Open Access

Role of macrophage polarization in osteoarthritis (Review)

  • Authors:
    • Baisen Chen
    • Hongxiang Hong
    • Yuyu Sun
    • Chu Chen
    • Chunshuai Wu
    • Guanhua Xu
    • Guofeng Bao
    • Zhiming Cui
  • View Affiliations

  • Published online on: November 9, 2022     https://doi.org/10.3892/etm.2022.11693
  • Article Number: 757
  • Copyright: © Chen 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

Osteoarthritis (OA) is a disease involving the whole joint that seriously reduces the living standards of individuals. Traditional treatments include physical therapy, administration of anti‑inflammatory and analgesic drugs and injection of glucocorticoids or hyaluronic acid into the joints. However, these methods have limited efficacy and it is difficult to reverse the progression of OA, therefore it is urgent to find new effective treatment methods. Immune microenvironment is significant in the occurrence and development of OA. Recent studies have shown that macrophages are important targets for the treatment of OA. Macrophages are polarized into M1 pro‑inflammatory phenotype and M2 anti‑inflammatory phenotype under stimulation of different factors, which release and regulate inflammatory response and cartilage growth. Accumulating studies have tried to alleviate OA by regulating macrophage homeostasis. The present study summarized the related studies, discuss the mechanism of various therapeutic reagents on OA, expound the molecular mechanism of drug effect on OA and attempted to provide clues for the treatment of OA.

1. Introduction

Osteoarthritis (OA) is a joint disease with a high prevalence and serious consequences. It leads to pain and decline in the quality of life of affected individuals (1,2). OA is characterized by synovitis, osteophyte formation and degeneration of joint-related tissues (such as articular cartilage), which may eventually result in changes in bone structure and disability of patients (3). Traditional treatments include physical therapy, anti-inflammatory and analgesic drugs and injecting glucocorticoid or hyaluronic acid into the joint (4-6). However, at present, most OA therapies have limited efficacy in symptom relief. There are still no drugs that can reverse the pathological process of OA (7). Although there is continuing research to reveal the mechanism of OA, more effective treatments are required. For the present review, the following terms were searched using the PubMed database (https://pubmed.ncbi.nlm.nih.gov/): ‘osteoarthritis’, ‘macrophage polarization’, ‘treatment’ and ‘cartilage damage’; only papers published in English were assessed.

In OA, immune cells secrete inflammatory factors or mediators affecting cartilage structure through intercellular association to participate in the pathological process. Neutrophils promote OA by secreting proinflammatory mediators and producing enzymes that degrade cartilage. Dendritic cells activate T helper (Th)1 and Th17 cells, which produce pro-inflammatory cytokines, resulting in cartilage degradation, while Th2 cells secrete anti-inflammatory factors to protect cartilage. In addition, Th2 cells can activate B cells to secrete proinflammatory factors and antibodies to inhibit cartilage repair. Treg cells have beneficial effects on OA. They do not directly participate in cartilage repair, but act on neutrophils to secrete anti-inflammatory factors. Natural killer cells are closely related to the differentiation of mesenchymal stem cells (MSCs) and osteoclasts. However, the largest number of immune cell types in OA synovium is macrophages (8). Macrophages, as an important part of the immune system, have long been considered to be an crucial participant in OA (9).

Macrophages, derived from hematopoietic stem cells, are immune cells that serve a key role in the immune response (10). Macrophages are activated by environmental factors such as TNF-α, IFN-γ and lipopolysaccharide (LPS) (8). When activated, they can absorb innate antigens or invading pathogens, express costimulatory molecules and secrete more inflammatory cytokines, which are essential for immune response and cartilage reconstruction (11). Macrophages are mainly polarized into M1 and M2 phenotypes (8). After being stimulated by IFN-γ, M0 macrophages are polarized into M1 macrophages, thereby expressing pro-inflammatory cytokines and participating in the process of pro-inflammation and degradation of extracellular matrix (ECM) (9). When M0 macrophages are stimulated by IL-4/IL-13, they polarized into M2 macrophages and express anti-inflammatory cytokines and thus exert anti-inflammatory and chondrogenic effects (12). Macrophage polarization participates in a number of pathological processes, including metabolic changes, virus infection, inflammatory environment and tumor immunity (8,9,11). Recent research has shown that it is beneficial to slow the progress of OA by regulating the changes of macrophage phenotype (12). This suggests that macrophage polarization is an emerging target to treat OA. The present study collected studies on the regulation of macrophage polarization and OA progress in order to elaborate its mechanism and provide clues for the subsequent development of treatment methods based on the regulation of macrophage phenotype.

2. Development of OA

OA may be caused by factors such as heredity and mechanical load (13-15). OA can influence the entire joint, including cartilage, synovial tissue, subchondral bone, joint capsule, ligaments and periarticular muscles (16). However, cartilage degeneration may be the most important pathological process of OA and accumulating researches focus on preventing its degeneration (3). The inflammatory environment generated by cartilage injury will cause chondrocyte apoptosis and hypertrophy, ECM decomposition, eventually leading to cartilage injury and aggravating OA (17). Cartilage covers the end of bone and is a type of hyaline cartilage without nerves and blood vessels (18). Typically, it consists of ECM and chondrocytes (19). The ECM consists of matrix, collagen type 1A (col1A) and collagen type 2A1 (col12A1) (20). Proteoglycan is the main component of matrix and participates in chondrocyte synthesis and degradation (12). The cascade effect is amplified by the production of MMP or a disintegrin-like and metalloproteinase with thrombospondin motifs (ADAMTS) by chondrocytes, synovium fibroblasts and macrophages when mechanical loading on cartilage causes damage (8). The ECM degradation enzyme ADAMTS5 is a proteoglycan enzyme of the ADAMTS family (21), whose main function is to clear large proteoglycans such as aggrecan, leading to the destruction of cartilage and OA (22). MMPs, especially collagenase MMP1 and MMP13, are believed to cut collagen type II, the main structural component of cartilage, leading to irreversible loss of ECM structure and function (23). Therefore, the regulation of cartilage homeostasis is particularly important for the development of OA.

Existing studies have shown that the imbalance of ECM composition synthesis and degradation is an important marker of exacerbating OA (8,12,23,24). However, the mechanism of imbalance between cartilage damage and repair remains to be elucidated. Trauma can lead to bone and joint friction or increased enzyme activity, followed by the formation of ‘wear’ particles that are then eaten by resident macrophages (8). However, once the immune system is unable to eliminate these ‘wear’ particles completely, they act as mediators of inflammation, inducing chondrocytes to release degrading enzymes (22). Synovial macrophages phagocytose molecules produced by collagen or proteoglycan breakdown and release pro-inflammatory cytokines such as TNF-α, IL-1 and IL-6(23). These cytokines can act on chondrocytes to further release degradation enzymes and prevent the generation of type II collagen, thus further increasing cartilage degradation (25). In conclusion, the mechanism of cartilage destruction aggravating OA may be: i) The increased production of degrading enzymes leads to decreased synthesis or increased decomposition of collagen fibers or ii) the apoptosis and senescence of chondrocytes. The morphologic change of articular cartilage goes through several processes: At first, fibrotic areas and cracks appear on the surface of the cartilage, leading to cartilaginous softening and cartilage thickness reduction (24). When the articular cartilage is completely destroyed, the underlying subchondral bone plate is completely exposed (25). The longer OA lasts, the more pronounced these changes become.

There are now a variety of induction methods to damage joints in animals and build models to simulate OA (26-28). One type of model is to simulate OA by surgical instability of joint structure and joint injury. These include anterior cruciate ligament (ACLT) transection or medial meniscus (DMM) instability, which may result in changes in the mechanical load of the joint (26). In addition, a closed non-invasive injury model has been established, which can simulate OA injury after mechanical injury regardless of whether there is ACL fracture or not (27). Another model triggers joint damage by injecting drugs into the joint that disrupt cartilage homeostasis. Intra-articular injection of monosodium iodoacetate (MIA) inhibits glycolysis of chondrocytes, which leads to chondrocyte death and induces inflammation, thus simulating OA (26). Occasionally collagenase is injected into the joint, which will destroy the stability of the joint and cause inflammation, leading to OA-like joint injury (26).

3. Macrophages and OA

OA is characterized by the infiltration and invasion of macrophages, the release of a great quantity of pro-inflammatory and pro-catabolic mediators in the articular cavity and the increase of synovial vascular distribution. These mediators are recognized by synovial immune cells and activate the immune cells to release more cytokines, resulting in chronic and repeated inflammation. In short, these fragments released into the synovial cavity stimulate macrophages to produce and release inflammatory mediators (such as cytokines, chemokines, lipid mediators and death-associated molecular patterns itself) into the synovial fluid (29). In turn, these mediators activate chondrocytes that produce metalloproteinases, leading to a vicious cycle between cartilage and synovium (30). The pathological process is generally described as mechanical stress directly damages cartilage or activates chondrocytes, producing abnormal levels of degradation enzymes and reactive oxygen species (ROS), leading to cartilage destruction. Subsequently, microcrystals, osteochondral fragments and ECM degradation products are released from the articular cavity. These molecules and products trigger inflammatory macrophages to secrete chemokines, cytokines, MMP, ROS and lipid mediators. They can directly degrade ECM components or imbalance of chondrocyte homeostasis, leading to imbalance of cartilage matrix degradation and synthesis, thereby aggravating OA.

Macrophages can regulate the local microenvironment under physiological and pathological conditions and respond quickly to various stimuli (31). After stimulation, macrophages are mainly activated into two phenotypes: M1 and M2. Macrophages is polarized to M1 type by IFN-γ released by Th1 cell and has high expression of proinflammatory cytokines, including TNF-α, IL-1β, IL-6, IL-12, IL-23 and cyclooxygenase-2 (COX-2), therefore, have strong pro-inflammatory and pro-tumor functions (32). M2 macrophages are polarized by the STAT pathway activated by Th2 cytokines such as IL4/IL-13 through interleukin receptor, which up regulates arginine-1(Arg-1), CC motif chemokine ligand (CCL) 17 and CCL2, so it has anti-inflammatory effect (33) (Fig. 1). Th1 cells release IFN-γ to polarize M0 macrophages into M1 macrophages, resulting in pro-inflammation and ECM degradation; Th2 cells release IL-4 and IL-13 polarizes M0 macrophages into M2 macrophages, resulting in anti-inflammation and chondrogenesis.

Therefore, macrophages are not only essential immune cells, but also attractive therapeutic targets. In-depth understanding of the molecular characteristics related to the dynamic changes of macrophage polarization and their pathways is crucial to elucidate the molecular basis of disease progression and design of new macrophage therapy strategies (34). Identifying precise targets that control the transition between given steady states is a huge challenge.

4. Macrophages are emerging targets for OA treatment

OA is an immune disease caused by dysfunction of immune microenvironment. Reconstructing the balance of immune microenvironment by regulating the polarization of macrophages is an effective measure to treat OA. The factors affecting OA development include chemical components and biomolecules. Increasing studies have shed light on regulating inflammation by reversing macrophage polarization and progress has been made. The following is a review of the regulation of macrophages on OA, providing clues for a deeper understanding of the regulation mechanism of macrophages in OA (Table I).

Table I

OA treatment strategies based on macrophage.

Table I

OA treatment strategies based on macrophage.

TypeNameOA sample; cellsOA sample; animal
Chemical compound   
 LiraglutideMouse primary chondrocytes, RAW 264.7 cellsMIA-induced OA mouse model
  Tert-butylhydroquinoneMouse chondrocytes and synovial macrophagesDMM-induced OA mouse model
 AngelicinMouse bone marrow macrophageDMM-induced OA mouse model
 FargesinMouse primary chondrocytes, RAW 264.7 cellsOA mouse model induced by injection of collagenase
 Pseudolaric acid BRAW 264.7 cells, bone marrow derived macrophages (BMDM)DMM-induced OA mouse model
 Eucommia ulmoides PolysaccharidesRAW 264.7 cellsAnterior cruciate ligament transection rabbit model
Cell   
 Human adipose stem cellsPrimary human adipose stem cells, RAW264.7 cellsMIA-induced OA mouse model
 Human umbilical cord mesenchymal stem cells3rd-5th generation human umbilical cord mesenchymal stem cells, CP-R092 cells, mouse synovial macrophagesAnterior cruciate ligament transection mouse model
 Human umbilical cord mesenchymal stem cellsPrimary human umbilical cord mesenchymal stem cells, mouse derived macrophages and rat derived chondrocytesAnterior cruciate ligament transection mouse model
 Transient receptor potential vanillin 1RAW 264.7 cellsMIA-induced OA rat model
Protein   
 Milk fat globule epidermal growth factor 8 8Mouse primary chondrocytes, RAW 264.7 cellsMIA-induced OA mouse model
 E3 ubiquitin ligase itch proteinMouse bone marrow macrophageMIA-induced OA mouse model

[i] OA, osteoarthritis; MIA, monosodium iodoacetate; DMM, destabilization of the medial meniscus.

Compounds. Liraglutide

Glucagon like peptide-1 (GLP-1) is secreted by intestinal L cells and is a gastrointestinal hormone that processes food intake. Insulin has a series of extrapancreatic functions associated with anti-inflammatory properties (35). The GLP-1 analogue liraglutide is a human GLP-1 applied in the patients with diabetes (36). New data suggest that liraglutide inhibits ROS generation, pro-inflammatory cytokine secretion (37).

In vitro, when liraglutide is added to LPS-stimulated RAW264.7 cells, M1 macrophage phenotype reverts to M2 phenotype (38). Liraglutide significantly reduced the expression of MMP and ADAMTS and made chondrocytes anti-catabolic. From a cellular perspective, the decrease of proinflammatory factors in macrophages and chondrocytes is dose-dependent (38). In vivo results show that in the MIA-induced model, liraglutide injection significantly reduced the secretion of IL-6, nitric oxide and prostaglandin E2 and improved the synovitis severity score in mice (38).

Liraglutide has been applied in clinical trials. A randomized controlled trial applied liraglutide in patients with knee arthritis, but it did not reduce knee pain compared with placebo (39). More clinical experiments are needed to verify the anti-inflammatory effect of liraglutide. In conclusion, liraglutide may improve OA by changing macrophage polarization and reducing cartilage degradation.

Tert-butylhydroquinone. Tert-butylhydroquinone (tBHQ) is a compound with strong antioxidant activity (40). There is increasing evidence that tBHQ may be used to treat diseases related to oxidative stress (41).

In vitro, tBHQ significantly reduces the expression of phosphorylated (p-)STAT1 and p-STAT3 induced by LPS and effectively inhibits the M1 polarization of synovial macrophages. From a signaling pathway perspective, tBHQ inhibited the production of ROS through MAPK and NF-κB signaling pathways, which can alleviate OA (42). In addition, IL-1β can induce chondrocyte apoptosis, promote inflammation and inhibit differentiation. tBHQ treatment prevents this effect of IL-1β (42). In DMM-induced mouse model, tBHQ significantly reduces the expression levels of IL-6, TNF-α and IL-1β and inhibits the development of OA in mice (42). In brief, tBHQ may be used for treating OA.

Angelicin. In China, Angelica sinensis is a commonly used herbal medicine to treat a variety of ailments. It is reported that Angelica sinensis extract has antioxidant and anti-OA effects (43,44). Angelicin (ANG) is an active ingredient isolated from Angelica sinensis, which has the functions of anticoagulation, analgesia and hemostasis. Therefore, ANG is considered to have a strong immunomodulatory effect (45).

ANG significantly inhibits the transformation of macrophages from M0 to M1 and the repolarization from M2 to M1 is also significantly inhibited (46). From the perspective of signaling pathway, ANG regulated macrophage polarization by activating the STAT pathway to reduce secretion of pro-inflammatory factors. In in vivo experiments, ANG effectively inhibits LPS-induced M1 polarization (46). The shape of cartilage in the ANG treatment group was similar to that of healthy cartilage, with smooth cartilage surface and complete cartilage matrix. Although experimental data indicate that ANG is effective in alleviating OA, more studies are needed to expand the applicability of this study. Overall, these results suggest that ANG has certain curative effects on OA.

Fargesin. Fargesin is a type of lignan with pharmacological activity and is extracted from Flos magnoliae (FM), which has long been used in the treatment of emphysema, sinusitis and headaches. It is reported that FM extracts have significant anti-allergic and anti-inflammatory effects (47). Recent studies have found that fargesin is critical for inflammatory response and glucose metabolism (48).

In terms of signaling pathways, fargesin promotes the polarization of macrophages into M2 through p38/ERK, P65/NF-κB pathways. In addition, anti-inflammatory factors in synovium are increased after fargesin treatment (49). Following fargesin treatment, the markers of cartilage catabolism (MMP13, collagen type X and Runt-related transcription factor 2) decrease and the markers of cartilage formation (col2a1 and Sox9) increase. This is through the mechanism of paracrine macrophages. In vivo, fargesin treatment alleviates synovitis and cartilage damage in CIOA-induced mice (49). Therefore, in-depth study of the signaling pathways regulating macrophage polarization and reprogramming or preventing macrophages from para-secreting cytokines to chondrocytes may be one direction for alleviation of OA.

Pseudolauric acid B. Pseudolaric acid B (PAB) is extracted from the dried cortex of the roots of Pseudolarix kaempferi Gord. It has a number of properties, such as antifungal, anti-angiogenesis and anti-inflammation (50). PAB may inhibit angiogenesis by promoting proteasome-mediated degradation (51).

In vitro experiments show that PAB inhibits the degradation of IκBα by inhibiting the phosphorylation of P65. At the same time, PAB inhibits NF-κB signaling by stabilizing peroxisome proliferator activated receptor γ to prevent M1 polarization and angiogenesis, thereby further reducing synovitis and OA progression (51). In vivo, mice treated with PAB show significantly reduced cartilage destruction and improved synovitis (52). These results expand the potential clinical application of PAB and strengthen the possibility that early targeting of synovitis or inhibiting angiogenesis may prevent OA (52).

Eucommia ulmoides polysaccharide (EUP). EU is a traditional herbal medicine that has been used in China for thousands of years (53). In recent years, a polysaccharide extracted from EU, as a drug component, has been studied for its immunomodulatory effect (53). It is reported that EUP has anti-inflammatory and anti-hypertensive effects. In terms of osteogenesis, it inhibits osteoclast and promotes osteogenesis (54-56).

EUP inhibits the expression of cytokines IL-6, IL-1β and IL-18, which inhibit chondrocyte apoptosis. In the extracellular matrix, cytokines activate MMPs to cut collagen fibers, so as to alleviate the progress of OA. In addition, the expression of osteogenesis and cartilage regeneration related genes such as BMP-6, TGF-β and Arg-1 increase under the treatment of EUP (57). At the same time, EUP treatment significantly decreases the expression of M1 macrophage markers (F4/80 and inducible nitric oxide synthase), while a M2 macrophage marker (CD206) increases (57). Micro-CT of living rabbit model showed that cartilage regeneration and subchondral bone reconstruction were evident following EUP treatment.

A retrospective study combined meloxicam with EUP in patients with OA showed significant improvement in pain, stiffness and dysfunction (58). Therefore, it can be considered that EUP alleviates the progress of OA and has the potential to treat OA.

Cells. Human adipose stem cells (hASCs)

hASCs are differentiated from mesenchymal stem cells (MSCs), which can secrete growth factors, chemokines and cytokines or other substances to improve the surrounding microenvironment and promote the growth of surrounding cells (59). hASCs secrete extracellular vesicles (EVs) to convey information between cells. EVs as carriers carry protein and other signal molecules, serving a role in targeting cells, such as chondrocytes and monocytes (60). EVs can improve OA by protecting chondrocytes from apoptosis and stimulating macrophages to polarize into the anti-inflammatory phenotype. Therefore, hASCs have been applied in regulating cell growth, regulating inflammatory response and repairing tissue defects (60,61).

In vitro, LPS was added to RAW264.7 cells to induce inflammation. The expression of proinflammatory cytokines (COX-2, IL-1, IL-6 and TNF-α) decreased significantly following hASCs-EVs treatment. In addition, hASCs-EVs promoted the transformation of macrophage to M2 subtype (62). In a mouse OA model, the number of M1 macrophages in OA synovium decrease and the expression of proinflammatory IL-1β in synovium and cartilage is also inhibited following intra-articular injection of EVs (62). In addition, hASCs-EVs treatment can regulate immune reactivity and effectively prevent proteoglycan degradation. It also reduces cartilage damage and slowed the progression of OA (62). In the DMM model, hASCs-EVs reduces the number of MMP-13 positive cells and reduces cartilage damage (62). Although this previous article shows that hASCs-EVs are helpful in reducing cartilage damage and OA, it is necessary to optimize the injection dose and study the mechanism in the future. Chen et al (63) studied the injection of ADSCs into the joint to treat knee OA and it effectively reduced pain scores and improved functional scores. In conclusion, hASCs-EVs should be considered as a potential treatment for OA.

Human umbilical cord mesenchymal stem cells (hUC-MSCs). There are two studies that reveal the effect of EVs secreted by hUC-MSCs on OA. The first study shows that hUC-MSC-EVs regulated OA in vitro mainly through the following: i) Releasing EVs to inhibit the infiltration of synovial M1 macrophages; ii) reducing the expression of CD14 and IL-1β, and iii) increasing the expression of CD206 and IL-10. In vivo experimental data show that in a rat model, hUC-MSC-EVs treatment can reverse the cartilage rupture and subchondral bone thickening (64).

The second study shows that hUC-MSC-EVs may transmit target proteins and microRNA through PI3K/Akt signaling pathway, so as to achieve the repolarization of M2 macrophages. In addition, the paracrine secretion of polarized M2 macrophages induced by hUC-MSC-EVs decreases the expression of MMP and apoptosis of chondrocytes. An in vivo experimental study reported that hUC-MSC-EVs can improve the progression of OA caused by ACLT (65).

Although hUC-MSCs have been proved to be effective in alleviating OA, there are still a number of problems with sEVs. The pain caused by repeated injections and the optimal dosage of injections are all difficult issues (64). However, hUC-MSC and their derivatives (such as EVs) have made remarkable achievements in promoting OA repair. These two studies further elucidated the molecular mechanism of hUC-MSC-EVs in the remission of OA and greatly improved the possibility of clinical application.

Proteins. Transient receptor potential vanilloid 1 (TRPV1)

TRPV1 is highly expressed on primary afferent neurons and participates in the conduction of pain (66). Activation of TRPV1 is closely associated with inflammation. However, TRPV1 exhibits both pro-inflammatory and anti-inflammatory effects, suggesting that the mechanism by which TRPV1 regulates inflammation may be complex (67).

In vitro experimental studies show that at the level of signaling pathways, TRPV1 inhibits the polarization of M1 macrophages in synovium through Ca2+/CaMKII/Nrf2 axis, thereby reversing the progress of OA (68). A rat OA model showed that activation of TRPV1 could reduce knee swelling, improve synovitis score and reduce M1 macrophage level, so as to reduce cartilage destruction and osteophyte formation (68).

In a study in which the target disease was knee OA, the pain of patients was significantly reduced after the application of TRPV1 antagonist (69). In conclusion, this previous paper clarifies that TRPV1 is an attractive research direction and reveals a new mechanism of local capsaicin in the treatment of OA.

Milk fat globule epidermal growth factor 8 (MFG-E8)

MFG-E8, also known as lactomyxin, is a peripheral secreted glycoprotein that can phagocytize apoptotic cells (70). In addition to being used to clear apoptotic cells, accumulating evidence shows that MFG-E8 serves a critical role in inhibiting inflammation, repairing injury, arterial remodeling and improving prognosis (70).

At the signaling pathway level, MFG-E8 targets chondrocyte growth and macrophage repolarization through the NF-κB pathway to prevent OA (71). In addition, lack of MFG-E8 leads to dysregulation of articular cartilage synthesis and decomposition homeostasis. Exogenous administration of MFG-E8 can promote the transformation of macrophages into M2 and reduce the release of inflammatory cytokines during the development of OA (72). In a DMM-mediated mouse model, MFG-E8 can prevent OA from developing from cartilage injury to osteophyte formation (72).

In conclusion, these results broaden the field of clinical application of MFG-E8. Targeting MFG-E8 through intra-articular supplements represents a method to delay the development of OA.

E3 ubiquitin ligase ITCH protein

Ubiquitin E1, E2 and E3 ligases can mediate ubiquitination. This modification occurs at the level of protein translation. Proteins usually degrade and lose their biological functions after ubiquitination. ITCH is a monomeric protein that serves a key role in different cellular environments, including DNA damage, immune response, T cell differentiation and cell death (73).

In vitro data show that ITCH-deficient macrophageswill promote the pro-inflammatory polarization of macrophages and aggravate the progress of post-traumatic OA (PTOA). In vivo studies show that in the mouse OA model, the decrease of ITCH protein level is related to the severity of PTOA, indicating that the decrease of ITCH protein level may aggravate human OA (74).

In conclusion, ubiquitination and degradation of ITCH protein may be involved in PTOA process and studying the mechanism of its degradation may be an exciting direction for preventing the progression of OA (Fig. 2).

Fig. 2 shows the relevant pathways of substances involved in macrophage polarization regulation in different studies. A previous study reported that the pathway underlying how hUC-MSCs regulate macrophage polarization is unknown (62), whereas another previous study reported that hUC-MSCs act through the PI3K/Akt pathway (64).

5. Conclusion

OA is a chronic disease that causes physical disability. By 2030, the number of OA patients in the United States is expected to increase to ~67 million (75). Traditional treatments include physical therapy, anti-inflammatory and analgesic drugs and injections of glucocorticoids or hyaluronic acid into the joints. However, these therapies are ineffective and have side effects. For example, non-steroidal anti-inflammatory drugs are associated with cardiovascular risk and gastrointestinal bleeding problems (76,77). The optimal dose and time of intra-articular hormone injection remain controversial (6). Gene editing, including RNAi and CRISPR/Cas9 is an emerging therapy for OA. RNAi is regulated by small interfering RNA (siRNA) at the level of gene transcription (78). In OA, a number of animal studies confirm that RNAi can reduce cartilage degradation and alleviate OA (79-81). However, there are currently no clinical trials using siRNA for OA treatment. CRISPR/Ccas9 has great potential in the treatment of OA. One study summarizes the potential targets of CRISPR/Cas9 in the treatment of OA (82). However, CRISPR/Cas9 has ethical and effective targeting range problems. Notably, gene editing can be easily completed using CRISPR/Cas9, which raises concerns regarding the application of this technology to embryos (83). Single guide RNA (sgRNA) can only bind to a specific region near the PAM sequence on DNA (82). The PAM sequence for Cas9 is 5'-NGG-3', where ‘N’ can be any nucleotide base, but the third base must be G. This results in a significant reduction in the potential target locations for DNA editing (83).

However, these OA treatment methods can only improve the symptoms of OA but cannot reverse its process. Immune microenvironment serves an important role in OA (18). Osteoarthritis is characterized by the invasion and activation of macrophages and lymphocytes. Macrophages will polarize into M1 macrophages or M2 macrophages following stimulation. Regulating the phenotype of macrophages is important in the treatment of OA and may be the most promising treatment.

The novelty of the present study is from the perspective of immunology; it collated the latest studies of the enriched signal pathways and target molecules of macrophage polarization in OA, so as to provide insights into the treatment of OA. The pathways involved are ERK, PI3K/Akt, MAPK, STAT and NF-κB pathways, different substances including compounds, cells and proteins can alleviate OA by regulating the balance of M1 and M2 macrophages polarization. This suggests that maintaining the homeostasis of M1 macrophages and M2 macrophages is important for the treatment of OA. In addition, in the in vivo models of these studies, cartilage destruction has been improved, which again shows that cartilage homeostasis is very important for the development of OA.

Unfortunately, most of these studies are only in the preclinical stage and further experiments are needed to verify the efficacy of their clinical use. It may bring more benefits to the future research on macrophage regulation.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the Science and Technology Project of Nantong City (grant no. JC2020013) and Postgraduate Research and Practice Innovation Program of Jiangsu Province (grant no. KYCX21_3107).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

BSC, HXH and ZMC contributed to the study design, participated in the review process and prepared the first draft. YYS, CC and GFB contributed to collecting the relevant literature and important information, generating figures and modified the manuscript. BSC, ZMC, CSW and GHX conceived the review and approved the final version of the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

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 

Zhang Y and Jordan JM: Epidemiology of osteoarthritis. Clin Geriatr Med. 26:355–369. 2010.PubMed/NCBI View Article : Google Scholar

2 

Wallace IJ, Worthington S, Felson DT, Jurmain RD, Wren KT, Maijanen H, Woods RJ and Lieberman DE: Knee osteoarthritis has doubled in prevalence since the mid-20th century. Proc Natl Acad Sci USA. 114:9332–9336. 2017.PubMed/NCBI View Article : Google Scholar

3 

Young DA, Barter MJ and Wilkinson DJ: Recent advances in understanding the regulation of metalloproteinases. F1000Res. 8(F1000 Faculty Rev-195)2019.PubMed/NCBI View Article : Google Scholar

4 

Glazier RH, Dalby DM, Badley EM, Hawker GA, Bell MJ, Buchbinder R and Lineker SC: Management of common musculoskeletal problems: A survey of Ontario primary care physicians. CMAJ. 158:1037–1040. 1998.PubMed/NCBI

5 

Gnylorybov AM, Ter-Vartanian SK, Golovach IY, Vyrva OE, Burianov OA, Yesirkepova GS, Irismetov ME, Rizamuhamedova MZ, Vardanyan VS and Ginosyan KV: Expert opinion on the extensive use of prescription crystalline glucosamine sulfate in the multimodal treatment of osteoarthritis in Ukraine, Kazakhstan, Uzbekistan, and Armenia. Clin Med Insights Arthritis Musculoskelet Disord. 13(1179544120946743)2020.PubMed/NCBI View Article : Google Scholar

6 

Nowaczyk A, Szwedowski D, Dallo I and Nowaczyk J: Overview of first-line and second-line pharmacotherapies for osteoarthritis with special focus on intra-articular treatment. Int J Mol Sci. 23(1566)2022.PubMed/NCBI View Article : Google Scholar

7 

Le Graverand-Gastineau MP: Disease modifying osteoarthritis drugs: Facing development challenges and choosing molecular targets. Curr Drug Targets. 11:528–535. 2010.PubMed/NCBI View Article : Google Scholar

8 

Thomson A and Hilkens CMU: Synovial macrophages in osteoarthritis: The key to understanding pathogenesis? Front Immunol. 12(678757)2021.PubMed/NCBI View Article : Google Scholar

9 

Zhu X, Lee CW, Xu H, Wang YF, Yung PSH, Jiang Y and Lee OK: Phenotypic alteration of macrophages during osteoarthritis: A systematic review. Arthritis Res Ther. 23(110)2021.PubMed/NCBI View Article : Google Scholar

10 

Rao KN and Brown MA: Mast cells: Multifaceted immune cells with diverse roles in health and disease. Ann N Y Acad Sci. 1143:83–104. 2008.PubMed/NCBI View Article : Google Scholar

11 

Xie J, Huang Z, Yu X, Zhou L and Pei F: Clinical implications of macrophage dysfunction in the development of osteoarthritis of the knee. Cytokine Growth Factor Rev. 46:36–44. 2019.PubMed/NCBI View Article : Google Scholar

12 

Zhang H, Cai D and Bai X: Macrophages regulate the progression of osteoarthritis. Osteoarthritis Cartilage. 28:555–561. 2020.PubMed/NCBI View Article : Google Scholar

13 

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

14 

Jeffries MA: Osteoarthritis year in review 2018: Genetics and epigenetics. Osteoarthritis Cartilage. 27:371–377. 2019.PubMed/NCBI View Article : Google Scholar

15 

DeFrate LE, Kim-Wang SY, Englander ZA and McNulty AL: Osteoarthritis year in review 2018: Mechanics. Osteoarthritis Cartilage. 27:392–400. 2019.PubMed/NCBI View Article : Google Scholar

16 

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

17 

Nunez-Carro C, Blanco-Blanco M, Montoya T, Villagran-Andrade KM, Hermida-Gomez T, Blanco FJ and de Andres MC: Histone extraction from human articular cartilage for the study of epigenetic regulation in osteoarthritis. Int J Mol Sci. 23(3355)2022.PubMed/NCBI View Article : Google Scholar

18 

Li M, Yin H, Yan Z, Li H, Wu J, Wang Y, Wei F, Tian G, Ning C, Li H, et al: The immune microenvironment in cartilage injury and repair. Acta Biomater. 140:23–42. 2022.PubMed/NCBI View Article : Google Scholar

19 

Bhat S, Tripathi A and Kumar A: Supermacroprous chitosan-agarose-gelatin cryogels: In vitro characterization and in vivo assessment for cartilage tissue engineering. J R Soc Interface. 8:540–554. 2011.PubMed/NCBI View Article : Google Scholar

20 

Sandell LJ, Morris N, Robbins JR and Goldring MB: Alternatively spliced type II procollagen mRNAs define distinct populations of cells during vertebral development: Differential expression of the amino-propeptide. J Cell Biol. 114:1307–1319. 1991.PubMed/NCBI View Article : Google Scholar

21 

Stocker W and Bode W: Structural features of a superfamily of zinc-endopeptidases: The metzincins. Curr Opin Struct Biol. 5:383–390. 1995.PubMed/NCBI View Article : Google Scholar

22 

Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj and Horwitz E: Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 8:315–317. 2006.PubMed/NCBI View Article : Google Scholar

23 

Chan CM, Macdonald CD, Litherland GJ, Wilkinson DJ, Skelton A, Europe-Finner GN and Rowan AD: Cytokine-induced MMP13 expression in human chondrocytes is dependent on activating transcription factor 3 (ATF3) Regulation. J Biol Chem. 292:1625–1636. 2017.PubMed/NCBI View Article : Google Scholar

24 

Heijink A, Vanhees M, van den Ende K, van den Bekerom MP, van Riet RP, Van Dijk CN and Eygendaal D: Biomechanical considerations in the pathogenesis of osteoarthritis of the elbow. Knee Surg Sports Traumatol Arthrosc. 24:2313–2318. 2016.PubMed/NCBI View Article : Google Scholar

25 

Stannus O, Jones G, Cicuttini F, Parameswaran V, Quinn S, Burgess J and Ding C: Circulating levels of IL-6 and TNF-α are associated with knee radiographic osteoarthritis and knee cartilage loss in older adults. Osteoarthritis Cartilage. 18:1441–1447. 2010.PubMed/NCBI View Article : Google Scholar

26 

Kuyinu EL, Narayanan G, Nair LS and Laurencin CT: Animal models of osteoarthritis: Classification, update, and measurement of outcomes. J Orthop Surg Res. 11(19)2016.PubMed/NCBI View Article : Google Scholar

27 

Blaker CL, Clarke EC and Little CB: Using mouse models to investigate the pathophysiology, treatment, and prevention of post-traumatic osteoarthritis. J Orthop Res. 35:424–439. 2017.PubMed/NCBI View Article : Google Scholar

28 

Man GS and Mologhianu G: Osteoarthritis pathogenesis-a complex process that involves the entire joint. J Med Life. 7:37–41. 2014.PubMed/NCBI

29 

Gordon S: Pattern recognition receptors: Doubling up for the innate immune response. Cell. 111:927–930. 2002.PubMed/NCBI View Article : Google Scholar

30 

Nefla M, Holzinger D, Berenbaum F and Jacques C: The danger from within: Alarmins in arthritis. Nat Rev Rheumatol. 12:669–683. 2016.PubMed/NCBI View Article : Google Scholar

31 

Ito T, Kurata N and Fukunaga Y: Tissue-Resident Macrophages in the Stria Vascularis. Front Neurol. 13(818395)2022.PubMed/NCBI View Article : Google Scholar

32 

Davies LC and Taylor PR: Tissue-resident macrophages: Then and now. Immunology. 144:541–548. 2015.PubMed/NCBI View Article : Google Scholar

33 

Wynn TA and Vannella KM: Macrophages in tissue repair, regeneration, and fibrosis. Immunity. 44:450–462. 2016.PubMed/NCBI View Article : Google Scholar

34 

Geiß C, Salas E, Guevara-Coto J, Regnier-Vigouroux A and Mora-Rodriguez RA: Multistability in macrophage activation pathways and metabolic implications. Cells. 11(404)2022.PubMed/NCBI View Article : Google Scholar

35 

Andersen A, Lund A, Knop FK and Vilsboll T: Glucagon-like peptide 1 in health and disease. Nat Rev Endocrinol. 14:390–403. 2018.PubMed/NCBI View Article : Google Scholar

36 

Schisano B, Harte AL, Lois K, Saravanan P, Al-Daghri N, Al-Attas O, Knudsen LB, McTernan PG, Ceriello A and Tripathi G: GLP-1 analogue, Liraglutide protects human umbilical vein endothelial cells against high glucose induced endoplasmic reticulum stress. Regul Pept. 174:46–52. 2012.PubMed/NCBI View Article : Google Scholar

37 

Mehan S, Bhalla S, Siddiqui EM, Sharma N, Shandilya A and Khan A: Potential roles of glucagon-like peptide-1 and its analogues in dementia targeting impaired insulin secretion and neurodegeneration. Degener Neurol Neuromuscul Dis. 12:31–59. 2022.PubMed/NCBI View Article : Google Scholar

38 

Meurot C, Martin C, Sudre L, Breton J, Bougault C, Rattenbach R, Bismuth K, Jacques C and Berenbaum F: Liraglutide, a glucagon-like peptide 1 receptor agonist, exerts analgesic, anti-inflammatory and anti-degradative actions in osteoarthritis. Sci Rep. 12(1567)2022.PubMed/NCBI View Article : Google Scholar

39 

Gudbergsen H, Overgaard A, Henriksen M, Waehrens EE, Bliddal H, Christensen R, Nielsen SM, Boesen M, Knop FK, Astrup A, et al: Liraglutide after diet-induced weight loss for pain and weight control in knee osteoarthritis: A randomized controlled trial. Am J Clin Nutr. 113:314–323. 2021.PubMed/NCBI View Article : Google Scholar

40 

Li J, Bi Y, Yang H and Wang D: Antioxidative properties and interconversion of tert-Butylhydroquinone and tert-Butylquinone in Soybean Oils. J Agric Food Chem. 65:10598–10603. 2017.PubMed/NCBI View Article : Google Scholar

41 

Song H, Xu Y, Yang X, Rong X, Wang Y and Wei N: Tertiary butylhydroquinone alleviates gestational diabetes mellitus in C57BL/KsJ-Lep db/+ mice by suppression of oxidative stress. J Cell Biochem. 120:15310–15319. 2019.PubMed/NCBI View Article : Google Scholar

42 

Zhang H, Li J, Xiang X, Zhou B, Zhao C, Wei Q, Sun Y, Chen J, Lai B, Luo Z and Li A: Tert-butylhydroquinone attenuates osteoarthritis by protecting chondrocytes and inhibiting macrophage polarization. Bone Joint Res. 10:704–713. 2021.PubMed/NCBI View Article : Google Scholar

43 

Wu SJ, Ng LT and Lin CC: Antioxidant activities of some common ingredients of traditional chinese medicine, Angelica sinensis, Lycium barbarum and Poria cocos. Phytother Res. 18:1008–1012. 2004.PubMed/NCBI View Article : Google Scholar

44 

Qin J, Liu YS, Liu J, Li J, Tan Y, Li XJ, Magdalou J, Mei QB, Wang H and Chen LB: Effect of angelica sinensis polysaccharides on osteoarthritis in vivo and in vitro: A possible mechanism to promote proteoglycans synthesis. Evid Based Complement Alternat Med. 2013(794761)2013.PubMed/NCBI View Article : Google Scholar

45 

Lampronti I, Manzione MG, Sacchetti G, Ferrari D, Spisani S, Bezzerri V, Finotti A, Borgatti M, Dechecchi MC, Miolo G, et al: Differential effects of angelicin analogues on NF-κB Activity and IL-8 gene expression in cystic fibrosis IB3-1 cells. Mediators Inflamm. 2017(2389487)2017.PubMed/NCBI View Article : Google Scholar

46 

Tian Z, Zeng F, Zhao C and Dong S: Angelicin alleviates post-trauma osteoarthritis progression by regulating macrophage polarization via STAT3 signaling pathway. Front Pharmacol. 12(669213)2021.PubMed/NCBI View Article : Google Scholar

47 

Hong PTL, Kim HJ, Kim WK and Nam JH: Flos magnoliae constituent fargesin has an anti-allergic effect via ORAI1 channel inhibition. Korean J Physiol Pharmacol. 25:251–258. 2021.PubMed/NCBI View Article : Google Scholar

48 

Lee GE, Lee CJ, An HJ, Kang HC, Lee HS, Lee JY, Oh SR, Cho SJ, Kim DJ and Cho YY: Fargesin Inhibits EGF-Induced cell transformation and colon cancer cell growth by suppression of CDK2/Cyclin E signaling pathway. Int J Mol Sci. 22(2073)2021.PubMed/NCBI View Article : Google Scholar

49 

Lu J and Zhang H, Pan J, Hu Z, Liu L, Liu Y, Yu X, Bai X, Cai D and Zhang H: Fargesin ameliorates osteoarthritis via macrophage reprogramming by downregulating MAPK and NF-κB pathways. Arthritis Res Ther. 23(142)2021.PubMed/NCBI View Article : Google Scholar

50 

Guan D, Li C, Lv X and Yang Y: Pseudolaric acid B inhibits PAX2 expression through Wnt signaling and induces BAX expression, therefore promoting apoptosis in HeLa cervical cancer cells. J Gynecol Oncol. 30(e77)2019.PubMed/NCBI View Article : Google Scholar

51 

Li MH, Miao ZH, Tan WF, Yue JM, Zhang C, Lin LP, Zhang XW and Ding J: Pseudolaric acid B inhibits angiogenesis and reduces hypoxia-inducible factor 1alpha by promoting proteasome-mediated degradation. Clin Cancer Res. 10:8266–8274. 2004.PubMed/NCBI View Article : Google Scholar

52 

Lu J, Guan H, Wu D, Hu Z, Zhang H, Jiang H, Yu J, Zeng K, Li H, Zhang H, et al: Pseudolaric acid B ameliorates synovial inflammation and vessel formation by stabilizing PPARγ to inhibit NF-κB signalling pathway. J Cell Mol Med. 25:6664–6678. 2021.PubMed/NCBI View Article : Google Scholar

53 

He X, Wang J, Li M, Hao D, Yang Y, Zhang C, He R and Tao R: Eucommia ulmoides Oliv: Ethnopharmacology, phytochemistry and pharmacology of an important traditional Chinese medicine. J Ethnopharmacol. 151:78–92. 2014.PubMed/NCBI View Article : Google Scholar

54 

Hong YK, Liu WJ, Li T and She SY: Optimization of extraction of Eucommia ulmoides polysaccharides by response surface methodology. Carbohydr Polym. 92:1761–1766. 2013.PubMed/NCBI View Article : Google Scholar

55 

Deng Y, Ma F, Ruiz-Ortega LI, Peng Y, Tian Y, He W and Tang B: Fabrication of strontium Eucommia ulmoides polysaccharides and in vitro evaluation of their osteoimmunomodulatory property. Int J Biol Macromol. 140:727–735. 2019.PubMed/NCBI View Article : Google Scholar

56 

Gao W, Feng Z, Zhang S, Wu B, Geng X, Fan G, Duan Y, Li K, Liu K and Peng C: Anti-Inflammatory and Antioxidant Effect of Eucommia ulmoides Polysaccharide in Hepatic Ischemia-Reperfusion Injury by Regulating ROS and the TLR-4-NF-κB Pathway. Biomed Res Int. 2020(1860637)2020.PubMed/NCBI View Article : Google Scholar

57 

Sun Y, Huang K, Mo L, Ahmad A, Wang D, Rong Z, Peng H, Cai H and Liu G: Eucommia ulmoides polysaccharides attenuate rabbit osteoarthritis by regulating the function of macrophages. Front Pharmacol. 12(730557)2021.PubMed/NCBI View Article : Google Scholar

58 

Hu CX, Hu KY and Wang JF: Potential role of the compound Eucommia bone tonic granules in patients with osteoarthritis and osteonecrosis: A retrospective study. World J Clin Cases. 8:46–53. 2020.PubMed/NCBI View Article : Google Scholar

59 

Mazini L, Rochette L, Admou B, Amal S and Malka G: Hopes and limits of adipose-derived stem cells (ADSCs) and mesenchymal stem cells (MSCs) in wound healing. Int J Mol Sci. 21(1306)2020.PubMed/NCBI View Article : Google Scholar

60 

Cosenza S, Ruiz M, Toupet K, Jorgensen C and Noel D: Mesenchymal stem cells derived exosomes and microparticles protect cartilage and bone from degradation in osteoarthritis. Sci Rep. 7(16214)2017.PubMed/NCBI View Article : Google Scholar

61 

Furuta T, Miyaki S, Ishitobi H, Ogura T, Kato Y, Kamei N, Miyado K, Higashi Y and Ochi M: Mesenchymal stem cell-derived exosomes promote fracture Healing in a mouse model. Stem Cells Transl Med. 5:1620–1630. 2016.PubMed/NCBI View Article : Google Scholar

62 

Woo CH, Kim HK, Jung GY, Jung YJ, Lee KS, Yun YE, Han J, Lee J, Kim WS, Choi JS, et al: Small extracellular vesicles from human adipose-derived stem cells attenuate cartilage degeneration. J Extracell Vesicles. 9(1735249)2020.PubMed/NCBI View Article : Google Scholar

63 

Chen CF, Hu CC, Wu CT, Wu HH, Chang CS, Hung YP, Tsai CC and Chang Y: Treatment of knee osteoarthritis with intra-articular injection of allogeneic adipose-derived stem cells (ADSCs) ELIXCYTE®: a phase I/II, randomized, active-control, single-blind, multiple-center clinical trial. Stem Cell Res Ther. 12(562)2021.PubMed/NCBI View Article : Google Scholar

64 

Tang S, Chen P, Zhang H, Weng H, Fang Z, Chen C, Peng G, Gao H, Hu K, Chen J, et al: Comparison of curative effect of human umbilical cord-derived mesenchymal stem cells and their small extracellular vesicles in treating osteoarthritis. Int J Nanomedicine. 16:8185–8202. 2021.PubMed/NCBI View Article : Google Scholar

65 

Li K, Yan G, Huang H, Zheng M, Ma K, Cui X, Lu D, Zheng L, Zhu B, Cheng J and Zhao J: Anti-inflammatory and immunomodulatory effects of the extracellular vesicles derived from human umbilical cord mesenchymal stem cells on osteoarthritis via M2 macrophages. J Nanobiotechnology. 20(38)2022.PubMed/NCBI View Article : Google Scholar

66 

Mickle AD, Shepherd AJ and Mohapatra DP: Sensory TRP channels: The key transducers of nociception and pain. Prog Mol Biol Transl Sci. 131:73–118. 2015.PubMed/NCBI View Article : Google Scholar

67 

Zhang X, Ye L, Huang Y, Ding X and Wang L: The potential role of TRPV1 in pulmonary hypertension: Angel or demon? Channels (Austin). 13:235–246. 2019.PubMed/NCBI View Article : Google Scholar

68 

Lv Z, Xu X, Sun Z, Yang YX, Guo H, Li J, Sun K, Wu R, Xu J, Jiang Q, et al: TRPV1 alleviates osteoarthritis by inhibiting M1 macrophage polarization via Ca2+/CaMKII/Nrf2 signaling pathway. Cell Death Dis. 12(504)2021.PubMed/NCBI View Article : Google Scholar

69 

Mayorga AJ, Flores CM, Trudeau JJ, Moyer JA, Shalayda K, Dale M, Frustaci ME, Katz N, Manitpisitkul P, Treister R, et al: A randomized study to evaluate the analgesic efficacy of a single dose of the TRPV1 antagonist mavatrep in patients with osteoarthritis. Scand J Pain. 17:134–143. 2017.PubMed/NCBI View Article : Google Scholar

70 

Yi YS: Functional Role of Milk fat globule-epidermal growth factor VIII in macrophage-mediated inflammatory responses and inflammatory/autoimmune diseases. Mediators Inflamm. 2016(5628486)2016.PubMed/NCBI View Article : Google Scholar

71 

Uchiyama A, Motegi SI, Sekiguchi A, Fujiwara C, Perera B, Ogino S, Yokoyama Y and Ishikawa O: Mesenchymal stem cells-derived MFG-E8 accelerates diabetic cutaneous wound healing. J Dermatol Sci. 86:187–197. 2017.PubMed/NCBI View Article : Google Scholar

72 

Lu Y, Liu L, Pan J, Luo B, Zeng H, Shao Y, Zhang H, Guan H, Guo D, Zeng C, et al: MFG-E8 regulated by miR-99b-5p protects against osteoarthritis by targeting chondrocyte senescence and macrophage reprogramming via the NF-κB pathway. Cell Death Dis. 12(533)2021.PubMed/NCBI View Article : Google Scholar

73 

Han S, Zhang Y, Guo C and Chang C: The E3 protein ubiquitin ligase Itch is a potential target in myeloid malignancies with marrow fibrosis. Transl Cancer Res. 10:2368–2378. 2021.PubMed/NCBI View Article : Google Scholar

74 

Lin X, Wang W, McDavid A, Xu H, Boyce BF and Xing L: The E3 ubiquitin ligase Itch limits the progression of post-traumatic osteoarthritis in mice by inhibiting macrophage polarization. Osteoarthritis Cartilage. 29:1225–1236. 2021.PubMed/NCBI View Article : Google Scholar

75 

Hootman JM and Helmick CG: Projections of US prevalence of arthritis and associated activity limitations. Arthritis Rheum. 54:226–229. 2006.PubMed/NCBI View Article : Google Scholar

76 

Cutolo M, Berenbaum F, Hochberg M, Punzi L and Reginster JY: Commentary on recent therapeutic guidelines for osteoarthritis. Semin Arthritis Rheum. 44:611–617. 2015.PubMed/NCBI View Article : Google Scholar

77 

Zhang W, Nuki G, Moskowitz RW, Abramson S, Altman RD, Arden NK, Bierma-Zeinstra S, Brandt KD, Croft P, Doherty M, et al: OARSI recommendations for the management of hip and knee osteoarthritis: Part III: Changes in evidence following systematic cumulative update of research published through January 2009. Osteoarthritis Cartilage. 18:476–499. 2010.PubMed/NCBI View Article : Google Scholar

78 

Rai MF, Pan H, Yan H, Sandell LJ, Pham CTN and Wickline SA: Applications of RNA interference in the treatment of arthritis. Transl Res. 214:1–16. 2019.PubMed/NCBI View Article : Google Scholar

79 

Ye C, Bhan AK, Deshpande V, Shankar P and Manjunath N: Silencing TNF-α in macrophages and dendritic cells for arthritis treatment. Scand J Rheumatol. 42:266–269. 2013.PubMed/NCBI View Article : Google Scholar

80 

Shi Q, Rondon-Cavanzo EP, Dalla Picola IP, Tiera MJ, Zhang X, Dai K, Benabdoune HA, Benderdour M and Fernandes JC: In vivo therapeutic efficacy of TNFα silencing by folate-PEG-chitosan-DEAE/siRNA nanoparticles in arthritic mice. Int J Nanomedicine. 13:387–402. 2018.PubMed/NCBI View Article : Google Scholar

81 

Lee SJ, Lee A, Hwang SR, Park JS, Jang J, Huh MS, Jo DG, Yoon SY, Byun Y, Kim SH, et al: TNF-α gene silencing using polymerized siRNA/thiolated glycol chitosan nanoparticles for rheumatoid arthritis. Mol Ther. 22:397–408. 2014.PubMed/NCBI View Article : Google Scholar

82 

Tanikella AS, Hardy MJ, Frahs SM, Cormier AG, Gibbons KD, Fitzpatrick CK and Oxford JT: Emerging gene-editing modalities for osteoarthritis. Int J Mol Sci. 21(6046)2020.PubMed/NCBI View Article : Google Scholar

83 

Nishimasu H, Shi X, Ishiguro S, Gao L, Hirano S, Okazaki S, Noda T, Abudayyeh OO, Gootenberg JS, Mori H, et al: Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science. 361:1259–1262. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2022
Volume 24 Issue 6

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
Chen B, Hong H, Sun Y, Chen C, Wu C, Xu G, Bao G and Cui Z: Role of macrophage polarization in osteoarthritis (Review). Exp Ther Med 24: 757, 2022
APA
Chen, B., Hong, H., Sun, Y., Chen, C., Wu, C., Xu, G. ... Cui, Z. (2022). Role of macrophage polarization in osteoarthritis (Review). Experimental and Therapeutic Medicine, 24, 757. https://doi.org/10.3892/etm.2022.11693
MLA
Chen, B., Hong, H., Sun, Y., Chen, C., Wu, C., Xu, G., Bao, G., Cui, Z."Role of macrophage polarization in osteoarthritis (Review)". Experimental and Therapeutic Medicine 24.6 (2022): 757.
Chicago
Chen, B., Hong, H., Sun, Y., Chen, C., Wu, C., Xu, G., Bao, G., Cui, Z."Role of macrophage polarization in osteoarthritis (Review)". Experimental and Therapeutic Medicine 24, no. 6 (2022): 757. https://doi.org/10.3892/etm.2022.11693