Open Access

Knee osteoarthritis: Current status and research progress in treatment (Review)

  • Authors:
    • Ruizhi Geng
    • Jiayi Li
    • Chen Yu
    • Chaoqun Zhang
    • Fei Chen
    • Jie Chen
    • Haonan Ni
    • Jiaxu Wang
    • Kaiqiang Kang
    • Ziqi Wei
    • Yongqing Xu
    • Tao Jin
  • View Affiliations

  • Published online on: August 25, 2023     https://doi.org/10.3892/etm.2023.12180
  • Article Number: 481
  • Copyright: © Geng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Knee osteoarthritis (KOA) is a common chronic articular disease worldwide. It is also the most common form of OA and is characterized by high morbidity and disability rates. With the gradual increase in life expectancy and ageing population, KOA not only affects the quality of life of patients, but also poses a burden on global public health. OA is a disease of unknown etiology and complex pathogenesis. It commonly affects joints subjected to greater loads and higher levels of activity. The knee joint, which is the most complex joint of the human body and bears the greatest load among all joints, is therefore most susceptible to development of OA. KOA lesions may involve articular cartilage, synovium, joint capsule and periarticular muscles, causing irreversible articular damage. Factors such as mechanical overload, inflammation, metabolism, hormonal changes and ageing serve key roles in the acceleration of KOA progression. The clinical diagnosis of KOA is primarily based on combined analysis of symptoms, signs, imaging and laboratory examination results. At present, there is no cure for KOA and the currently available therapies primarily focus on symptomatic treatment and delay of disease progression. Knee replacement surgery is typically performed in patients with advanced disease. The current study presents a review of epidemiological characteristics, risk factors, histopathological manifestations, pathogenesis, diagnosis, treatment modalities and progress in KOA research.

1. Introduction

Knee osteoarthritis (KOA) is a multi-etiological, chronic disabling disease that affects the entire knee joint, which is the most common site of involvement in OA (1). KOA is classified as primary or secondary depending on etiology. The pathogenesis of primary KOA is complex and involves numerous factors, such as mechanical stress, inflammation, metabolism, immunity and genetics, with age, genetics, body weight, sex and race being risk factors (2,3). By contrast, secondary KOA is primarily caused by either trauma, congenital articular dysplasia or iatrogenic injury. The pathological changes of KOA are not passive degenerative or wear-and-tear lesions but active changes caused by an imbalance between articular tissue damage and repair (4). They are typically accompanied by lesions in articular or subchondral bones, ligaments, synovium, joint capsule and periarticular muscular structures (5). The primary clinical manifestations are pain and limited mobility, which reduce the quality of life of patients (6). Surveys have indicated a higher incidence of KOA symptoms in the Chinese population aged ≥60 (19.4%) (7), with the incidence rate in women (10.3%) being higher than that in men (5.7%) (8). The ageing population has led to an increase in the proportion of older people with OA. Therefore, OA has become a major healthcare and economic burden (9). Currently available pharmacological therapies such as NSAIDs are palliative as they are mainly aimed at symptomatic relief of inflammation and pain and the delay of disease progression (10). There are presently no drugs that halt disease progression or reverse pathological changes in the entire joint and most patients with end-stage KOA require surgical treatment (11,12). The definition of KOA, as well as identification of risk factors and pathophysiological mechanisms, are being improved and developed as researchers investigate pathogenesis of the disease (13). However, the scientific, rational and effective treatment of KOA remains a challenge in clinical practice (14). The present study reviews the epidemiological characteristics, risk factors, histopathological manifestation, pathogenesis, diagnosis, treatment modalities and the latest KOA research.

2. Epidemiology

KOA primarily occurs in ≥50-year-old individuals (15). It is a chronic degenerative joint disease that clinically manifests as pain, joint deformity and limited mobility that typically causes disability (16). With the acceleration of population ageing and the increase in the proportion of obese individuals, KOA is the 11th leading cause of disability worldwide and ranks 38th among disability factors that affect life expectancy (17). In developed regions, such as North America, Western Europe, Japan, Australia and New Zealand, the prevalence of KOA is ~22.0% in men aged ≥80 and 30.3% in elderly women; in the Western Pacific region, the corresponding prevalence rates are 13.0 and 20.5%, respectively, with prevalence increasing considerably in individuals aged ≥45 (14.1% for men and 22.8% for women) (18). The prevalence of KOA in China is ~18% (19,20). Prevalence rate is notably higher among women than men, especially in the population aged ≥60 years (10.0% for men and 37.3% for women) (19,20). The median age of KOA diagnosis is 55 years (21). With rapid increase in the elderly population of China, the incidence of KOA has risen. Besides causing physical pain and dysfunction, KOA leads to an increase in anxiety, helplessness, depression and social barriers at both social and psychological level; this affects daily life, social function and quality of life of patients, brings economic burdens and pressure to families and social healthcare and poses a challenge to social health (22). The pain and disability caused by KOA posed economic burdens worldwide, with the cost amounting to 1.0-2.5% of the gross national product in developed countries (23).

3. Risk factors

Age

In the United States, Osteoarthritis is the second leading cause of incapacity in men >50 years of age (24). The disease is also one of the main causes of disability (24) and affects the quality of life and economic status of patients. Therefore, greater emphasis on the prevention and treatment of KOA in older people is important to decrease disability rates and improve quality of life in the elderly population.

Genetics

Family history is a risk factor for KOA. In a previous study, the heritability frequencies of femoral, tibial, patellar and total cartilage volumes were estimated to be 61, 76, 66 and 73%, respectively (25). Given the relatively high heritability of tibial cartilage volume and greater susceptibility of the elderly to tibial fractures, attention should be paid to the protection of the tibia during middle and older age. KOA is also associated with abnormalities in the COL2A1 gene, which is associated with type II collagen synthesis. One-third of the mutations in the COL2A1 gene are dominant negative mutations, affecting glycine residues in the α1 chain G-X-Y repeat sequence. These mutations disrupt the triple helix structure of collagen and are common in type II achondroplasia and hypochondroplasia (26). Which suggests that KOA is influenced by genetics (27).

Body mass index

High body mass index is a risk factor for KOA. A previous study reported a 35% increase in KOA risk with every 5-unit increase in body mass index (28). This is primarily attributed to the fact that greater body weight increases the weight-bearing pressure of the knee joint, thereby increasing the probability of KOA.

Sex

Sex differences exist in the incidence rates of KOA. In rural Tianjin, the prevalence of KOA was higher in women than in men (14.1% vs. 6.5%) (29-32). The causes of sex differences in KOA incidence remain unclear and may be associated with estrogen levels (33).

Race

Race affects the incidence of KOA. Through the investigation of the prevalence of KOA among the elderly population in an urban area in China and the white population in the United States, it was shown that the prevalence rates of radiological and clinical KOA in Chinese women were 46.6 and 15.4%, respectively, which were considerably higher than the rates in white American women of the same age group, 34.8 and 11.6% respectively. The radiological and clinical KOA prevalence rates in elderly Chinese men were 27.6 and 7.1% (34), respectively, similar to those in white American men, 30.8 and 6.9% respectively (35).

Trauma

KOA is associated with articular injuries, including articular surface fractures, joint dislocation and ligament and meniscus injury (36). These traumatic injuries increase the incidence of KOA to different extents. For example, there a near 7-fold and 8-fold increase in the odds for the development of KOA post ACL injury and ACL reconstruction have been reported (37).

Other risk factors

Humid, cold and dark living environments (38-40), joint load (41) and professional sports training (42) are risk factors for KOA.

KOA occurs as a result of combined effects of multiple factors, but there is currently no consensus on the exact risk factors. Specifically, age, genetics, body weight, sex, race and trauma have been recognized as risk factors by a number of studies, but opinion is divided on the roles of diet, smoking and exercise, which require further investigation (43-46). This article summarizes the risk factors, pathogenesis and treatment of KOA in the form of graphs (Fig. 1).

4. Histopathological manifestation

The primary histopathological characteristics of KOA include degradation and degenerative changes of the articular cartilage matrix. However, these are not limited to articular cartilage and may even involve the entire joint. Other manifestations include articular cartilage softening, fibrosis, ulceration, and the loss of articular cartilage, synovial hyperemia, swelling and hyperplasia, subchondral bone sclerosis and eburnation and osteophyte and subchondral cyst formation (47,48).

5. Pathogenesis

Factors influencing cartilage synthesis and degradation

During development of KOA, the balance between cartilage synthesis and degradation is primarily affected by pro-degenerative cytokines, such as interleukins (ILs) and tumour necrosis factor-α (TNF-α), and pro-anabolic factors such as insulin-like growth factor 1 (IGF-1), transforming growth factor-β (TGF-β) and bone morphogenetic proteins (BMPs) (49). Pro-degenerative factors primarily include IL-1β, -6, -15, -17 and -18 and TNF-α, with IL-1β and TNF-α being strong activators of cartilage extracellular matrix (ECM) degradation. Pro-anabolic factors include IGF-1, members of the TGF-β family, BMP-2 and -7, IL-4 and -10 and anti-inflammatory or regulatory cytokines secreted by the synovium, cartilage or other tissues, including fibroblast growth factor 2 and IL-4, -6, -10 and -14 (50-52). Free radicals are also associated with cartilage destruction and matrix degradation in KOA. For example, reactive oxygen species can inhibit the proliferation of articular chondrocytes, promote their apoptosis and cause an imbalance in synthesis and degradation of collagen and proteoglycans (PGs) in the cartilage matrix, thereby inducing cartilage injury (53).

Imbalance between ECM synthesis and degradation

ECM, synthesized and secreted by chondrocytes, is primarily composed of water, collagen and PGs. These components form a complex network structure that provides nutrients and support to chondrocytes and serves as a site for physiological activity of chondrocytes (54). A previous study showed that cartilage injury in KOA occurs primarily in the form of ECM degeneration (55), which manifests as an imbalance between degradation and synthesis. Signaling pathways associated with degradation comprise MAPKs, NF-κB, the Wnt/β-catenin and Notch signaling pathways and toll-like receptors (56).

Excessive chondrocyte apoptosis

A small amount of apoptosis occurs in normal cartilage and is generally limited to the surface layer. It is an essential physiological process for growth, development, functional regulation and maintenance of internal environment stability in cartilage (57,58). However, excessive apoptosis of cartilage is a pathological process and one of the pathogenic mechanisms of KOA (59). In a previous study, flow cytometry data showed that ~5% of normal chondrocytes and 22% of OA chondrocytes undergo apoptosis. Staining of cartilage sections revealed that apoptotic cells existed on the surface and middle cartilage layers, with PGs notably decreased in apoptotic regions, and the number of apoptotic cells was positively associated with KOA severity (60). Chondrocyte apoptosis in KOA is primarily regulated by the mitochondrial, death receptor and endoplasmic reticulum stress response pathways (61,62). A previous study investigated the widely recognized Fas pathway among death receptor pathways, and the NO pathway among the various mitochondrial pathways (62-66). Genes involved in regulation of apoptosis mainly include the BCL-2 and interlukin-1β-converting enzyme (ICE) gene families and the p53 and c-myc genes.

Subchondral bone lesions

Subchondral bone hardening and osteophyte formation, known as bone remodeling, are characteristic manifestations of KOA (67). Studies have shown that subchondral bone lesions appear during the early stage of KOA and may occur earlier than cartilage degeneration (68). Although there is a lack of consensus in this aspect, it has been widely recognized that subchondral bone lesions promote cartilage degeneration (68-70).

6. Diagnosis

KOA is primarily diagnosed based on clinical symptoms, signs, imaging examinations and laboratory examinations.

Symptoms

KOA primarily manifests as knee joint pain that is aggravated by fatigue or weather changes and cold and wet weather, and relieved after rest or the application of heat. Patients typically experience either dull and needle-like pain or joint soreness (71). The pain originates from tissue around the knee joint, such as subchondral bone microfractures, synovitis, joint capsular strain caused by osteohypertrophy and increased intraosseous venous pressure (72). In the early stage of disease, pain is only felt during continuous loading or overuse of the joint. When joint weakness or movement disorders develop during middle-to-late stages of the disease, pain occurs with mild exertion or during rest or night-time (73). Patients also experience limited joint mobility upon waking in the morning, known as morning stiffness, which usually lasts <30 min (74) and is limited to the affected joints.

Signs

A physical examination should be performed to determine whether symptoms are associated with joints. It usually consists of joint palpation, testing of the active and passive range of motion of joints and assessment of pain using the pain index. Common signs include the following: i) Tenderness and tactile pain in the knee joint and joint edges, with tender points mostly located near the joint line, joint capsule and insertion points of the collateral ligament. This may be associated with muscle spasms, bursitis and tendonitis (71); ii) hypertrophy and swelling of the knee joint resulting from osteophyte formation leading to osteohypertrophy with or without aseptic inflammatory exudation from the synovial membrane within the joint, which may be accompanied by an increase in local skin temperature of the knee joint (75); iii) obviously palpable articular crepitus sounds during active or passive joint flexion and extension, primarily related to articular cartilage detachment and degeneration of joints such as the patella and femur, which result in uneven articular surfaces that rub against each other and the presence of cartilage fragments in the joint (5); iv) limited joint range of motion and movement abnormalities, which may be related to pain, exudate adhesion, flexion contracture, muscle spasms, cartilage loss, degeneration, mechanical obstruction caused by cartilage or the menisci or intra-articular loose bodies (76) and v) joint varus deformity or valgus deformity (77).

Imaging examinations

X-ray is the first-choice imaging modality in the diagnosis of KOA. In general, anteroposterior and lateral radiographs of the knee joint, particularly the weight-bearing views, are required for the comparison of bilateral knee joints (78). In the early stage of disease, X-ray findings are usually negative, with small osteophytes occasionally seen on the superior and inferior patellar edges. Advanced disease manifests as narrowing of the joint space, bone sclerosis, cystic changes, osteophyte formation along joint edges, subchondral ossification or cystic changes, or joint deformation (79). Further disease progression leads to the occurrence of manifestations such as subchondral bone osteosclerosis, subarticular cysts, bone resorption and intra-articular loose bodies (80). The Chinese Medical Association Guidelines for the Diagnosis and Treatment of OA (2018 edition) summarized the three typical X-ray manifestations of OA, namely asymmetric joint space narrowing in the affected joints, subchondral bone sclerosis or cystic changes and osteophyte formation along joint edges (39).

Computed tomography (CT) and magnetic resonance imaging (MRI) are primarily used for differential diagnosis. CT possesses limitations, such as for certain minor soft tissue injuries, CT images may not be sensitive enough and is less commonly used in clinical practice (81). It is primarily used to locate loose bodies or debris in the joint, and to determine patellofemoral joint structure (82). MRI can show lesions in the cartilage, bone marrow and soft tissue of the knee joint, provide indications of early cartilage and synovial lesions and reveal structural abnormalities in periarticular tissues such as ligaments and menisci (83). It also enables the evaluation of bone resorption around the knee joint. With semi-quantitative and quantitative analysis of cartilage, the degree of lesions can be evaluated in a more accurate manner, which provides early diagnostic value (84). Quantitative T2-weighted MRI may serve as an indicator for early KOA diagnosis and evaluation of disease condition (85-89), but it is of limited use in the guidance of clinical treatment of KOA (90) as it is difficult to distinguish inflammatory arthritis OA with MRI.

Ultrasound examinations are of use in detecting joint exudation, cartilage lesions and popliteal cysts and in guiding punctures and injections in joints and the surrounding soft tissue (91). Radionuclide bone imaging assists in determining the metabolic status of osseous tissue, which contributes to early diagnosis (92).

Laboratory examination

At present, no specific clinical laboratory markers of KOA are known (30). A previous study showed that ESR is associated with joint function limitation, whereas CRP is associated with severe KOA (93). However, further research is required to ascertain whether ESR and CRP serve as indicators for disease diagnosis and monitoring (93). Serum IGF-1 expression and matrix metalloproteinase (MMP)-3 expression are associated with osteophyte formation and articular index in patients with KOA, respectively (94,95).

7. Research in KOA treatment

The Guidelines for the Diagnosis and Treatment of OA (2018 Edition) published in China first proposed the concept and strategy of stepwise treatment. Basic treatment, including patient education, exercise therapy, physical therapy and action support therapy, is applicable to all patients with OA. Appropriate basic treatment regimens can be selected for patients with early-stage KOA. For those with aggravated disease, secondary pharmacological treatment can be administered with the appropriate drug administration routes and drug types selected based on affected sites and risk factors of the patient. If basic and pharmacological treatment prove ineffective, surgical treatment can be performed. The surgical plan should be developed by taking the lesion site and degree, general condition and willingness of the patient into consideration (39). At present, the treatment principle for KOA is combined use of non-pharmacological and pharmacological therapies, with surgery only performed when necessary. Treatment of symptomatic KOA should be aimed at the following: i) Control 0of symptoms; ii) improvement of limited joint function; iii) enhancement of quality of life; iv) reduction of disability rate and v) avoidance of excessive medication. In 2000, the American College of Rheumatology (ACR) and the European League Against Rheumatism (EULAR) recommended the following treatment options for patients with KOA: Non-pharmacological, pharmacological and surgical treatment options (96).

Non-pharmacological treatment

Health education of patients. KOA-related knowledge can be provided to patients through health education to promote positive and optimistic attitudes towards the disease. Measures such as maintenance of good lifestyle habits, regular exercise, avoidance of weight-bearing activities and body weight reduction contribute to the enhancement of self-management and exert good preventive and stabilizing effects during the rehabilitation stage. Therefore, health education is essential for treatment of KOA.

Exercise therapy. Exercise therapy, which possesses advantages such as ease of operation, lack of adverse reaction (including liver and kidney damage and gastrointestinal irritation), effective pain relief and improvement of joint function, is by various guidelines as the first-choice treatment method for KOA (97). Various exercise training regimens have been reported to have therapeutic effects on KOA. Aerobic exercise is the most widely used and can reduce pain and improve physical function (98). Strength training is the most effective exercise therapy against muscle weakness (99). The research basis for exercise training in patients with KOA has been stated in recommended regimens for non-pharmacological treatment of arthritis proposed by the EULAR in 2013(53). Forms of exercise therapy recommended by various guidelines on KOA treatment published worldwide primarily include aerobic and aquatic exercise and muscle strength training (97).

Physiotherapy. Clinical application of physiotherapy improves blood circulation, provides anti-inflammatory and analgesic effects, and assists in the alleviation of symptoms (100). Physiotherapy techniques primarily used in the treatment of KOA include hot compression, aquatic therapy, acupuncture and electromagnetic and ultra-short-wave therapy (101,102).

Pharmacological treatment

EULAR has classified pharmacological agents for KOA treatment into two primary categories, namely symptom- and disease-modifying drugs (103-105). Traditional Chinese medicine and Chinese herbal medications can also be used for the treatment of KOA (106). Modern pharmacological studies have shown that Yanghe decoction has the effect of preventing chondrocyte apoptosis and exerts an anti-inflammatory action during KOA treatment (107,108).

Symptom-modifying drugs. Acetaminophen is the most representative analgesic and the primary drug used to control symptoms in patients with mild-to-moderate KOA. It causes less gastrointestinal irritation, has a good safety and tolerability profile and is less likely to cause gastrointestinal bleeding, which makes it suitable for use in elderly patients. However, its analgesic effects are relatively weak, making it more suitable for patients with early-stage KOA or milder symptoms. In 2000, the ACR and EULAR recommended acetaminophen as the first-choice therapy for the treatment of OA, but large doses or long-term use may lead to liver or kidney damage (109).

Non-steroidal anti-inflammatory drugs (NSAIDs) exert anti-inflammatory effects by decreasing the synthesis of prostaglandins through the inhibition of cyclooxygenase (COX) and are used as first-line therapy in KOA treatment (110). NSAIDs include salicylic and phenylpropionic acid, acetic acid derivatives, fenamates, enolic acids, naphthalenones and COX inhibitors, with acetylsalicylic acid, ibuprofen, naproxen, indomethacin and sulindac commonly used in clinical practice (111). Traditional NSAIDs produce toxic side effects due to the lack of selectivity towards COX-1 or COX-2(112). To address this, second-generation NSAIDs, a class of COX-2 selective inhibitors, have been designed to avoid adverse reactions associated with traditional NSAIDs, including gastrointestinal complications, cardiovascular events, renal toxicity, the exacerbation of hypertension and fluid retention (113). Representative drugs include celecoxib and rofecoxib. Studies have demonstrated that celecoxib enables an increase in PG content of the cartilage matrix, cartilage repair and delay of cartilage degeneration (114,115). However, COX-2 selective inhibitors increase risk of cardiovascular disease (116). Therefore, consideration of various factors and rational drug use is necessary for the clinical application of NSAIDs.

Inhibition of inflammation and alleviation of symptoms can be achieved through aspiration of joint effusion and subsequent injection of corresponding medications (117). Steroid hormones (118) and hyaluronic acid (119) are commonly used as intracavitary injection drugs. The intra-articular injection of glucocorticoids enables inhibition of abnormal connective tissue proliferation, reduction of synovitis, significant alleviation of joint pain and improvement of joint function (120). However, it is important to note that repeated injections of intra-articular glucocorticoids over a long period of time can lead to cartilage loss (121). Hyaluronic acid attenuates joint pain, increases joint mobility, decreases synovitis and promotes anabolism of chondrocytes in patients with OA, but the exact mechanisms of action require further investigation (122). Both glucocorticoids and hyaluronic acid effectively relieve joint pain and swelling but should not be used repeatedly due to the damaging effects of injections on cartilage, such as scratching of the cartilage by the syringe needle (123,124). Additionally, intra-articular injections of autologous platelet-rich plasma for treatment of KOA have demonstrated good therapeutic effects (125,126) and possess good prospects for application in clinical practice.

Antidepressants (127) such as duloxetine provide antidepressant, central pain-suppressing and anxiolytic effects. Topical medications such as capsaicin (128), diclofenac diethylamine cream (129) and methyl salicylate (130) relieve the symptoms of joint pain to a certain extent. A study has also shown that statins decrease the incidence of KOA and delay its progression (131). Other studies have demonstrated that salmon calcitonin not only improves symptoms of KOA, but also exerts cartilage-protecting effects (132,133).

Disease-modifying drugs. As the basic substance in the synthesis of polyglucosamine and hyaluronic acid, glucosamine is an important component required for PG synthesis in articular cartilage matrix. It acts selectively on articular cartilage, promotes PG and hyaluronic acid synthesis, maintains morphology and structure of the cartilage ECM and serves an important role in articular cartilage repair and synovial fluid production (134). Other effects include inhibition of the generation of free oxygen radicals, collagenases, prostaglandin E2 and phospholipase A2, which protect articular cartilage and contribute to the delay of OA progression (135). In European countries, glucosamine sulphate is the first-choice prescription drug for the treatment of KOA and is thought to be delay or reverse pathological changes of OA (136). However, adverse reactions such as nausea and vomiting, abdominal distension and diarrhea and bone aches and pain may arise during clinical application of glucosamine sulphate.

Chondroitin sulphate is glycosaminoglycan that promotes the anabolism of chondrocytes, stimulates PG and hyaluronic acid synthesis and inhibits ECM degradation by enzymes, which contribute to relief of joint pain and improvement of joint function (137).

Diacerein is an anthraquinone is extracted from Da Huang (Radix et Rhizoma Rhei). It inhibits inflammatory factors and MMPs, promotes the synthesis of growth factors and stabilizes lysosomal membranes, thereby achieving anti-inflammatory, analgesic and cartilage-protecting effects (138). A previous study demonstrated that the combined use of glucosamine and diacerein improves meniscal damage (139). Diarrhea may occur as a rare adverse reaction.

MMPs may degrade type II collagen and PGs in the ECM, which results in cartilage destruction (140). Tetracyclines (141) inhibit MMP-1 release from type B synoviocytes, as well as nitrous oxide production. This leads to the reduction of damage to knee articular cartilage by inflammatory mediators, which contributes to the improvement of disease in patients with KOA (141). The development of MMP inhibitors has gained increasing research interest in the field of pharmacological therapies for OA (142).

Antioxidants (143) such as superoxide dismutase (144), vitamins C, D and E can provide cartilage-protecting effects by blocking free oxygen radical pathways (145-148). Other drugs such as calcitonin (149) and statins (150) alleviate the disease condition of patients with KOA.

Surgical treatment

Surgical treatment is recommended for patients with advanced KOA who respond poorly after 6 months of conservative treatment, and whose condition severely affects daily living (103,151). However, the effects of surgical treatment may be influenced by factors such as pain, joint function, anatomical factors and patient's physical function (152). Current surgical treatment methods for KOA include knee arthroscopy (153), bone marrow stimulation (154) and osteotomy (155-157) and joint replacement (158), fusion (159) and arthroplasty (160). For patients with early-to-mid-stage KOA, joint arthroscopy is a better option as it provides the advantages of a smaller wound and rapid recovery (153). Artificial joint replacement, which is the terminal treatment for KOA, may be considered for patients with advanced KOA who experience joint ankylosis, severe damage to joint structures and intense pain (158). However, there is need for further exploration into the prevention and treatment of postoperative complications following artificial joint replacement (159). Osteotomy techniques such as high tibial osteotomy are primarily used for the treatment of KOA with varus and valgus deformities (161).

8. Conclusion

KOA is a common chronic disease in orthopedic practice and one of the primary causes of disability and pain in older people (162). Although etiology and pathogenesis of KOA have not yet been fully elucidated, research on its pathogenic mechanisms has shifted from the macroscopic level, including looking at biomechanics, articular cartilage and subchondral bone lesions to the microscopic level, including investigation of inflammatory cytokines and neural mechanisms (140). However, current research remains limited (9). For example, certain progress has been made in the investigation of KOA-associated inflammatory cytokines (163) from the single-factor perspective, but the complex mechanisms of association among multiple factors have not yet been determined (164). Given the lack of early symptoms and signs and specific diagnostic indicators in imaging and laboratory examinations, differential diagnosis of KOA in clinical practice is of importance. Clinicians are required to make judgments based on analysis of risk factors, symptoms, signs and results of imaging and laboratory examinations. Identifying specific diagnostic markers via measurement of markers in the peripheral blood, joint fluid and other body fluids of patients may facilitate early detection, diagnosis and treatment of KOA. However, the combination of various factors, such as risk factors, symptoms, signs, imaging and laboratory findings is required for confirmed diagnosis and staging to provide a basis for clinical treatment.

Numerous factors, including risk factors, psychosocial factors, symptoms, signs, imaging and laboratory findings, should be taken into consideration for the determination of KOA treatment regimens as disease characteristics and general conditions differ between individuals. Prevention should take priority, followed by individualized and standardized treatment via formulation of treatment regimens suited to every patient to maximize clinical effects. KOA treatment is currently limited to symptomatic therapy such as anti-inflammatory treatment, analgesia and limited functional improvement. In previous studies, researchers have developed pharmacological agents to promote cartilage repair and regeneration (165,166), inhibit cartilage degradation (167), promote cartilage matrix secretion (168), specifically inhibit inflammatory factors and pathways (169) and maintain the joint cavity environment (170,171). However, the majority of these drugs are still undergoing clinical trials. In addition, sociopsychological support should also be provided to patients during treatment process to assist in the recovery of social function. With the continuous improvements in understanding of the pathogenesis of KOA, it is anticipated that drugs that directly target pathogenic factors of the disease will emerge in the future and create new breakthroughs in disease treatment. It is anticipated that KOA may be prevented, controlled or cured through active intervention rather than passive treatment. Future research efforts should be directed towards the prevention of KOA, etiological treatment, maximum recovery of joint structure and function and the limitation or even reversal of KOA progression.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the Yunnan Provincial Clinical Orthopedic Trauma Medical Center (grant no. ZX20191001) and the Science and Technology Plan Project of The Science and Technology Department of Yunnan Province (grant no. 202101AY070001-296).

Availability of data and materials

Not applicable.

Authors' contributions

YX and TJ conceived the study. RG and JL designed the study and drafted, reviewed and edited the manuscript. CY, CZ, FC and JW wrote the manuscript. JC, HN, KK and ZW analysed the literature. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Prieto-Alhambra D, Judge A, Javaid MK, Cooper C, Diez-Perez A and Arden NK: Incidence and risk factors for clinically diagnosed knee, hip and hand osteoarthritis: Influences of age, gender and osteoarthritis affecting other joints. Ann Rheum Dis. 73:1659–1664. 2014.PubMed/NCBI View Article : Google Scholar

2 

Busija L, Bridgett L, Williams SR, Osborne RH, Buchbinder R, March L and Fransen M: Osteoarthritis. Best Pract Res Clin Rheumatol. 24:757–768. 2010.PubMed/NCBI View Article : Google Scholar

3 

Felson DT, Lawrence RC, Dieppe PA, Hirsch R, Helmick CG, Jordan JM, Kington RS, Lane NE, Nevitt MC, Zhang Y, et al: Osteoarthritis: New insights. Part 1: The disease and its risk factors. Ann Intern Med. 133:635–646. 2000.PubMed/NCBI View Article : Google Scholar

4 

Michael JW, Schlüter-Brust KU and Eysel P: The epidemiology, etiology, diagnosis, and treatment of osteoarthritis of the knee. Dtsch Arztebl Int. 107:152–162. 2010.PubMed/NCBI View Article : Google Scholar

5 

Andriacchi TP, Mündermann A, Smith RL, Alexander EJ, Dyrby CO and Koo S: A framework for the in vivo pathomechanics of osteoarthritis at the knee. Ann Biomed Eng. 32:447–457. 2004.PubMed/NCBI View Article : Google Scholar

6 

Dawson J, Linsell L, Zondervan K, Rose P, Carr A, Randall T and Fitzpatrick R: Impact of persistent hip or knee pain on overall health status in elderly people: A longitudinal population study. Arthritis Rheum. 53:368–374. 2005.PubMed/NCBI View Article : Google Scholar

7 

Xiang YJ and Dai SM: Prevalence of rheumatic diseases and disability in China. Rheumatol Int. 29:481–490. 2009.PubMed/NCBI View Article : Google Scholar

8 

Tang X, Wang S, Zhan S, Niu J, Tao K, Zhang Y and Lin J: The prevalence of symptomatic knee osteoarthritis in China: Results from the China Health and retirement longitudinal study. Arthritis Rheumatol. 68:648–653. 2016.PubMed/NCBI View Article : Google Scholar

9 

Sarzi-Puttini P, Cimmino MA, Scarpa R, Caporali R, Parazzini F, Zaninelli A, Atzeni F and Canesi B: Osteoarthritis: An overview of the disease and its treatment strategies. Semin Arthritis Rheum. 35:1–10. 2005.PubMed/NCBI View Article : Google Scholar

10 

Bannuru RR, Osani MC, Vaysbrot EE, Arden NK, Bennell K, Bierma-Zeinstra SMA, Kraus VB, Lohmander LS, Abbott JH, Bhandari M, et al: OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthritis Cartilage. 27:1578–1589. 2019.PubMed/NCBI View Article : Google Scholar

11 

Brophy RH and Fillingham YA: AAOS Clinical practice guideline summary: Management of osteoarthritis of the Knee (Nonarthroplasty), Third edition. J Am Acad Orthop Surg. 30:e721–e729. 2022.PubMed/NCBI View Article : Google Scholar

12 

Peng H, Ou A, Huang X, Wang C, Wang L, Yu T and Zhang Y and Zhang Y: Osteotomy around the knee: The surgical treatment of osteoarthritis. Orthop Surg. 13:1465–1473. 2021.PubMed/NCBI View Article : Google Scholar

13 

Jang S, Lee K and Ju JH: Recent updates of diagnosis, pathophysiology, and treatment on osteoarthritis of the knee. Int J Mol Sci. 22(2619)2021.PubMed/NCBI View Article : Google Scholar

14 

Bichsel D, Liechti FD, Schlapbach JM and Wertli MM: Cross-sectional analysis of recommendations for the treatment of hip and knee osteoarthritis in clinical guidelines. Arch Phys Med Rehabil. 103:559–569.e5. 2022.PubMed/NCBI View Article : Google Scholar

15 

Bahar E, Shamarina D, Sergerie Y and Mukherjee P: Descriptive overview of pertussis epidemiology among older adults in Europe during 2010-2020. Infect Dis Ther. 11:1821–1838. 2022.PubMed/NCBI View Article : Google Scholar

16 

Madry H, Kon E, Condello V, Peretti GM, Steinwachs M, Seil R, Berruto M, Engebretsen L, Filardo G and Angele P: Early osteoarthritis of the knee. Knee Surg Sports Traumatol Arthrosc. 24:1753–1762. 2016.PubMed/NCBI View Article : Google Scholar

17 

Cross M, Smith E, Hoy D, Nolte S, Ackerman I, Fransen M, Bridgett L, Williams S, Guillemin F, Hill CL, et al: The global burden of hip and knee osteoarthritis: estimates from the global burden of disease 2010 study. Ann Rheum Dis. 73:1323–1330. 2014.PubMed/NCBI View Article : Google Scholar

18 

Woolf AD and Pfleger B: Burden of major musculoskeletal conditions. Bull World Health Organ. 81:646–656. 2003.PubMed/NCBI

19 

Abbassy AA, Trebinjac S and Kotb N: The use of cellular matrix in symptomatic knee osteoarthritis. Bosn J Basic Med Sci. 20:271–274. 2020.PubMed/NCBI View Article : Google Scholar

20 

Zhang Y, Xu L, Nevitt MC, Aliabadi P, Yu W, Qin M, Lui LY and Felson DT: Comparison of the prevalence of knee osteoarthritis between the elderly Chinese population in Beijing and whites in the United States: The Beijing Osteoarthritis Study. Arthritis Rheum. 44:2065–2071. 2001.PubMed/NCBI View Article : Google Scholar

21 

(AUS) AOA: Lay summary 2015 annual report hip and knee replacement: Suppliment report, 2015.

22 

Zhou Z, Cui J, Wu S, Geng Z and Su J: Silk fibroin-based biomaterials for cartilage/osteochondral repair. Theranostics. 12:5103–5124. 2022.PubMed/NCBI View Article : Google Scholar

23 

Hiligsmann M, Cooper C, Arden N, Boers M, Branco JC, Luisa Brandi M, Bruyère O, Guillemin F, Hochberg MC, Hunter DJ, et al: Health economics in the field of osteoarthritis: An Expert's consensus paper from the European Society for Clinical and Economic Aspects of Osteoporosis and Osteoarthritis (ESCEO). Semin Arthritis Rheum. 43:303–313. 2013.PubMed/NCBI View Article : Google Scholar

24 

Arden N and Nevitt MC: Osteoarthritis: Epidemiology. Best Pract Res Clin Rheumatol. 20:3–25. 2006.PubMed/NCBI View Article : Google Scholar

25 

Hunter DJ, Snieder H, March L and Sambrook PN: Genetic contribution to cartilage volume in women: A classical twin study. Rheumatology (Oxford). 42:1495–1500. 2003.PubMed/NCBI View Article : Google Scholar

26 

Barat-Houari M, Sarrabay G, Gatinois V, Fabre A, Dumont B, Genevieve D and Touitou I: Mutation update for COL2A1 gene variants associated with type II collagenopathies. Hum Mutat. 37:7–15. 2016.PubMed/NCBI View Article : Google Scholar

27 

Chapman K and Valdes AM: Genetic factors in OA pathogenesis. Bone. 51:258–264. 2012.PubMed/NCBI View Article : Google Scholar

28 

Jiang L, Tian W, Wang Y, Rong J, Bao C, Liu Y, Zhao Y and Wang C: Body mass index and susceptibility to knee osteoarthritis: A systematic review and meta-analysis. Joint Bone Spine. 79:291–297. 2012.PubMed/NCBI View Article : Google Scholar

29 

Ji S, Liu L, Li J, Zhao G, Cai Y, Dong Y, Wang J and Wu S: Prevalence and factors associated with knee osteoarthritis among middle-aged and elderly individuals in rural Tianjin: A population-based cross-sectional study. J Orthop Surg Res. 18(266)2023.PubMed/NCBI View Article : Google Scholar

30 

Hunter DJ and Felson DT: Osteoarthritis. BMJ. 332:639–642. 2006.PubMed/NCBI View Article : Google Scholar

31 

Jordan JM, Luta G, Renner JB, Dragomir A, Hochberg MC and Fryer JG: Ethnic differences in self-reported functional status in the rural south: The Johnston County Osteoarthritis Project. Arthritis Care Res. 9:483–491. 1996.PubMed/NCBI View Article : Google Scholar

32 

Srikanth VK, Fryer JL, Zhai G, Winzenberg TM, Hosmer D and Jones G: A meta-analysis of sex differences prevalence, incidence and severity of osteoarthritis. Osteoarthritis Cartilage. 13:769–781. 2005.PubMed/NCBI View Article : Google Scholar

33 

Shu Z, Miao X, Tang T, Zhan P, Zeng L and Jiang Y: The GSK-3β/β-catenin signaling pathway is involved in HMGB1-induced chondrocyte apoptosis and cartilage matrix degradation. Int J Mol Med. 45:769–778. 2020.PubMed/NCBI View Article : Google Scholar

34 

Xu L, Nevitt MC, Zhang Y, Yu W, Alibadi P and Felson DT: High prevalence of knee, but not hip or hand osteoarthritis in Beijing elders: Comparison with data of Caucasian in United States. Zhonghua Yi Xue Za Zhi. 83:1206–1209. 2003.PubMed/NCBI(In Chinese).

35 

Guccione AA, Felson DT, Anderson JJ, Anthony JM, Zhang Y, Wilson PW, Kelly-Hayes M, Wolf PA, Kreger BE and Kannel WB: The effects of specific medical conditions on the functional limitations of elders in the Framingham Study. Am J Public Health. 84:351–358. 1994.PubMed/NCBI View Article : Google Scholar

36 

Øiestad BE, Engebretsen L, Storheim K and Risberg MA: Knee osteoarthritis after anterior cruciate ligament injury: A systematic review. Am J Sports Med. 37:1434–1443. 2009.PubMed/NCBI View Article : Google Scholar

37 

Webster KE and Hewett TE: Anterior cruciate ligament injury and knee osteoarthritis: An umbrella systematic review and meta-analysis. Clin J Sport Med. 32:145–152. 2022.PubMed/NCBI View Article : Google Scholar

38 

Xie F, Li SC, Fong KY, Lo NN, Yeo SJ, Yang KY and Thumboo J: What health domains and items are important to patients with knee osteoarthritis? A focus group study in a multiethnic urban Asian population. Osteoarthritis Cartilage. 14:224–230. 2006.PubMed/NCBI View Article : Google Scholar

39 

Stitik TP and Foye PM: The prevalence of knee pain and symptomatic knee osteoarthritis among veteran traumatic amputees and nonamputees. Arch Phys Med Rehabil. 86(1273)2005.PubMed/NCBI View Article : Google Scholar

40 

Wigley RD, Zhang NZ, Zeng QY, Shi CS, Hu DW, Couchman K, Duff IF and Bennett PH: Rheumatic diseases in China: ILAR-China study comparing the prevalence of rheumatic symptoms in northern and southern rural populations. J Rheumatol. 21:1484–1490. 1994.PubMed/NCBI

41 

Palmer KT: Occupational activities and osteoarthritis of the knee. Br Med Bull. 102:147–170. 2012.PubMed/NCBI View Article : Google Scholar

42 

Lau EC, Cooper C, Lam D, Chan VN, Tsang KK and Sham A: Factors associated with osteoarthritis of the hip and knee in Hong Kong Chinese: Obesity, joint injury, and occupational activities. Am J Epidemiol. 152:855–862. 2000.PubMed/NCBI View Article : Google Scholar

43 

Fuller-Thomson E, Stefanyk M and Brennenstuhl S: The robust association between childhood physical abuse and osteoarthritis in adulthood: Findings from a representative community sample. Arthritis Rheum. 61:1554–1562. 2009.PubMed/NCBI View Article : Google Scholar

44 

Rossignol M, Leclerc A, Allaert FA, Rozenberg S, Valat JP, Avouac B, Coste P, Litvak E and Hilliquin P: Primary osteoarthritis of hip, knee, and hand in relation to occupational exposure. Occup Environ Med. 62:772–777. 2005.PubMed/NCBI View Article : Google Scholar

45 

Ni J, Wang P, Yin KJ, Huang JX, Tian T, Cen H, Sui C, Xu Z and Pan HF: Does smoking protect against developing osteoarthritis? Evidence from a genetically informed perspective. Semin Arthritis Rheum. 55(152013)2022.PubMed/NCBI View Article : Google Scholar

46 

Morales-Ivorra I, Romera-Baures M, Roman-Viñas B and Serra-Majem L: Osteoarthritis and the Mediterranean Diet: A systematic review. Nutrients. 10(1030)2018.PubMed/NCBI View Article : Google Scholar

47 

Uryu N, Okada K and Kawakita K: Analgesic effects of indirect moxibustion on an experimental rat model of osteoarthritis in the knee. Acupunct Med. 25:175–183. 2007.PubMed/NCBI View Article : Google Scholar

48 

Torelli SR, Rahal SC, Volpi RS, Sequeira JL and Grassioto IQ: Histopathological evaluation of treatment with chondroitin sulphate for osteoarthritis induced by continuous immobilization in rabbits. J Vet Med A Physiol Pathol Clin Med. 52:45–51. 2005.PubMed/NCBI View Article : Google Scholar

49 

Kim JR, Yoo JJ and Kim HA: Therapeutics in osteoarthritis based on an understanding of its molecular pathogenesis. Int J Mol Sci. 19(674)2018.PubMed/NCBI View Article : Google Scholar

50 

Wojdasiewicz P, Poniatowski ŁA and Szukiewicz D: The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of osteoarthritis. Mediators Inflamm. 2014(561459)2014.PubMed/NCBI View Article : Google Scholar

51 

Wang Q, Pan X, Wong HH, Wagner CA, Lahey LJ, Robinson WH and Sokolove J: Oral and topical boswellic acid attenuates mouse osteoarthritis. Osteoarthritis Cartilage. 22:128–132. 2014.PubMed/NCBI View Article : Google Scholar

52 

Yoshida A, Morihara T, Matsuda K, Sakamoto H, Arai Y, Kida Y, Kawata M and Kubo T: Immunohistochemical analysis of the effects of estrogen on intraarticular neurogenic inflammation in a rat anterior cruciate ligament transection model of osteoarthritis. Connect Tissue Res. 53:197–206. 2012.PubMed/NCBI View Article : Google Scholar

53 

Sutipornpalangkul W, Morales NP and Harnroongroj T: Free radicals in primary knee osteoarthritis. J Med Assoc Thai. 92 (Suppl 6):S268–S274. 2009.PubMed/NCBI

54 

Guilak F, Nims RJ, Dicks A, Wu CL and Meulenbelt I: Osteoarthritis as a disease of the cartilage pericellular matrix. Matrix Biol. 71-72:40–50. 2018.PubMed/NCBI View Article : Google Scholar

55 

Aurich M, Squires GR, Reiner A, Mollenhauer JA, Kuettner KE, Poole AR and Cole AA: Differential matrix degradation and turnover in early cartilage lesions of human knee and ankle joints. Arthritis Rheum. 52:112–119. 2005.PubMed/NCBI View Article : Google Scholar

56 

Yao Q, Wu X, Tao C, Gong W, Chen M, Qu M, Zhong Y, He T, Chen S and Xiao G: Osteoarthritis: Pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther. 8(56)2023.PubMed/NCBI View Article : Google Scholar

57 

Taniguchi N, Caramés B, Ronfani L, Ulmer U, Komiya S, Bianchi ME and Lotz M: Aging-related loss of the chromatin protein HMGB2 in articular cartilage is linked to reduced cellularity and osteoarthritis. Proc Natl Acad Sci USA. 106:1181–1186. 2009.PubMed/NCBI View Article : Google Scholar

58 

Johnson EO, Charchandi A, Babis GC and Soucacos PN: Apoptosis in osteoarthritis: Morphology, mechanisms, and potential means for therapeutic intervention. J Surg Orthop Adv. 17:147–152. 2008.PubMed/NCBI

59 

Kawaguchi H: Endochondral ossification signals in cartilage degradation during osteoarthritis progression in experimental mouse models. Mol Cells. 25:1–6. 2008.PubMed/NCBI

60 

Hashimoto S, Ochs RL, Rosen F, Quach J, McCabe G, Solan J, Seegmiller JE, Terkeltaub R and Lotz M: Chondrocyte-derived apoptotic bodies and calcification of articular cartilage. Proc Natl Acad Sci USA. 95:3094–3099. 1998.PubMed/NCBI View Article : Google Scholar

61 

Liu L, Luo P, Yang M, Wang J, Hou W and Xu P: The role of oxidative stress in the development of knee osteoarthritis: A comprehensive research review. Front Mol Biosci. 9(1001212)2022.PubMed/NCBI View Article : Google Scholar

62 

Ryu JH, Shin Y, Huh YH, Yang S, Chun CH and Chun JS: Hypoxia-inducible factor-2α regulates Fas-mediated chondrocyte apoptosis during osteoarthritic cartilage destruction. Cell Death Differ. 19:440–450. 2012.PubMed/NCBI View Article : Google Scholar

63 

Chen H, Wang J, Hu B, Wu X, Chen Y, Li R and Yuan W: MiR-34a promotes Fas-mediated cartilage endplate chondrocyte apoptosis by targeting Bcl-2. Mol Cell Biochem. 406:21–30. 2015.PubMed/NCBI View Article : Google Scholar

64 

Chou CT, Yang JS and Lee MR: Apoptosis in rheumatoid arthritis-expression of Fas, Fas-L, p53, and Bcl-2 in rheumatoid synovial tissues. J Pathol. 193:110–116. 2001.PubMed/NCBI View Article : Google Scholar

65 

Lee MS, Trindade MC, Ikenoue T, Goodman SB, Schurman DJ and Smith RL: Regulation of nitric oxide and bcl-2 expression by shear stress in human osteoarthritic chondrocytes in vitro. J Cell Biochem. 90:80–86. 2003.PubMed/NCBI View Article : Google Scholar

66 

Chen WP, Jin GJ, Xiong Y, Hu PF, Bao JP and Wu LD: Rosmarinic acid down-regulates NO and PGE2 expression via MAPK pathway in rat chondrocytes. J Cell Mol Med. 22:346–353. 2018.PubMed/NCBI View Article : Google Scholar

67 

Bijlsma JW, Berenbaum F and Lafeber FP: Osteoarthritis: An update with relevance for clinical practice. Lancet. 377:2115–2126. 2011.PubMed/NCBI View Article : Google Scholar

68 

Wu X, Crawford R, Xiao Y, Mao X and Prasadam I: Osteoarthritic subchondral bone release exosomes that promote cartilage degeneration. Cells. 10(251)2021.PubMed/NCBI View Article : Google Scholar

69 

Mansell JP, Collins C and Bailey AJ: Bone, not cartilage, should be the major focus in osteoarthritis. Nat Clin Pract Rheumatol. 3:306–307. 2007.PubMed/NCBI View Article : Google Scholar

70 

Ajami S, Javaheri B, Chang YM, Maruthainar N, Khan T, Donaldson J, Pitsillides AA and Liu C: Spatial links between subchondral bone architectural features and cartilage degeneration in osteoarthritic joints. Sci Rep. 12(6694)2022.PubMed/NCBI View Article : Google Scholar

71 

de Rooij M, van der Leeden M, Heymans MW, Holla JF, Häkkinen A, Lems WF, Roorda LD, Veenhof C, Sanchez-Ramirez DC, de Vet HC and Dekker J: Prognosis of pain and physical functioning in patients with knee osteoarthritis: A systematic review and meta-analysis. Arthritis Care Res (Hoboken). 68:481–492. 2016.PubMed/NCBI View Article : Google Scholar

72 

Emmert D, Rasche T, Stieber C, Conrad R and Mücke M: Knee pain-symptoms, diagnosis and therapy of osteoarthritis. MMW Fortschr Med. 160:58–64. 2018.PubMed/NCBI View Article : Google Scholar : (In German).

73 

Sinusas K: Osteoarthritis: Diagnosis and treatment. Am Fam Physician. 85:49–56. 2012.PubMed/NCBI

74 

Kolasinski SL, Neogi T, Hochberg MC, Oatis C, Guyatt G, Block J, Callahan L, Copenhaver C, Dodge C, Felson D, et al: 2019 American college of Rheumatology/Arthritis foundation guideline for the management of osteoarthritis of the hand, hip, and knee. Arthritis Care Res (Hoboken). 72:149–162. 2020.PubMed/NCBI View Article : Google Scholar

75 

Pessler F, Dai L, Diaz-Torne C, Gomez-Vaquero C, Paessler ME, Zheng DH, Einhorn E, Range U, Scanzello C and Schumacher HR: The synovitis of ‘Non-Inflammatory’ orthopaedic arthropathies: A quantitative histological and immunohistochemical analysis. Ann Rheum Dis. 67:1184–1187. 2008.PubMed/NCBI View Article : Google Scholar

76 

Katsuragawa Y, Saitoh K, Tanaka N, Wake M, Ikeda Y, Furukawa H, Tohma S, Sawabe M, Ishiyama M, Yagishita S, et al: Changes of human menisci in osteoarthritic knee joints. Osteoarthritis Cartilage. 18:1133–1143. 2010.PubMed/NCBI View Article : Google Scholar

77 

Krackow KA, Mandeville DS, Rachala SR, Bayers-Thering M and Osternig LR: Torsion deformity and joint loading for medial knee osteoarthritis. Gait Posture. 33:625–629. 2011.PubMed/NCBI View Article : Google Scholar

78 

Incze MA: I have arthritis of the knees: What Should I Do? JAMA Intern Med. 179(736)2019.PubMed/NCBI View Article : Google Scholar

79 

Huang J, Chen X, Xia M, Lv S and Tong P: West Lake staging: A new staging system orchestrated by X-ray and MRI on knee osteoarthritis. J Orthop Surg (Hong Kong). 29(23094990211049587)2021.PubMed/NCBI View Article : Google Scholar

80 

Sukerkar PA and Doyle Z: Imaging of osteoarthritis of the knee. Radiol Clin North Am. 60:605–616. 2022.PubMed/NCBI View Article : Google Scholar

81 

Zarringam D, Saris DBF and Bekkers JEJ: The value of SPECT/CT for knee osteoarthritis: A systematic review. Cartilage. 12:431–437. 2021.PubMed/NCBI View Article : Google Scholar

82 

Bousson V, Lowitz T, Laouisset L, Engelke K and Laredo JD: CT imaging for the investigation of subchondral bone in knee osteoarthritis. Osteoporos Int. 23 (Suppl 8):S861–S865. 2012.PubMed/NCBI View Article : Google Scholar

83 

Watson PJ, Carpenter TA, Hall LD and Tyler JA: Cartilage swelling and loss in a spontaneous model of osteoarthritis visualized by magnetic resonance imaging. Osteoarthritis Cartilage. 4:197–207. 1996.PubMed/NCBI View Article : Google Scholar

84 

Hayashi D, Guermazi A, Kwoh CK, Hannon MJ, Moore C, Jakicic JM, Green SM and Roemer FW: Semiquantitative assessment of subchondral bone marrow edema-like lesions and subchondral cysts of the knee at 3T MRI: A comparison between intermediate-weighted fat-suppressed spin echo and Dual Echo Steady State sequences. BMC Musculoskelet Disord. 12(198)2011.PubMed/NCBI View Article : Google Scholar

85 

Blumenkrantz G and Majumdar S: Quantitative magnetic resonance imaging of articular cartilage in osteoarthritis. Eur Cell Mater. 13:76–86. 2007.PubMed/NCBI View Article : Google Scholar

86 

Mosher TJ and Dardzinski BJ: Cartilage MRI T2 relaxation time mapping: Overview and applications. Semin Musculoskelet Radiol. 8:355–368. 2004.PubMed/NCBI View Article : Google Scholar

87 

Nissi MJ, Rieppo J, Töyräs J, Laasanen MS, Kiviranta I, Jurvelin JS and Nieminen MT: T(2) relaxation time mapping reveals age- and species-related diversity of collagen network architecture in articular cartilage. Osteoarthritis Cartilage. 14:1265–1271. 2006.PubMed/NCBI View Article : Google Scholar

88 

Dardzinski BJ, Mosher TJ, Li S, Van Slyke MA and Smith MB: Spatial variation of T2 in human articular cartilage. Radiology. 205:546–550. 1997.PubMed/NCBI View Article : Google Scholar

89 

Liebl H, Joseph G, Nevitt MC, Singh N, Heilmeier U, Subburaj K, Jungmann PM, McCulloch CE, Lynch JA, Lane NE and Link TM: Early T2 changes predict onset of radiographic knee osteoarthritis: Data from the osteoarthritis initiative. Ann Rheum Dis. 74:1353–1359. 2015.PubMed/NCBI View Article : Google Scholar

90 

Conaghan P: Is MRI useful in osteoarthritis? Best Pract Res Clin Rheumatol. 20:57–68. 2006.PubMed/NCBI View Article : Google Scholar

91 

Zhao X, Gong W, Li X, Yang W, Yang D and Liu Z: Back propagation neural network-based ultrasound image for diagnosis of cartilage lesions in knee osteoarthritis. J Healthc Eng. 2021(2584291)2021.PubMed/NCBI View Article : Google Scholar

92 

McCrae F, Shouls J, Dieppe P and Watt I: Scintigraphic assessment of osteoarthritis of the knee joint. Ann Rheum Dis. 51:938–942. 1992.PubMed/NCBI View Article : Google Scholar

93 

Garnero P, Mazières B, Guéguen A, Abbal M, Berdah L, Lequesne M, Nguyen M, Salles JP, Vignon E and Dougados M: Cross-sectional association of 10 molecular markers of bone, cartilage, and synovium with disease activity and radiological joint damage in patients with hip osteoarthritis: The ECHODIAH cohort. J Rheumatol. 32:697–703. 2005.PubMed/NCBI

94 

Schouten JS, Van den Ouweland FA, Valkenburg HA and Lamberts SW: Insulin-like growth factor-1: A prognostic factor of knee osteoarthritis. Br J Rheumatol. 32:274–280. 1993.PubMed/NCBI View Article : Google Scholar

95 

Manicourt DH, Fujimoto N, Obata K and Thonar EJ: Serum levels of collagenase, stromelysin-1, and TIMP-1. Age- and sex-related differences in normal subjects and relationship to the extent of joint involvement and serum levels of antigenic keratan sulfate in patients with osteoarthritis. Arthritis Rheum. 37:1774–1783. 1994.PubMed/NCBI View Article : Google Scholar

96 

Zhang W, Doherty M, Arden N, Bannwarth B, Bijlsma J, Gunther KP, Hauselmann HJ, Herrero-Beaumont G, Jordan K, Kaklamanis P, et al: EULAR evidence based recommendations for the management of hip osteoarthritis: Report of a task force of the EULAR Standing Committee for International Clinical Studies Including Therapeutics (ESCISIT). Ann Rheum Dis. 64:669–681. 2005.PubMed/NCBI View Article : Google Scholar

97 

Zeng CY, Zhang ZR, Tang ZM and Hua FZ: Benefits and mechanisms of exercise training for knee osteoarthritis. Front Physiol. 12(794062)2021.PubMed/NCBI View Article : Google Scholar

98 

Wellsandt E and Golightly Y: Exercise in the management of knee and hip osteoarthritis. Curr Opin Rheumatol. 30:151–159. 2018.PubMed/NCBI View Article : Google Scholar

99 

Li Y, Su Y, Chen S, Zhang Y, Zhang Z, Liu C, Lu M, Liu F, Li S, He Z, et al: The effects of resistance exercise in patients with knee osteoarthritis: A systematic review and meta-analysis. Clin Rehabil. 30:947–959. 2016.PubMed/NCBI View Article : Google Scholar

100 

Page CJ, Hinman RS and Bennell KL: Physiotherapy management of knee osteoarthritis. Int J Rheum Dis. 14:145–151. 2011.PubMed/NCBI View Article : Google Scholar

101 

van Doormaal MCM, Meerhoff GA, Vliet Vlieland TPM and Peter WF: A clinical practice guideline for physical therapy in patients with hip or knee osteoarthritis. Musculoskeletal Care. 18:575–595. 2020.PubMed/NCBI View Article : Google Scholar

102 

Peter WF, Jansen MJ, Hurkmans EJ, Bloo H, Dekker J, Dilling RG, Hilberdink W, Kersten-Smit C, de Rooij M, Veenhof C, et al: Physiotherapy in hip and knee osteoarthritis: Development of a practice guideline concerning initial assessment, treatment and evaluation. Acta Reumatol Port. 36:268–281. 2011.PubMed/NCBI

103 

Kloppenburg M, Kroon FP, Blanco FJ, Doherty M, Dziedzic KS, Greibrokk E, Haugen IK, Herrero-Beaumont G, Jonsson H, Kjeken I, et al: 2018 update of the EULAR recommendations for the management of hand osteoarthritis. Ann Rheum Dis. 78:16–24. 2019.PubMed/NCBI View Article : Google Scholar

104 

Rausch Osthoff AK, Niedermann K, Braun J, Adams J, Brodin N, Dagfinrud H, Duruoz T, Esbensen BA, Günther KP, Hurkmans E, et al: 2018 EULAR recommendations for physical activity in people with inflammatory arthritis and osteoarthritis. Ann Rheum Dis. 77:1251–1260. 2018.PubMed/NCBI View Article : Google Scholar

105 

Kroon FPB, Carmona L, Schoones JW and Kloppenburg M: Efficacy and safety of non-pharmacological, pharmacological and surgical treatment for hand osteoarthritis: A systematic literature review informing the 2018 update of the EULAR recommendations for the management of hand osteoarthritis. RMD Open. 4(e000734)2018.PubMed/NCBI View Article : Google Scholar

106 

Wang HM, Liu JN and Zhao Y: Progress on integrated Chinese and Western medicine in the treatment of osteoarthritis. Chin J Integr Med. 16:378–384. 2010.PubMed/NCBI View Article : Google Scholar

107 

Xu X, Wan Y, Gong L, Ma Z and Xu T: Chinese herbal medicine Yanghe decoction for knee osteoarthritis: A protocol for systematic review and meta-analysis. Medicine (Baltimore). 99(e21877)2020.PubMed/NCBI View Article : Google Scholar

108 

Xia H, Cao D and Yang F, Yang W, Li W, Liu P, Wang S and Yang F: Jiawei Yanghe decoction ameliorates cartilage degradation in vitro and vivo via Wnt/β-catenin signaling pathway. Biomed Pharmacother. 122(109708)2020.PubMed/NCBI View Article : Google Scholar

109 

Hochberg MC and Dougados M: Pharmacological therapy of osteoarthritis. Best Pract Res Clin Rheumatol. 15:583–593. 2001.PubMed/NCBI View Article : Google Scholar

110 

Nelson AE, Allen KD, Golightly YM, Goode AP and Jordan JM: A systematic review of recommendations and guidelines for the management of osteoarthritis: The chronic osteoarthritis management initiative of the U.S. bone and joint initiative. Semin Arthritis Rheum. 43:701–712. 2014.PubMed/NCBI View Article : Google Scholar

111 

Bindu S, Mazumder S and Bandyopadhyay U: Non-steroidal anti-inflammatory drugs (NSAIDs) and organ damage: A current perspective. Biochem Pharmacol. 180(114147)2020.PubMed/NCBI View Article : Google Scholar

112 

Bacchi S, Palumbo P, Sponta A and Coppolino MF: Clinical pharmacology of non-steroidal anti-inflammatory drugs: A review. Antiinflamm Antiallergy Agents Med Chem. 11:52–64. 2012.PubMed/NCBI View Article : Google Scholar

113 

Jones R: Nonsteroidal anti-inflammatory drug prescribing: Past, present, and future. Am J Med. 110 (Suppl):4S–7S. 2001.PubMed/NCBI View Article : Google Scholar

114 

Puljak L, Marin A, Vrdoljak D, Markotic F, Utrobicic A and Tugwell P: Celecoxib for osteoarthritis. Cochrane Database Syst Rev. 5(CD009865)2017.PubMed/NCBI View Article : Google Scholar

115 

Mastbergen SC, Bijlsma JW and Lafeber FP: Selective COX-2 inhibition is favorable to human early and late-stage osteoarthritic cartilage: A human in vitro study. Osteoarthritis Cartilage. 13:519–526. 2005.PubMed/NCBI View Article : Google Scholar

116 

Solomon SD, McMurray JJ, Pfeffer MA, Wittes J, Fowler R, Finn P, Anderson WF, Zauber A, Hawk E and Bertagnolli M: Adenoma Prevention with Celecoxib (APC) Study Investigators. Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N Engl J Med. 352:1071–1080. 2005.PubMed/NCBI View Article : Google Scholar

117 

Elsawy NA, Ibrahiem AH, Younis GA, Meheissen MA and Abdel-Fattah YH: Clinical examination, ultrasound assessment and aspiration of knee effusion in primary knee osteoarthritis patients. J Orthop Surg Res. 18(422)2023.PubMed/NCBI View Article : Google Scholar

118 

Henricsdotter C, Ellegaard K, Klokker L, Bartholdy C, Bandak E, Bartels EM, Bliddal H and Henriksen M: Changes in ultrasound assessed markers of inflammation following intra-articular steroid injection combined with exercise in knee osteoarthritis: Exploratory outcome from a randomized trial. Osteoarthritis Cartilage. 24:814–821. 2016.PubMed/NCBI View Article : Google Scholar

119 

Wang SZ, Guo YD, Zhang XJ and Wang C: Intra-articular injection of compound betamethasone and hyaluronic acid for the treatment of moderate-severe knee osteoarthritis: A randomized controlled trial. Zhongguo Gu Shang. 34:424–428. 2021.PubMed/NCBI View Article : Google Scholar : (In Chinese).

120 

Deyle GD, Allen CS, Allison SC, Gill NW, Hando BR, Petersen EJ, Dusenberry DI and Rhon DI: Physical therapy versus glucocorticoid injection for osteoarthritis of the knee. N Engl J Med. 382:1420–1429. 2020.PubMed/NCBI View Article : Google Scholar

121 

Zhang Y, Ruan G, Zheng P, Huang S, Zhou X, Liu X, Hu W, Feng H, Lin Y, He J, et al: Efficacy and safety of GLucocorticoid injections into InfrapaTellar faT pad in patients with knee ostEoarthRitiS: Protocol for the GLITTERS randomized controlled trial. Trials. 24(6)2023.PubMed/NCBI View Article : Google Scholar

122 

Jin L, Xu K, Liang Y, Du P, Wan S and Jiang C: Effect of hyaluronic acid on cytokines and immune cells change in patients of knee osteoarthritis. BMC Musculoskelet Disord. 23(812)2022.PubMed/NCBI View Article : Google Scholar

123 

Georgiev T and Angelov AK: Modifiable risk factors in knee osteoarthritis: Treatment implications. Rheumatol Int. 39:1145–1157. 2019.PubMed/NCBI View Article : Google Scholar

124 

Liao CD, Chen HC, Huang MH, Liou TH, Lin CL and Huang SW: Comparative efficacy of intra-articular injection, physical therapy, and combined treatments on pain, function, and sarcopenia indices in knee osteoarthritis: A network meta-analysis of randomized controlled trials. Int J Mol Sci. 24(6078)2023.PubMed/NCBI View Article : Google Scholar

125 

Xie X, Zhang C and Tuan RS: Biology of platelet-rich plasma and its clinical application in cartilage repair. Arthritis Res Ther. 16(204)2014.PubMed/NCBI View Article : Google Scholar

126 

Duymus TM, Mutlu S, Dernek B, Komur B, Aydogmus S and Kesiktas FN: Choice of intra-articular injection in treatment of knee osteoarthritis: Platelet-rich plasma, hyaluronic acid or ozone options. Knee Surg Sports Traumatol Arthrosc. 25:485–492. 2017.PubMed/NCBI View Article : Google Scholar

127 

Taqi A, Gran S and Knaggs RD: Current use of analgesics and the risk of falls in people with knee osteoarthritis: A population-based cohort study using primary care and hospital records. Osteoarthr Cartil Open. 3(100165)2021.PubMed/NCBI View Article : Google Scholar

128 

Sharma SK, Vij AS and Sharma M: Mechanisms and clinical uses of capsaicin. Eur J Pharmacol. 720:55–62. 2013.PubMed/NCBI View Article : Google Scholar

129 

Liu J and Zang YJ: Comparative study between three analgesic agents for the pain management during extracorporeal shock wave lithotripsy. Urol J. 10:942–945. 2013.PubMed/NCBI

130 

Guo J, Hu X, Wang J, Yu B, Li J, Chen J, Nie X, Zheng Z, Wang S and Qin Q: Safety and efficacy of compound methyl salicylate liniment for topical pain: A multicenter real-world study in China. Front Pharmacol. 13(1015941)2022.PubMed/NCBI View Article : Google Scholar

131 

Riddle DL, Moxley G and Dumenci L: Response to comments in: Statin use is associated with reduced incidence and progression of knee osteoarthritis in the Rotterdam study by Clockaerts et al. Ann Rheum Dis. 72(e12)2013.PubMed/NCBI View Article : Google Scholar

132 

Sondergaard BC, Madsen SH, Segovia-Silvestre T, Paulsen SJ, Christiansen T, Pedersen C, Bay-Jensen AC and Karsdal MA: Investigation of the direct effects of salmon calcitonin on human osteoarthritic chondrocytes. BMC Musculoskelet Disord. 11(62)2010.PubMed/NCBI View Article : Google Scholar

133 

Armagan O, Serin DK, Calisir C, Dokumacioglu A, Ozgen M, Oner S and Alatas O: Inhalation therapy of calcitonin relieves osteoarthritis of the knee. J Korean Med Sci. 27:1405–1410. 2012.PubMed/NCBI View Article : Google Scholar

134 

Tanaka H, Ueta Y, Yamashita U, Kannan H and Yamashita H: Biphasic changes in behavioral, endocrine, and sympathetic systems in adjuvant arthritis in Lewis rats. Brain Res Bull. 39:33–37. 1996.PubMed/NCBI View Article : Google Scholar

135 

Sawitzke AD, Shi H, Finco MF, Dunlop DD, Harris CL, Singer NG, Bradley JD, Silver D, Jackson CG, Lane NE, et al: Clinical efficacy and safety of glucosamine, chondroitin sulphate, their combination, celecoxib or placebo taken to treat osteoarthritis of the knee: 2-year results from GAIT. Ann Rheum Dis. 69:1459–1464. 2010.PubMed/NCBI View Article : Google Scholar

136 

Jordan KM, Arden NK, Doherty M, Bannwarth B, Bijlsma JW, Dieppe P, Gunther K, Hauselmann H, Herrero-Beaumont G, Kaklamanis P, et al: EULAR Recommendations 2003: An evidence based approach to the management of knee osteoarthritis: Report of a Task Force of the Standing Committee for International Clinical Studies Including Therapeutic Trials (ESCISIT). Ann Rheum Dis. 62:1145–1155. 2003.PubMed/NCBI View Article : Google Scholar

137 

Bishnoi M, Jain A, Hurkat P and Jain SK: Chondroitin sulphate: A focus on osteoarthritis. Glycoconj J. 33:693–705. 2016.PubMed/NCBI View Article : Google Scholar

138 

Fidelix TS, Macedo CR, Maxwell LJ and Fernandes Moça Trevisani V: Diacerein for osteoarthritis. Cochrane Database Syst Rev: CD005117, 2014.

139 

Reginster JY, Deroisy R, Rovati LC, Lee RL, Lejeune E, Bruyere O, Giacovelli G, Henrotin Y, Dacre JE and Gossett C: Long-term effects of glucosamine sulphate on osteoarthritis progression: A randomised, placebo-controlled clinical trial. Lancet. 357:251–256. 2001.PubMed/NCBI View Article : Google Scholar

140 

Loeser RF, Goldring SR, Scanzello CR and Goldring MB: Osteoarthritis: A disease of the joint as an organ. Arthritis Rheum. 64:1697–1707. 2012.PubMed/NCBI View Article : Google Scholar

141 

Sadowski T and Steinmeyer J: Effects of tetracyclines on the production of matrix metalloproteinases and plasminogen activators as well as of their natural inhibitors, tissue inhibitor of metalloproteinases-1 and plasminogen activator inhibitor-1. Inflamm Res. 50:175–182. 2001.PubMed/NCBI View Article : Google Scholar

142 

Malemud CJ: Inhibition of MMPs and ADAM/ADAMTS. Biochem Pharmacol. 165:33–40. 2019.PubMed/NCBI View Article : Google Scholar

143 

Rosenbaum CC, O'Mathúna DP, Chavez M and Shields K: Antioxidants and antiinflammatory dietary supplements for osteoarthritis and rheumatoid arthritis. Altern Ther Health Med. 16:32–40. 2010.PubMed/NCBI

144 

Gui T, Luo L, Chhay B, Zhong L, Wei Y, Yao L, Yu W, Li J, Nelson CL, Tsourkas A, et al: Superoxide dismutase-loaded porous polymersomes as highly efficient antioxidant nanoparticles targeting synovium for osteoarthritis therapy. Biomaterials. 283(121437)2022.PubMed/NCBI View Article : Google Scholar

145 

Mathieu S, Soubrier M, Peirs C, Monfoulet LE, Boirie Y and Tournadre A: A Meta-analysis of the impact of nutritional supplementation on osteoarthritis symptoms. Nutrients. 14(1607)2022.PubMed/NCBI View Article : Google Scholar

146 

Yao H, Xu J, Wang J, Zhang Y, Zheng N, Yue J, Mi J, Zheng L, Dai B, Huang W, et al: Combination of magnesium ions and vitamin C alleviates synovitis and osteophyte formation in osteoarthritis of mice. Bioact Mater. 6:1341–1352. 2021.PubMed/NCBI View Article : Google Scholar

147 

Suantawee T, Tantavisut S, Adisakwattana S, Tanavalee A, Yuktanandana P, Anomasiri W, Deepaisarnsakul B and Honsawek S: Oxidative stress, vitamin e, and antioxidant capacity in knee osteoarthritis. J Clin Diagn Res. 7:1855–1859. 2013.PubMed/NCBI View Article : Google Scholar

148 

Bhattacharya I, Saxena R and Gupta V: Efficacy of vitamin E in knee osteoarthritis management of North Indian geriatric population. Ther Adv Musculoskelet Dis. 4:11–19. 2012.PubMed/NCBI View Article : Google Scholar

149 

Karsdal MA, Tanko LB, Riis BJ, Sondergard BC, Henriksen K, Altman RD, Qvist P and Christiansen C: Calcitonin is involved in cartilage homeostasis: Is calcitonin a treatment for OA? Osteoarthritis Cartilage. 14:617–624. 2006.PubMed/NCBI View Article : Google Scholar

150 

Saberianpour S, Abolbashari S, Modaghegh MHS, Karimian MS, Eid AH, Sathyapalan T and Sahebkar A: Therapeutic effects of statins on osteoarthritis: A review. J Cell Biochem. 123:1285–1297. 2022.PubMed/NCBI View Article : Google Scholar

151 

Cook JL and Payne JT: Surgical treatment of osteoarthritis. Vet Clin North Am Small Anim Pract. 27:931–944. 1997.PubMed/NCBI View Article : Google Scholar

152 

Gomoll AH, Filardo G, de Girolamo L, Espregueira-Mendes J, Marcacci M, Rodkey WG, Steadman JR, Zaffagnini S and Kon E: Surgical treatment for early osteoarthritis. Part I: Cartilage repair procedures. Knee Surg Sports Traumatol Arthrosc. 20:450–466. 2012.PubMed/NCBI View Article : Google Scholar

153 

Krych AJ, Bert JM and Levy BA: Treatment of OA of the knee in the middle-aged athlete: The role of arthroscopy. Sports Med Arthrosc Rev. 21:23–30. 2013.PubMed/NCBI View Article : Google Scholar

154 

Gobbi A, Lane JG and Dallo I: Editorial commentary: Cartilage restoration-what is currently available? Arthroscopy. 36:1625–1628. 2020.PubMed/NCBI View Article : Google Scholar

155 

Yasunaga Y, Fujii J, Tanaka R, Yasuhara S, Yamasaki T, Adachi N and Ochi M: Rotational acetabular osteotomy. Clin Orthop Surg. 9:129–135. 2017.PubMed/NCBI View Article : Google Scholar

156 

Li OL, Pritchett S, Giffin JR and Spouge ARI: High Tibial osteotomy: An update for radiologists. AJR Am J Roentgenol. 218:701–712. 2022.PubMed/NCBI View Article : Google Scholar

157 

Han J, Zeng Z, Pei F and Zheng T: An implementation study of periarticular knee osteotomy in the treatment of knee osteoarthritis. Am J Transl Res. 13:4771–4779. 2021.PubMed/NCBI

158 

Liao CD, Huang SW, Huang YY and Lin CL: Effects of Sarcopenic obesity and its confounders on knee range of motion outcome after total knee replacement in older adults with knee osteoarthritis: A retrospective study. Nutrients. 13(3717)2021.PubMed/NCBI View Article : Google Scholar

159 

Liu CY, Li CD, Wang L, Ren S, Yu FB, Li JG, Ma JX and Ma XL: Function scores of different surgeries in the treatment of knee osteoarthritis: A PRISMA-compliant systematic review and network-meta analysis. Medicine (Baltimore). 97(e10828)2018.PubMed/NCBI View Article : Google Scholar

160 

Xue YY, Shi JN, Zhang K, Zhang HH and Yan SH: The effects of total knee arthroplasty on knee proprioception of patients with knee osteoarthritis: A meta-analysis. J Orthop Surg Res. 17(258)2022.PubMed/NCBI View Article : Google Scholar

161 

He M, Zhong X, Li Z, Shen K and Zeng W: Progress in the treatment of knee osteoarthritis with high Tibial osteotomy: A systematic review. Syst Rev. 10(56)2021.PubMed/NCBI View Article : Google Scholar

162 

Dell'Isola A, Allan R, Smith SL, Marreiros SS and Steultjens M: Identification of clinical phenotypes in knee osteoarthritis: A systematic review of the literature. BMC Musculoskelet Disord. 17(425)2016.PubMed/NCBI View Article : Google Scholar

163 

Lin M, Li X, Liang W, Liu J, Guo J, Zheng J and Liu X: Needle-knife therapy improves the clinical symptoms of knee osteoarthritis by inhibiting the expression of inflammatory cytokines. Exp Ther Med. 7:835–842. 2014.PubMed/NCBI View Article : Google Scholar

164 

Du X, Liu ZY, Tao XX, Mei YL, Zhou DQ, Cheng K, Gao SL, Shi HY, Song C and Zhang XM: Research progress on the pathogenesis of knee osteoarthritis. Orthop Surg: Jul 12, 2023 doi: 10.1111/os.13809 (Epub ahead of print).

165 

Dai W, Cheng J, Yan W, Cao C, Zhao F, Li Q, Hu X, Wang J and Ao Y: Enhanced osteochondral repair with hyaline cartilage formation using an extracellular matrix-inspired natural scaffold. Sci Bull (Beijing): Aug 1, 2023 doi: 10.1016/j.scib.2023.07.050 (Epub ahead of print).

166 

Li C, Liu Y, Weng T, Yang M, Wang X and Chai W: Fabrication of injectable Kartogenin-conjugated composite hydrogel with a sustained drug release for cartilage repair. Pharmaceutics. 15(1949)2023.PubMed/NCBI View Article : Google Scholar

167 

Deng C, Chen Y, Zhao X, Yu L, Xiao Y, Li H, Zhang Y, Ai K, Zhou D, Bai X, et al: Apoptotic neutrophil membrane-camouflaged liposomes for dually targeting synovial macrophages and fibroblasts to attenuate osteoarthritis. ACS Appl Mater Interfaces: Jul 31, 2023 doi: 10.1021/acsami.3c05861 (Epub ahead of print).

168 

Liu R, Liu Z, Chen H, He S, Wang S, Dai J and Li X: Ginkgolide K delays the progression of osteoarthritis by regulating YAP to promote the formation of cartilage extracellular matrix. Phytother Res: Aug 1, 2023 doi: 10.1002/ptr.7953 (Epub ahead of print).

169 

Kuppa SS, Kim HK, Kang JY, Lee SC, Yang HY, Sankaranarayanan J and Seon JK: Polynucleotides suppress inflammation and stimulate matrix synthesis in an in vitro Cell-based osteoarthritis model. Int J Mol Sci. 24(12282)2023.PubMed/NCBI View Article : Google Scholar

170 

Chen S, Meng C, He Y, Xu H, Qu Y, Wang Y, Fan Y, Huang X and You H: An in vitro and in vivo study: Valencene protects cartilage and alleviates the progression of osteoarthritis by anti-oxidative stress and anti-inflammatory effects. Int Immunopharmacol. 123(110726)2023.PubMed/NCBI View Article : Google Scholar

171 

Wang X, Cai Y, Wu C, Liang J, Tang K, Lin Z, Chen L, Lu Y and Wang Q: Conversion of senescent cartilage into a pro-chondrogenic microenvironment with antibody-functionalized copper sulfate nanoparticles for efficient osteoarthritis therapy. J Nanobiotechnology. 21(258)2023.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

October-2023
Volume 26 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Geng R, Li J, Yu C, Zhang C, Chen F, Chen J, Ni H, Wang J, Kang K, Wei Z, Wei Z, et al: Knee osteoarthritis: Current status and research progress in treatment (Review). Exp Ther Med 26: 481, 2023
APA
Geng, R., Li, J., Yu, C., Zhang, C., Chen, F., Chen, J. ... Jin, T. (2023). Knee osteoarthritis: Current status and research progress in treatment (Review). Experimental and Therapeutic Medicine, 26, 481. https://doi.org/10.3892/etm.2023.12180
MLA
Geng, R., Li, J., Yu, C., Zhang, C., Chen, F., Chen, J., Ni, H., Wang, J., Kang, K., Wei, Z., Xu, Y., Jin, T."Knee osteoarthritis: Current status and research progress in treatment (Review)". Experimental and Therapeutic Medicine 26.4 (2023): 481.
Chicago
Geng, R., Li, J., Yu, C., Zhang, C., Chen, F., Chen, J., Ni, H., Wang, J., Kang, K., Wei, Z., Xu, Y., Jin, T."Knee osteoarthritis: Current status and research progress in treatment (Review)". Experimental and Therapeutic Medicine 26, no. 4 (2023): 481. https://doi.org/10.3892/etm.2023.12180