [CrossRef] [PubMed]
  346. Majeed, M.; Majeed, S.; Narayanan, N.K.; Nagabhushanam, K. A Pilot, Randomized, Double-Blind, Placebo-Controlled Trial to Assess the Safety and Efficacy of a Novel Boswellia Serrata Extract in the Management of Osteoarthritis of the Knee. Phytother. Res. 2019, 33, 1457–1468. [Google Scholar] [CrossRef] [PubMed]
  347. Khoramjouy, M.; Bayanati, M.; Noori, S.; Faizi, M.; Zarghi, A. Effects of Ziziphus Jujuba Extract Alone and Combined with Boswellia Serrata Extract on Monosodium Iodoacetate Model of Osteoarthritis in Mice. Iran. J. Pharm. Res. 2022, 21, 134338. [Google Scholar] [CrossRef] [PubMed]
  348. Shin, M.R.; Kim, H.Y.; Choi, H.Y.; Park, K.S.; Choi, H.J.; Roh, S.S. Boswellia Serrata Extract, 5-Loxin®, Prevents Joint Pain and Cartilage Degeneration in a Rat Model of Osteoarthritis through Inhibition of Inflammatory Responses and Restoration of Matrix Homeostasis. Evid.-Based Complement. Altern. Med. 2022, 2022, 3067526. [Google Scholar] [CrossRef]
  349. Kulkarni, P.D.; Damle, N.D.; Singh, S.; Yadav, K.S.; Ghante, M.R.; Bhaskar, V.H.; Hingorani, L.; Gota, V.S. Double-Blind Trial of Solid Lipid Boswellia Serrata Particles (SLBSP) vs. Standardized Boswellia Serrata Gum Extract (BSE) for Osteoarthritis of Knee. Drug Metab. Pers. Ther. 2020, 35, 20200104. [Google Scholar] [CrossRef]
  350. Henrotin, Y.; Dierckxsens, Y.; Delisse, G.; Maes, N.; Albert, A. Curcuma Longa and Boswellia Serrata Extract Combination for Hand Osteoarthritis: An Open-Label Pre-Post Trial. Pharm. Biol. 2022, 60, 2295–2299. [Google Scholar] [CrossRef] [PubMed]
  351. Marefati, N.; Beheshti, F.; Memarpour, S.; Bayat, R.; Naser Shafei, M.; Sadeghnia, H.R.; Ghazavi, H.; Hosseini, M. The Effects of Acetyl-11-Keto-β-Boswellic Acid on Brain Cytokines and Memory Impairment Induced by Lipopolysaccharide in Rats. Cytokine 2020, 131, 155107. [Google Scholar] [CrossRef] [PubMed]
  352. Siemoneit, U.; Koeberle, A.; Rossi, A.; Dehm, F.; Verhoff, M.; Reckel, S.; Maier, T.J.; Jauch, J.; Northoff, H.; Bernhard, F.; et al. Inhibition of Microsomal Prostaglandin E2 Synthase-1 as a Molecular Basis for the Anti-Inflammatory Actions of Boswellic Acids from Frankincense. Br. J. Pharmacol. 2011, 162, 147–162. [Google Scholar] [CrossRef]
  353. Tausch, L.; Henkel, A.; Siemoneit, U.; Poeckel, D.; Kather, N.; Franke, L.; Hofmann, B.; Schneider, G.; Angioni, C.; Geisslinger, G.; et al. Identification of Human Cathepsin G As a Functional Target of Boswellic Acids from the Anti-Inflammatory Remedy Frankincense. J. Immunol. 2009, 183, 3433–3442. [Google Scholar] [CrossRef]
  354. Wang, H.; Zhang, C.; Wu, Y.; Ai, Y.; Lee, D.Y.W.; Dai, R. Comparative Pharmacokinetic Study of Two Boswellic Acids in Normal and Arthritic Rat Plasma after Oral Administration of Boswellia Serrata Extract or Huo Luo Xiao Ling Dan by LC-MS. Biomed. Chromatogr. 2014, 28, 1402–1408. [Google Scholar] [CrossRef] [PubMed]
  355. Huang, T.H.W.; Tran, V.H.; Duke, R.K.; Tan, S.; Chrubasik, S.; Roufogalis, B.D.; Duke, C.C. Harpagoside Suppresses Lipopolysaccharide-Induced INOS and COX-2 Expression through Inhibition of NF-ΚB Activation. J. Ethnopharmacol. 2006, 104, 149–155. [Google Scholar] [CrossRef] [PubMed]
  356. Schulze-Tanzil, G.; Hansen, C.; Shakibaei, M. Wirkung Des Extraktes Aus Harpagophytum Procumbens DC Auf Matrix-Metalloproteinasen in Menschlichen Knorpelzellen In Vitro. Arzneimittelforschung 2011, 54, 213–220. [Google Scholar] [CrossRef] [PubMed]
  357. Ncube, S.F.; McGaw, L.J.; Njoya, E.M.; Ndagurwa, H.G.T.; Mundy, P.J.; Sibanda, S. In Vitro Antioxidant Activity of Crude Extracts of Harpagophytum Zeyheri and Their Anti-Inflammatory and Cytotoxicity Activity Compared with Diclofenac. BMC Complement. Med. Ther. 2021, 21, 238. [Google Scholar] [CrossRef] [PubMed]
  358. Haseeb, A.; Ansari, M.Y.; Haqqi, T.M. Harpagoside Suppresses IL-6 Expression in Primary Human Osteoarthritis Chondrocytes. J. Orthop. Res. 2017, 35, 311–320. [Google Scholar] [CrossRef] [PubMed]
  359. Haseeb, A.; Leigh, D.; Haqqi, T.M. A Small Molecule Harpagoside Inhibits IL-1beta-Induced Expression of IL-6 by Blocking the Expression of C-FOS in Primary Human Osteoarthritis Chondrocytes. Osteoarthr. Cartil. 2015, 23, A155–A156. [Google Scholar] [CrossRef]
  360. Mariano, A.; Di Sotto, A.; Leopizzi, M.; Garzoli, S.; Di Maio, V.; Gullì, M.; Vedova, P.D.; Ammendola, S.; D’Abusco, A.S. Antiarthritic Effects of a Root Extract from Harpagophytum Procumbens DC: Novel Insights into the Molecular Mechanisms and Possible Bioactive Phytochemicals. Nutrients 2020, 12, 2545. [Google Scholar] [CrossRef] [PubMed]
  361. Farpour, H.R.; Rajabi, N.; Ebrahimi, B. The Efficacy of Harpagophytum Procumbens (Teltonal) in Patients with Knee Osteoarthritis: A Randomized Active-Controlled Clinical Trial. Evid. Based Complement. Altern. Med. 2021, 2021, 5596892. [Google Scholar] [CrossRef] [PubMed]
  362. Żȩgota, Z.; Goździk, J.; Głogowska-Szeląg, J. Prospective, Multicenter Evaluation of a Polyherbal Supplement alongside Standard-of-Care Treatment for Mild Knee Osteoarthritis. Adv. Orthop. 2021, 2021, 5589597. [Google Scholar] [CrossRef]
  363. Akhtar, N.; Haqqi, T.M. Current Nutraceuticals in the Management of Osteoarthritis: A Review. Ther. Adv. Musculoskelet. Dis. 2012, 4, 181–207. [Google Scholar] [CrossRef]
  364. Yang, Y.; Wang, L.; Yuan, M.; Yu, Q.; Fu, F. Anti-Inflammatory and Gastroprotective Effects of Escin. Nat. Prod. Commun. 2020, 15. [Google Scholar] [CrossRef]
  365. Wang, B.; Yang, R.; Ju, Q.; Liu, S.; Zhang, Y.; Ma, Y. Clinical Effects of Joint Application of β-Sodium Aescinate and Mannitol in Treating Early Swelling after Upper Limb Trauma Surgery. Exp. Ther. Med. 2016, 12, 3320–3322. [Google Scholar] [CrossRef] [PubMed]
  366. Idris, S.; Mishra, A.; Khushtar, M. Phytochemical, Ethanomedicinal and Pharmacological Applications of Escin from Aesculus hippocastanum L. Towards Future Medicine. J. Basic. Clin. Physiol. Pharmacol. 2020, 31, 20190115. [Google Scholar] [CrossRef]
  367. Wang, Z.; Chen, Q.; Li, B.; Xie, J.; Yang, X.; Zhao, K.; Wu, Y.; Ye, Z.; Chen, Z.; Qin, Z.; et al. Escin-Induced DNA Damage Promotes Escin-Induced Apoptosis in Human Colorectal Cancer Cells via P62 Regulation of the ATM/ΓH2AX Pathway. Acta Pharmacol. Sin. 2018, 39, 1645–1660. [Google Scholar] [CrossRef] [PubMed]
  368. Harikumar, K.B.; Sung, B.; Pandey, M.K.; Guha, S.; Krishnan, S.; Aggarwal, B.B. Escin, a Pentacyclic Triterpene, Chemosensitizes Human Tumor Cells through Inhibition of Nuclear Factor-ΚB Signaling Pathway. Mol. Pharmacol. 2010, 77, 818–827. [Google Scholar] [CrossRef]
  369. Jiang, N.; Xin, W.; Wang, T.; Zhang, L.; Fan, H.; Du, Y.; Li, C.; Fu, F. Protective Effect of Aescin from the Seeds of Aesculus hippocastanum on Liver Injury Induced by Endotoxin in Mice. Phytomedicine 2011, 18, 1276–1284. [Google Scholar] [CrossRef] [PubMed]
  370. Wang, Y.W.; Wang, S.J.; Zhou, Y.N.; Pan, S.H.; Sun, B. Escin Augments the Efficacy of Gemcitabine through Down-Regulation of Nuclear Factor-ΚB and Nuclear Factor-ΚB-Regulated Gene Products in Pancreatic Cancer Both In Vitro and In Vivo. J. Cancer Res. Clin. Oncol. 2012, 138, 785–797. [Google Scholar] [CrossRef] [PubMed]
  371. Borisenko, O.V.; Belen’kiǐ, D.A. Impact of Combined Therapy Using Glucosamine Sulfate and Anti-Inflammatory Agent on Pain Severity in Patients with Osteoarthritis: Prospective, Non-Controlled Postmarketing Study. Klin. Med. 2013, 91, 65–71. [Google Scholar]
  372. Zeng, X.; Wang, B.; Li, L.; Lei, T.; Liu, H.; Sun, Y. Therapeutic Effect Analysis of Sodium Aescinate Tablets on Knee Osteoarthritis Combined with Synovitis. J. Clin. Nurs. Res. 2021, 5, 1–6. [Google Scholar] [CrossRef]
  373. Maghsoudi, H.; Hallajzadeh, J.; Rezaeipour, M. Evaluation of the Effect of Polyphenol of Escin Compared with Ibuprofen and Dexamethasone in Synoviocyte Model for Osteoarthritis: An In Vitro Study. Clin. Rheumatol. 2018, 37, 2471–2478. [Google Scholar] [CrossRef]
  374. Fu, F.; Hou, Y.; Jiang, W.; Wang, R.; Liu, K. Escin: Inhibiting Inflammation and Promoting Gastrointestinal Transit to Attenuate Formation of Postoperative Adhesions. World J. Surg. 2005, 29, 1614–1620. [Google Scholar] [CrossRef] [PubMed]
  375. Matsuda, H.; Li, Y.; Yoshikawa, M. Possible Involvement of 5-HT and 5-HT2 Receptors in Acceleration of Gastrointestinal Transit by Escin Ib in Mice. Life Sci. 2000, 66, 2233–2238. [Google Scholar] [CrossRef] [PubMed]
  376. Singh, O.; Khanam, Z.; Misra, N.; Srivastava, M. Chamomile (Matricaria chamomilla L.): An Overview. Pharmacogn. Rev. 2011, 5, 82. [Google Scholar] [CrossRef]
  377. Ortiz, M.I.; Fernández-Martínez, E.; Soria-Jasso, L.E.; Lucas-Gómez, I.; Villagómez-Ibarra, R.; González-García, M.P.; Castañeda-Hernández, G.; Salinas-Caballero, M. Isolation, Identification and Molecular Docking as Cyclooxygenase (COX) Inhibitors of the Main Constituents of Matricaria chamomilla L. Extract and Its Synergistic Interaction with Diclofenac on Nociception and Gastric Damage in Rats. Biomed. Pharmacother. 2016, 78, 248–256. [Google Scholar] [CrossRef] [PubMed]
  378. Gosztola, B.; Sárosi, S.; Németh, E. Variability of the Essential Oil Content and Composition of Chamomile (Matricaria recutita L.) Affected by Weather Conditions. Nat. Prod. Commun. 2010, 5, 465–470. [Google Scholar] [CrossRef] [PubMed]
  379. Orav, A.; Raal, A.; Arak, E. Content and Composition of the Essential Oil of Chamomilla recutita (L.) Rauschert from Some European Countries. Nat. Prod. Res. 2010, 24, 48–55. [Google Scholar] [CrossRef] [PubMed]
  380. Akram, W.; Ahmed, S.; Rihan, M.; Arora, S.; Khalid, M.; Ahmad, S.; Ahmad, F.; Haque, S.; Vashishth, R. An Updated Comprehensive Review of the Therapeutic Properties of Chamomile (Matricaria chamomilla L.). Int. J. Food Prop. 2024, 27, 133–164. [Google Scholar] [CrossRef]
  381. Sah, A.; Naseef, P.P.; Kuruniyan, M.S.; Jain, G.K.; Zakir, F.; Aggarwal, G. A Comprehensive Study of Therapeutic Applications of Chamomile. Pharmaceuticals 2022, 15, 1284. [Google Scholar] [CrossRef] [PubMed]
  382. Catani, M.V.; Rinaldi, F.; Tullio, V.; Gasperi, V.; Savini, I. Comparative Analysis of Phenolic Composition of Six Commercially Available Chamomile (Matricaria chamomilla L.) Extracts: Potential Biological Implications. Int. J. Mol. Sci. 2021, 22, 10601. [Google Scholar] [CrossRef]
  383. Avonto, C.; Wang, M.; Chittiboyina, A.G.; Avula, B.; Zhao, J.; Khan, I.A. Hydroxylated Bisabolol Oxides: Evidence for Secondary Oxidative Metabolism in Matricaria chamomilla. J. Nat. Prod. 2013, 76, 1848–1853. [Google Scholar] [CrossRef]
  384. Asgharzade, S.; Rabiei, Z.; Rafieian-Kopaei, M. Effects of Matricaria chamomilla Extract on Motor Coordination Impairment Induced by Scopolamine in Rats. Asian Pac. J. Trop. Biomed. 2015, 5, 829–833. [Google Scholar] [CrossRef]
  385. Rafraf, M.; Zemestani, M.; Asghari-Jafarabadi, M. Effectiveness of Chamomile Tea on Glycemic Control and Serum Lipid Profile in Patients with Type 2 Diabetes. J. Endocrinol. Invest. 2015, 38, 163–170. [Google Scholar] [CrossRef]
  386. Bayliak, M.M.; Dmytriv, T.R.; Melnychuk, A.V.; Strilets, N.V.; Storey, K.B.; Lushchak, V.I. Chamomile as a Potential Remedy for Obesity and Metabolic Syndrome. EXCLI J. 2021, 20, 1261–1286. [Google Scholar] [CrossRef] [PubMed]
  387. Awaad, A.A.; El-Meligy, R.M.; Zain, G.M.; Safhi, A.A.; AL Qurain, N.A.; Almoqren, S.S.; Zain, Y.M.; Sesh Adri, V.D.; Al-Saikhan, F.I. Experimental and Clinical Antihypertensive Activity of Matricaria chamomilla Extracts and Their Angiotensin-converting Enzyme Inhibitory Activity. Phytother. Res. 2018, 32, 1564–1573. [Google Scholar] [CrossRef]
  388. Chandrashekhar, V.M.; Halagali, K.S.; Nidavani, R.B.; Shalavadi, M.H.; Biradar, B.S.; Biswas, D.; Muchchandi, I.S. Anti-Allergic Activity of German Chamomile (Matricaria recutita L.) in Mast Cell Mediated Allergy Model. J. Ethnopharmacol. 2011, 137, 336–340. [Google Scholar] [CrossRef] [PubMed]
  389. Saidi, R.; Heidari, H.; Sedehi, M.; Safdarian, B. Evaluating the Effect of Matricaria chamomilla and Melissa Officinalis on Pain Intensity and Satisfaction with Pain Management in Patients after Orthopedic Surgery. J. Herbmed Pharmacol. 2020, 9, 339–345. [Google Scholar] [CrossRef]
  390. Jabri, M.-A.; Aissani, N.; Tounsi, H.; Sakly, M.; Marzouki, L.; Sebai, H. Protective Effect of Chamomile (Matricaria recutita L.) Decoction Extract against Alcohol-Induced Injury in Rat Gastric Mucosa. Pathophysiology 2017, 24, 1–8. [Google Scholar] [CrossRef]
  391. Afrigan, L.; Jafari Anarkooli, I.; Sohrabi, D.; Abdanipour, A.; Yazdinezhad, A.; Sayyar, Z.; Ghorbanlou, M.; Arianmanesh, M. The Effect of Hydroethanolic Extract of Matricaria chamomilla on the Reproductive System of Male Rats Exposed to Formaldehyde. Andrologia 2019, 51, e13362. [Google Scholar] [CrossRef]
  392. Zargaran, A.; Borhani-Haghighi, A.; Faridi, P.; Daneshamouz, S.; Kordafshari, G.; Mohagheghzadeh, A. Potential Effect and Mechanism of Action of Topical Chamomile (Matricaria chammomila L.) Oil on Migraine Headache: A Medical Hypothesis. Med. Hypotheses 2014, 83, 566–569. [Google Scholar] [CrossRef]
  393. Srivastava, J.K.; Pandey, M.; Gupta, S. Chamomile, a Novel and Selective COX-2 Inhibitor with Anti-Inflammatory Activity. Life Sci. 2009, 85, 663–669. [Google Scholar] [CrossRef]
  394. Satyal, P.; Shrestha, S.; Setzer, W.N. Composition and Bioactivities of an (E)-β-Farnesene Chemotype of Chamomile (Matricaria chamomilla) Essential Oil from Nepal. Nat. Prod. Commun. 2015, 10, 1934578X1501000. [Google Scholar] [CrossRef]
  395. Sharifi, H.; Minaie, M.B.; Qasemzadeh, M.J.; Ataei, N.; Gharehbeglou, M.; Heydari, M. Topical Use of Matricaria recutita L (Chamomile) Oil in the Treatment of Monosymptomatic Enuresis in Children. J. Evid. Based Complement. Altern. Med. 2017, 22, 12–17. [Google Scholar] [CrossRef] [PubMed]
  396. Pelissolo, A. Efficacité et Tolérance de l’escitalopram Dans Les Troubles Anxieux : Revue de La Littérature. Encephale 2008, 34, 400–408. [Google Scholar] [CrossRef] [PubMed]
  397. Amsterdam, J.D.; Li, Y.; Soeller, I.; Rockwell, K.; Mao, J.J.; Shults, J. A Randomized, Double-Blind, Placebo-Controlled Trial of Oral Matricaria recutita (Chamomile) Extract Therapy for Generalized Anxiety Disorder. J. Clin. Psychopharmacol. 2009, 29, 378–382. [Google Scholar] [CrossRef] [PubMed]
  398. Miguel, F.G.; Cavalheiro, A.H.; Spinola, N.F.; Ribeiro, D.L.; Barcelos, G.R.M.; Antunes, L.M.G.; Hori, J.I.; Marquele-Oliveira, F.; Rocha, B.A.; Berretta, A.A. Validation of a RP-HPLC-DAD Method for Chamomile (Matricaria recutita) Preparations and Assessment of the Marker, Apigenin-7-Glucoside, Safety and Anti-Inflammatory Effect. Evid. -Based Complement. Altern. Med. 2015, 2015, 828437. [Google Scholar] [CrossRef] [PubMed]
  399. Mamalis, A.; Nguyen, D.-H.; Brody, N.; Jagdeo, J. The Active Natural Anti-Oxidant Properties of Chamomile, Milk Thistle, and Halophilic Bacterial Components in Human Skin In Vitro. J. Drugs Dermatol. 2013, 12, 780–784. [Google Scholar] [PubMed]
  400. Drummond, E.M.; Harbourne, N.; Marete, E.; Jacquier, J.C.; O’Riordan, D.; Gibney, E.R. An In Vivo Study Examining the Antiinflammatory Effects of Chamomile, Meadowsweet, and Willow Bark in a Novel Functional Beverage. J. Diet. Suppl. 2013, 10, 370–380. [Google Scholar] [CrossRef] [PubMed]
  401. Shoara, R.; Hashempur, M.H.; Ashraf, A.; Salehi, A.; Dehshahri, S.; Habibagahi, Z. Efficacy and Safety of Topical Matricaria chamomilla L. (Chamomile) Oil for Knee Osteoarthritis: A Randomized Controlled Clinical Trial. Complement. Ther. Clin. Pract. 2015, 21, 181–187. [Google Scholar] [CrossRef]
  402. Mushtaq, Z.; Sadeer, N.B.; Hussain, M.; Mahwish; Alsagaby, S.A.; Imran, M.; Mumtaz, T.; Umar, M.; Tauseef, A.; Al Abdulmonem, W.; et al. Therapeutical Properties of Apigenin: A Review on the Experimental Evidence and Basic Mechanisms. Int. J. Food Prop. 2023, 26, 1914–1939. [Google Scholar] [CrossRef]
  403. Lavanya, J.; Periyar Selvam, S.; Jeevitha Priya, M.; Preethi, J.; Aradana, M. Antioxidant and Antimicrobial Activity of Selected Medicinal Plants against Human Oral Pathogens. Int. J. Pharm. Pharm. Sci. 2016, 8, 71. [Google Scholar] [CrossRef]
  404. Nikseresht, M.; Kamali, A.; Rahimi, H.; Delaviz, H.; Toori, M.; Kashani, I.; Mahmoudi, R. The Hydroalcoholic Extract of Matricaria chamomilla Suppresses Migration and Invasion of Human Breast Cancer MDA-MB-468 and MCF-7 Cell Lines. Pharmacogn. Res. 2017, 9, 87. [Google Scholar] [CrossRef] [PubMed]
  405. Asadi, Z.; Ghazanfari, T.; Hatami, H. Anti-Inflammatory Effects of Matricaria chamomilla Extracts on BALB/c Mice Macrophages and Lymphocytes. Iran. J. Allergy Asthma Immunol. 2020. [Google Scholar] [CrossRef] [PubMed]
  406. Lee, Y.M.; Son, E.; Kim, S.-H.; Kim, D.-S. Protective Effects of Glycine Soja Leaf and Stem Extract against Chondrocyte Inflammation and Osteoarthritis. Int. J. Mol. Sci. 2023, 24, 4829. [Google Scholar] [CrossRef] [PubMed]
  407. Wang, K.-J.; Li, X.-H.; Zhang, J.-J.; Chen, H.; Zhang, Z.-L.; Yu, G.-D. Natural Introgression from Cultivated Soybean (Glycine Max) into Wild Soybean (Glycine Soja) with the Implications for Origin of Populations of Semi-Wild Type and for Biosafety of Wild Species in China. Genet. Resour. Crop Evol. 2010, 57, 747–761. [Google Scholar] [CrossRef]
  408. Wen, Z.; Ding, Y.; Zhao, T.; Gai, J. Genetic Diversity and Peculiarity of Annual Wild Soybean (G. Soja Sieb. et Zucc.) from Various Eco-Regions in China. Theor. Appl. Genet. 2009, 119, 371–381. [Google Scholar] [CrossRef] [PubMed]
  409. Kuroda, Y.; Kaga, A.; Tomooka, N.; Yano, H.; Takada, Y.; Kato, S.; Vaughan, D. QTL Affecting Fitness of Hybrids between Wild and Cultivated Soybeans in Experimental Fields. Ecol. Evol. 2013, 3, 2150–2168. [Google Scholar] [CrossRef] [PubMed]
  410. Chen, Q.; Wang, X.; Yuan, X.; Shi, J.; Zhang, C.; Yan, N.; Jing, C. Comparison of Phenolic and Flavonoid Compound Profiles and Antioxidant and α-Glucosidase Inhibition Properties of Cultivated Soybean (Glycine Max) and Wild Soybean (Glycine Soja). Plants 2021, 10, 813. [Google Scholar] [CrossRef]
  411. Jing, C.; Wen, Z.; Zou, P.; Yuan, Y.; Jing, W.; Li, Y.; Zhang, C. Consumption of Black Legumes Glycine Soja and Glycine Max Lowers Serum Lipids and Alters the Gut Microbiome Profile in Mice Fed a High-Fat Diet. J. Agric. Food Chem. 2018, 66, 7367–7375. [Google Scholar] [CrossRef]
  412. Loo, F.A.J.V.D.; Joosten, L.A.B.; Van Lent, P.L.E.M.; Arntz, O.J.; Van Den Berg, W.B. Role of Interleukin-1, Tumor Necrosis Factor α, and Interleukin-6 in Cartilage Proteoglycan Metabolism and Destruction Effect of in Situ Blocking in Murine Antigen- and Zymosan-induced Arthritis. Arthritis Rheum. 1995, 38, 164–172. [Google Scholar] [CrossRef]
  413. Dinarello, C.A. Immunological and Inflammatory Functions of the Interleukin-1 Family. Annu. Rev. Immunol. 2009, 27, 519–550. [Google Scholar] [CrossRef]
  414. Marcu, K.B.; Otero, M.; Olivotto, E.; Maria Borzi, R.; Goldring, M.B. NF-KappaB Signaling: Multiple Angles to Target OA. Curr. Drug Targets 2010, 11, 599–613. [Google Scholar] [CrossRef] [PubMed]
  415. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Clinical Aspects and Health Benefits of Ginger (Zingiber officinale) in Both Traditional Chinese Medicine and Modern Industry. Acta Agric. Scand. B Soil. Plant Sci. 2019, 69, 546–556. [Google Scholar] [CrossRef]
  416. Govindarajan, V.S.; Connell, D.W. Ginger—Chemistry, Technology, and Quality Evaluation: Part 1. C R C Crit. Rev. Food Sci. Nutr. 1983, 17, 1–96. [Google Scholar] [CrossRef] [PubMed]
  417. Butt, M.S.; Sultan, M.T. Ginger and Its Health Claims: Molecular Aspects. Crit. Rev. Food Sci. Nutr. 2011, 51, 383–393. [Google Scholar] [CrossRef] [PubMed]
  418. Young, H.-Y.; Luo, Y.-L.; Cheng, H.-Y.; Hsieh, W.-C.; Liao, J.-C.; Peng, W.-H. Analgesic and Anti-Inflammatory Activities of x-Gingerol. J. Ethnopharmacol. 2005, 96, 207–210. [Google Scholar] [CrossRef]
  419. Tripathi, S.; Maier, K.; Bruch, D.; Kittur, D. Ginger and Its Active Ingredient 6-Gingerol down Regulate pro-Inflammatory Cytokine Release by Macrophages. J. Surg. Res. 2006, 130, 318. [Google Scholar] [CrossRef]
  420. Al-Shibani, N.; Al-Kattan, R.; Alssum, L.; Allam, E. Effects of Ginger (Zingiber officinale) on Gingival Fibroblasts: An In Vitro Study. Clin. Exp. Dent. Res. 2022, 8, 906–911. [Google Scholar] [CrossRef] [PubMed]
  421. Thomson, M.; Al-Qattan, K.K.; Al-Sawan, S.M.; Alnaqeeb, M.A.; Khan, I.; Ali, M. The Use of Ginger (Zingiber officinale Rosc.) as a Potential Anti-Inflammatory and Antithrombotic Agent. Prostaglandins Leukot. Essent. Fat. Acids 2002, 67, 475–478. [Google Scholar] [CrossRef] [PubMed]
  422. Shen, C.-L.; Hong, K.-J.; Kim, S.W. Effects of Ginger (Zingiber officinale Rosc.) on Decreasing the Production of Inflammatory Mediators in Sow Osteoarthrotic Cartilage Explants. J. Med. Food 2003, 6, 323–328. [Google Scholar] [CrossRef]
  423. Ballester, P.; Cerdá, B.; Arcusa, R.; Marhuenda, J.; Yamedjeu, K.; Zafrilla, P. Effect of Ginger on Inflammatory Diseases. Molecules 2022, 27, 7223. [Google Scholar] [CrossRef]
  424. Kim, S.O.; Kundu, J.K.; Shin, Y.K.; Park, J.-H.; Cho, M.-H.; Kim, T.-Y.; Surh, Y.-J. x-Gingerol Inhibits COX-2 Expression by Blocking the Activation of P38 MAP Kinase and NF-ΚB in Phorbol Ester-Stimulated Mouse Skin. Oncogene 2005, 24, 2558–2567. [Google Scholar] [CrossRef]
  425. Al-Khayri, J.M.; Sahana, G.R.; Nagella, P.; Joseph, B.V.; Alessa, F.M.; Al-Mssallem, M.Q. Flavonoids as Potential Anti-Inflammatory Molecules: A Review. Molecules 2022, 27, 2901. [Google Scholar] [CrossRef] [PubMed]
  426. Mashhadi, N.S.; Ghiasvand, R.; Askari, G.; Hariri, M.; Darvishi, L.; Mofid, M.R. Anti-Oxidative and Anti-Inflammatory Effects of Ginger in Health and Physical Activity: Review of Current Evidence. Int. J. Prev. Med. 2013, 4, S36–S42. [Google Scholar] [PubMed]
  427. Ippoushi, K.; Azuma, K.; Ito, H.; Horie, H.; Higashio, H. x-Gingerol Inhibits Nitric Oxide Synthesis in Activated J774.1 Mouse Macrophages and Prevents Peroxynitrite-Induced Oxidation and Nitration Reactions. Life Sci. 2003, 73, 3427–3437. [Google Scholar] [CrossRef] [PubMed]
  428. Pan, M.; Hsieh, M.; Hsu, P.; Ho, S.; Lai, C.; Wu, H.; Sang, S.; Ho, C. 6-Shogaol Suppressed Lipopolysaccharide-induced Up-expression of INOS and COX-2 in Murine Macrophages. Mol. Nutr. Food Res. 2008, 52, 1467–1477. [Google Scholar] [CrossRef] [PubMed]
  429. Naderi, Z.; Mozaffari-Khosravi, H.; Dehghan, A.; Nadjarzadeh, A.; Huseini, H.F. Effect of Ginger Powder Supplementation on Nitric Oxide and C-Reactive Protein in Elderly Knee Osteoarthritis Patients: A 12-Week Double-Blind Randomized Placebo-Controlled Clinical Trial. J. Tradit. Complement. Med. 2016, 6, 199–203. [Google Scholar] [CrossRef] [PubMed]
  430. Piovezana Bossolani, G.D.; Silva, B.T.; Colombo Martins Perles, J.V.; Lima, M.M.; Vieira Frez, F.C.; Garcia de Souza, S.R.; Sehaber-Sierakowski, C.C.; Bersani-Amado, C.A.; Zanoni, J.N. Rheumatoid Arthritis Induces Enteric Neurodegeneration and Jejunal Inflammation, and Quercetin Promotes Neuroprotective and Anti-Inflammatory Actions. Life Sci. 2019, 238, 116956. [Google Scholar] [CrossRef] [PubMed]
  431. Bhaskar, S.; Sudhakaran, P.R.; Helen, A. Quercetin Attenuates Atherosclerotic Inflammation and Adhesion Molecule Expression by Modulating TLR-NF-ΚB Signaling Pathway. Cell Immunol. 2016, 310, 131–140. [Google Scholar] [CrossRef]
  432. Hu, Y.; Gui, Z.; Zhou, Y.; Xia, L.; Lin, K.; Xu, Y. Quercetin Alleviates Rat Osteoarthritis by Inhibiting Inflammation and Apoptosis of Chondrocytes, Modulating Synovial Macrophages Polarization to M2 Macrophages. Free Radic. Biol. Med. 2019, 145, 146–160. [Google Scholar] [CrossRef]
  433. 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. 2020, 48, 030006051987346. [Google Scholar] [CrossRef]
  434. Kirkland, J.L.; Tchkonia, T. Senolytic Drugs: From Discovery to Translation. J. Intern. Med. 2020, 288, 518–536. [Google Scholar] [CrossRef] [PubMed]
  435. Feng, K.; Chen, Z.; Pengcheng, L.; Zhang, S.; Wang, X. Quercetin Attenuates Oxidative Stress-induced Apoptosis via SIRT1/AMPK-mediated Inhibition of ER Stress in Rat Chondrocytes and Prevents the Progression of Osteoarthritis in a Rat Model. J. Cell Physiol. 2019, 234, 18192–18205. [Google Scholar] [CrossRef] [PubMed]
  436. Li, W.; Wang, Y.; Tang, Y.; Lu, H.; Qi, Y.; Li, G.; He, H.; Lu, F.; Yang, Y.; Sun, H. Quercetin Alleviates Osteoarthritis Progression in Rats by Suppressing Inflammation and Apoptosis via Inhibition of IRAK1/NLRP3 Signaling. J. Inflamm. Res. 2021, 14, 3393–3403. [Google Scholar] [CrossRef] [PubMed]
  437. Wang, X.-P.; Xie, W.-P.; Bi, Y.-F.; Wang, B.-A.; Song, H.-B.; Wang, S.-L.; Bi, R.-X. Quercetin Suppresses Apoptosis of Chondrocytes Induced by IL-1β via Inactivation of P38 MAPK Signaling Pathway. Exp. Ther. Med. 2021, 21, 468. [Google Scholar] [CrossRef] [PubMed]
  438. Heydari Nasrabadi, M.; Parsivand, M.; Mohammadi, N.; Asghari Moghaddam, N. Comparison of Elaeagnus angustifolia L. Extract and Quercetin on Mouse Model of Knee Osteoarthritis. J. Ayurveda Integr. Med. 2022, 13, 100529. [Google Scholar] [CrossRef] [PubMed]
  439. World Health Organization WHO Traditional Medicine Strategy World Health Organization. Available online: http://www.who.int/medicines/publications/traditional/trm_strategy14_23/en/ (accessed on 7 February 2024).
  440. van Breemen, R.B.; Fong, H.H.S.; Farnsworth, N.R. The Role of Quality Assurance and Standardization in the Safety of Botanical Dietary Supplements. Chem. Res. Toxicol. 2007, 20, 577–582. [Google Scholar] [CrossRef] [PubMed]
  441. Thakkar, S.; Anklam, E.; Xu, A.; Ulberth, F.; Li, J.; Li, B.; Hugas, M.; Sarma, N.; Crerar, S.; Swift, S.; et al. Regulatory Landscape of Dietary Supplements and Herbal Medicines from a Global Perspective. Regul. Toxicol. Pharmacol. 2020, 114, 104647. [Google Scholar] [CrossRef] [PubMed]
  442. Bagchi, D. Nutraceuticals and Functional Foods Regulations in the United States and around the World. Toxicology 2006, 221, 1–3. [Google Scholar] [CrossRef] [PubMed]
  443. World Health Organization. WHO Guidelines for Assessing Quality of Herbal Medicines with Reference to Contaminants and Residues; World Health Organization, Ed.; WHO Press: Geneva, Switzerland, 2007; ISBN 9789241594448. [Google Scholar]
  444. Bailey, R.L. Current Regulatory Guidelines and Resources to Support Research of Dietary Supplements in the United States. Crit. Rev. Food Sci. Nutr. 2020, 60, 298–309. [Google Scholar] [CrossRef]
  445. Helal, N.A.; Eassa, H.A.; Amer, A.M.; Eltokhy, M.A.; Edafiogho, I.; Nounou, M.I. Nutraceuticals’ Novel Formulations: The Good, the Bad, the Unknown and Patents Involved. Recent. Pat. Drug Deliv. Formul. 2019, 13, 105–156. [Google Scholar] [CrossRef]
  446. Ansari, M.Y.; Ahmad, N.; Haqqi, T.M. Oxidative Stress and Inflammation in Osteoarthritis Pathogenesis: Role of Polyphenols. Biomed. Pharmacother. 2020, 129, 110452. [Google Scholar] [CrossRef] [PubMed]
  447. Colletti, A.; Cicero, A.F.G. Nutraceutical Approach to Chronic Osteoarthritis: From Molecular Research to Clinical Evidence. Int. J. Mol. Sci. 2021, 22, 12920. [Google Scholar] [CrossRef] [PubMed]
  448. 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]
  449. Verma, S.; Pandey, A.K. Improving Bioavailability of Nutrients Through Nanotechnology. In Sustainable Agriculture Reviews 55; Springer: Cham, Switzerland, 2021; pp. 135–170. [Google Scholar]
  450. Huang, H.; Lou, Z.; Zheng, S.; Wu, J.; Yao, Q.; Chen, R.; Kou, L.; Chen, D. Intra-Articular Drug Delivery Systems for Osteoarthritis Therapy: Shifting from Sustained Release to Enhancing Penetration into Cartilage. Drug Deliv. 2022, 29, 767–791. [Google Scholar] [CrossRef]
  451. Patil, P.; Nene, S.; Shah, S.; Singh, S.B.; Srivastava, S. Exploration of Novel Drug Delivery Systems in Topical Management of Osteoarthritis. Drug Deliv. Transl. Res. 2023, 13, 531–546. [Google Scholar] [CrossRef]
  452. Han, R.; Wu, Y.; Han, Y.; Liu, X.; Liu, H.; Su, J. Engineered Plant Extracellular Vesicles for Autoimmune Diseases Therapy. Nano Res. 2024, 17, 2857–2873. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the pathological mechanisms in osteoarthritis (OA) highlighting the complex interplay among bone, cartilage, and synovial tissues [126]. The diagram illustrates how prolonged mechanical stress, wear, and trauma lead to the secretion of extracellular matrix (ECM) by cartilage cells. Key enzymes involved in ECM remodeling—such as MMPs, ADAMTSs, plasmin, plasminogen activator, and TIMPs—are shown to have disrupted activity, contributing to excessive cartilage catabolism. The role of interleukins, TGF-β, and BMP-2 in cartilage morphology, tissue homeostasis, and metabolism is depicted alongside the suppression of TIMPs. The feedback loop of MMP activation, TNF-α, and interleukin synthesis by synovial tissues and chondrocytes fosters a sustained inflammatory cascade. This leads to the release of NO and prostaglandin E2, proliferation of chondrocytes, and deposition of collagen type X, which undergoes calcification, exacerbating OA pathology. BMP-2: bone morphogenetic protein 2; PA: plasminogen activator; MMP: matrix metalloproteinase; TGF-β: transforming growth factor beta; TIMP: tissue inhibitor of metalloproteinase; TNF-α: tumor necrosis factor alpha; NO: nitric oxide; PGE2: Prostaglandin E2; IL-1: Interleukin-1; IL-8: Interleukin-8.
Figure 1. Schematic representation of the pathological mechanisms in osteoarthritis (OA) highlighting the complex interplay among bone, cartilage, and synovial tissues [126]. The diagram illustrates how prolonged mechanical stress, wear, and trauma lead to the secretion of extracellular matrix (ECM) by cartilage cells. Key enzymes involved in ECM remodeling—such as MMPs, ADAMTSs, plasmin, plasminogen activator, and TIMPs—are shown to have disrupted activity, contributing to excessive cartilage catabolism. The role of interleukins, TGF-β, and BMP-2 in cartilage morphology, tissue homeostasis, and metabolism is depicted alongside the suppression of TIMPs. The feedback loop of MMP activation, TNF-α, and interleukin synthesis by synovial tissues and chondrocytes fosters a sustained inflammatory cascade. This leads to the release of NO and prostaglandin E2, proliferation of chondrocytes, and deposition of collagen type X, which undergoes calcification, exacerbating OA pathology. BMP-2: bone morphogenetic protein 2; PA: plasminogen activator; MMP: matrix metalloproteinase; TGF-β: transforming growth factor beta; TIMP: tissue inhibitor of metalloproteinase; TNF-α: tumor necrosis factor alpha; NO: nitric oxide; PGE2: Prostaglandin E2; IL-1: Interleukin-1; IL-8: Interleukin-8.
Cimb 46 00251 g001
Table 1. Kellgren and Lawrence classification of radiological OA. The pathology is present starting from grade 2, even if of minimal severity [71].
Table 1. Kellgren and Lawrence classification of radiological OA. The pathology is present starting from grade 2, even if of minimal severity [71].
GradeKL Scale
0
None
No pathophysiologic involvement of osteoarthritis
1
Doubtful
Normal joint with only one tiny osteophyte
2
Minimal
Clear osteophytes at two spots with slight hardening of the bone under the cartilage and possible hollow areas, but normal joint gap and no distortion
3
Moderate
Moderate osteophytes presence, some bone malformation and shrinking of joint gap
4
Severe
Large presence of osteophytes and bone end impairment, loss of joint space, densification, and cysts
Table 2. Clinical trials on natural molecules in OA (information extracted from clinicaltrials.gov, accessed on 31 March 2024).
Table 2. Clinical trials on natural molecules in OA (information extracted from clinicaltrials.gov, accessed on 31 March 2024).
Molecule ClassMechanisms of ActionClinical StudiesClinicalTrials.gov IdentifierStatusOutcomeSponsor/
Collaborators
Curcumin
DiarylheptanoidAnti-inflammatory effect [243]The Efficacy and Safety of Curcuma Domestica Extracts and Ibuprofen for Therapy of Patients with Knee OsteoarthritisNCT00792818Phase 3—Completed
N = 367
Pain reduction and functional improvement comparable to ibuprofen but with lesser gastrointestinal side effects [297]Mahidol University, Salaya, Thailand
Evaluation of FLEXOFYTOL® Versus PLACEBO (COPRA)NCT02909621Phase 2—Completed
N = 150
Pain reduction when used as adjuvant to paracetamol and/or NSAIDs in comparison to placebo; suitable safety profile and lower Patient Global Assessment of Disease Activity reported in comparison to placebo [298]Tilman S.A., Baillonville, Belgium
Comparative Study of Turmeric Extract in Patients with ArthrosisNCT04500210Phase 3—Completed
N = 120
-Kaj Winther Hansen
Evaluation of the Efficacy of a Turmeric Extract (Arantal®) in Patients with Osteoarthritis of the Knee (Gonarthrosis)NCT00992004Phase 2—Completed
N = 280
-Bioxtract S.A., Gembloux, Belgium
Exploratory Non-Comparative Study to Evaluate the Efficacy of Highly Bioavailable Curcumin (Flexofytol) in Patients with Knee OsteoarthritisNCT01909037Early Phase 1—Completed
N = 22
Chondrogenic effect and inhibition of proinflammatory cytokines, prostanoids, and MMPs released by chondrocytes. Inhibition of TNF-α activity and production both in vitro and in vivo [299]Tilman S.A., Baillonville, Belgium
Effectiveness of Curcumin-based Food Supplement in Reducing Pain and Inflammatory Component in Osteoarthritis (FENOXI-1900)NCT04207021Not Applicable
N = 134
-KOS Care SRL—Istituto di Riabilitazione Santo Stefano
Combination of Curcuminoid with Acupressure for Inflammation and Pain in the Elderly with Osteoarthritis GenuNCT06105840Phase 2—Enrolling by invitation
N = 70
-Gadjah Mada University, Sleman, Indonesia
Randomized Trial of Regenexx Stem Cell Support FormulaNCT04661267Not Applicable
N = 80
-Regenexx LLC, Des Moines, IA, USA
Epigenorm Antivir Combined with Acupuncture for the Treatment of Osteoarthritis Patients Who Are Overweight or ObeseNCT03540186Not Applicable
N = 15
-Epigenorm Antivir Combined with Acupuncture for the Treatment of Osteoarthritis Patients Who Are Overweight or Obese
Bromelain
Proteinase-peptidaseAnti-inflammatory, analgesic, anti-edematous, and fibrinolytic effects [244]Study to Investigate the Mechanism of Action of an Oral Enzyme Treatment with Bromelain, Trypsin and Rutoside Versus Placebo in Subjects with OsTeoarthritis (WobeSmart)NCT05038410Not Applicable
N = 40
-Mucos Pharma GmbH & Co. KG, Berlin, Germany
Harpagophytum procumbens (devil’s claw)
Mix of phenolic acids and glycosides, triterpenes, phytosterols, iridoid glucosides like harpagoside and various flavonoidsAnti-rheumatic, anti-inflammatory, and analgesic effects [245]Trial Evaluating Devil’s Claw for the Treatment of Hip and Knee OsteoarthritisNCT00295490Phase 2—Completed
N = 67
-University of Southampton, UK
Clinical Efficacy and Safety of Loxacon Dietary Supplement Capsules at Patients with Knee ArthrosisNCT05925725Phase 4—Completed
N = 100
-Polyclinic of the Hospitaller Brothers of St. John of God, Budapest, Hungary
Boswellia serrata
TerpeneAnti-inflammatory effect [246]Efficacy of Myalgesin™ to Support Joint Function in Patients with Knee OsteoarthritisNCT00577330Phase 3—Not applicable
N = 110
-ProThera, Inc., Reno, NV, USA
A Study of the Feasibility of Using the Dietary Supplement “ARTNEO” in Patients with OsteoathritisNCT05975879Not Applicable
N = 212
-NPO Petrovax, Moscow, Russia
Clinical Efficacy and Safety of Loxacon Dietary Supplement Capsules at Patients with Knee ArthrosisNCT05925725Phase 4—Completed
N = 100
-Polyclinic of the Hospitaller Brothers of St. John of God, Budapest, Hungary
A Study to Assess Efficacy of Supporting Properties and Safety of ARTNEO in Patients with Knee OsteoarthritisNCT06032442Not Applicable
N = 70
-NPO Petrovax, Moscow, Russia
Management of Joint Pain Associated with Osteoarthritis of the Knee with Association of Plant ExtractsNCT02977936Not Applicable
N = 126
-PiLeJe, Paris, France
To Assess the Lanconone® (E-OA-07) Efficacy in Physical Activity-related Pain-LEAP Study (LEAP)NCT03262805Not Applicable
N = 73
-Vedic Lifesciences Pvt. Ltd., Mumbai, India
Effects of Glucosamine and Chondroitin Supplementation in Women with Knee Osteoarthritis Participating in an Exercise and Weight Loss ProgramNCT01271218Phase 4—Completed
N = 36
-Texas A&M University, College Station, TX, USA
Quercetin
FlavonoidAntioxidant [300]Effect of Natural Senolytic Agents & NLRP3 Inhibitors on OsteoarthritisNCT05276895Not Applicable
N = 60
OngoingAssiut University, Assiut, Egypt
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

Coppola, C.; Greco, M.; Munir, A.; Musarò, D.; Quarta, S.; Massaro, M.; Lionetto, M.G.; Maffia, M. Osteoarthritis: Insights into Diagnosis, Pathophysiology, Therapeutic Avenues, and the Potential of Natural Extracts. Curr. Issues Mol. Biol. 2024, 46, 4063-4105. https://doi.org/10.3390/cimb46050251

AMA Style

Coppola C, Greco M, Munir A, Musarò D, Quarta S, Massaro M, Lionetto MG, Maffia M. Osteoarthritis: Insights into Diagnosis, Pathophysiology, Therapeutic Avenues, and the Potential of Natural Extracts. Current Issues in Molecular Biology. 2024; 46(5):4063-4105. https://doi.org/10.3390/cimb46050251

Chicago/Turabian Style

Coppola, Chiara, Marco Greco, Anas Munir, Debora Musarò, Stefano Quarta, Marika Massaro, Maria Giulia Lionetto, and Michele Maffia. 2024. "Osteoarthritis: Insights into Diagnosis, Pathophysiology, Therapeutic Avenues, and the Potential of Natural Extracts" Current Issues in Molecular Biology 46, no. 5: 4063-4105. https://doi.org/10.3390/cimb46050251

Article Metrics

Back to TopTop