Next Article in Journal
CVAM: CNA Profile Inference of the Spatial Transcriptome Based on the VGAE and HMM
Next Article in Special Issue
LEO1 Is Required for Efficient Entry into Quiescence, Control of H3K9 Methylation and Gene Expression in Human Fibroblasts
Previous Article in Journal
Real-Time Assessment of Intracellular Metabolites in Single Cells through RNA-Based Sensors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Targets and Strategies for Rheumatoid Arthritis: From Signal Transduction to Epigenetic Aspect

1
State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
2
Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(5), 766; https://doi.org/10.3390/biom13050766
Submission received: 16 March 2023 / Revised: 17 April 2023 / Accepted: 21 April 2023 / Published: 28 April 2023
(This article belongs to the Special Issue Histone Modifications in Health and Diseases)

Abstract

:
Rheumatoid arthritis (RA) is a chronic autoimmune disease that can lead to joint damage and even permanent disability, seriously affecting patients’ quality of life. At present, the complete cure for RA is not achievable, only to relieve the symptoms to reduce the pain of patients. Factors such as environment, genes, and sex can induce RA. Presently, non-steroidal anti-inflammatory drugs, DRMADs, and glucocorticoids are commonly used in treating RA. In recent years, some biological agents have also been applied in clinical practice, but most have side effects. Therefore, finding new mechanisms and targets for treating RA is necessary. This review summarizes some potential targets discovered from the perspective of epigenetics and RA mechanisms.

1. Introduction

Rheumatoid arthritis (RA) is a chronic autoimmune disease that affects the joints and can lead to inflammation, pain, stiffness, and eventually, joint damage and disability. According to statistics, not only does the prevalence of rheumatoid arthritis vary widely among ethnic groups, but it is more common in women than in men. Lifestyle habits can also affect the incidence of RA. The impact of smoking on RA is enormous. Studies have shown that people who have smoked for 20 years are twice as likely to develop RA as people who do not smoke [1]. Relevant data also support the impact of nutritional habits on RA. Studies have shown that overeating food containing saturated fat, trans fat, high salt, and high calories will promote inflammation and induce insulin resistance, increasing the risk of RA [2,3]. Studies have shown that people with RA have different gut microbiomes compared to those without RA and that certain bacteria in the gut can affect inflammation and joint damage [4]. Data have shown that the diversity of intestinal flora in RA patients is lower than in patients without RA.
In the early stage of RA, the immune system is activated and inflammatory cells, such as macrophages, T cells, and B cells, infiltrate the synovial tissue of the affected joint. With the intensification of immune cell penetration, RA gradually becomes severe, eventually leading to hyperplasia of the lining layer and pannus formation. The healthy synovial tissue mainly contains macrophage-like synovial (MLS) and fibroblast-like synovial (FLS), while the synovial tissue of RA was heavily infiltrated with leukocytes [5]. Synovial inflammation in RA is maintained by interactions between T cells, macrophages, and FLS. In the joint, once the white blood cells are recruited and activated, they cannot be stopped, thus causing inflammation and causing damage to the joint [6]. Studies have shown that non-classical monocytes play an essential role in RA [7]. During an RA flare-up, neutrophils are activated and migrate to the joints, where they release inflammatory molecules that contribute to the destruction of joint tissue [8]. In the inflammatory milieu of the joints, it is theorized that the recruited monocytes differentiate into macrophages and produce inflammatory mediators, thus activating the immune system and inducing RA [9]. The abnormal proliferation and invasion of FLS caused by the secretion of pro-inflammatory factors play an important role in joint injury and inflammation. We summarize the structure of the joint and the changes in rheumatoid arthritis in Figure 1.
The development of RA: Under the stimulation of susceptibility factors, the adaptive immune system will recognize peptidyl arginine and peptidyl citrulline. Antigen-presenting cells (APCs) will transmit these changes to the lymphatic tissues to generate an immune response and autoantibodies. Then, FLS, APCs, and macrophages are activated to produce inflammatory cytokines (IL-6, 8, 17, etc.). Forming of the immune complex and activating of the complement will stimulate the immune system for a second time, increasing the production of cytokines and synovial vascular inflammation, eventually leading to synovial inflammation and bone destruction [10].
RA is characterized by inflammation of the synovial membrane, the proliferation of synovial cells and the formation of pannus, leading to the dysfunction of the synovial. At present, the treatment of RA is mainly to relieve pain, inhibit the proliferation of synovial cells, prevent the generation of pannus, regulate immune function and protect cartilage, and finally, achieve the goal of eliminating inflammation as far as possible [11,12]. Common drugs include non-steroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, and DMARDs (Disease-modifying anti-rheumatic drugs). NSAIDs’ primary mechanism of action is to inhibit cyclooxygenase activity from combating inflammation. NSAIDs are effective in the initial stages of RA and can be used in combination with DMARDs. However, long-term use of NSAIDs can cause adverse effects on the gastrointestinal tract and kidneys. A variety of cells are involved in the inflammatory response in RA. The effect of glucocorticoid is to relieve the pain and swelling of RA patients by inhibiting the activity of immune cells, including T cells and B cells, which play a role in the autoimmune response that leads to RA [13]. Glucocorticoids are divided into short-term (cortisone and cortisol), medium-term (methylprednisolone, prednisolone, methylprednisolone, and triamcinolone), and long-term (dexamethasone and betamethasone) treatments of RA [14]. In DMARDs, methotrexate (MTX) is often used in combination with a glucocorticoid as the primary drug for RA treatment due to its advantages, such as reliability, effectiveness, low cost, and few side effects. In addition, hydroxychloroquine, sulfasalazine, leflunomide, etc., can also be used in the treatment of RA.
Although there are many treatment options for RA, a large number of patients do not achieve remission, thus indicating the need for a more diverse approach to treatment. The pathogenesis of RA is complex and involves multiple factors, including genetic and epigenetic factors. Signal transduction pathways play a critical role in the pathogenesis of RA. The abnormal activation of various signaling pathways contributes to the production of proinflammatory cytokines, chemokines, and other inflammatory mediators. Epigenetic modifications, including DNA methylation, histone modifications, and noncoding RNA-mediated gene regulation, are also involved in the development and progression of RA. Epigenetic changes can alter gene expression patterns and contribute to the aberrant activation of signaling pathways and the dysregulation of immune responses. Aberrant signals in signal transduction and epigenetic modification often serve as targets for drug discovery [15]. In recent decades, the development of biologically targeted therapy has been very rapid and has provided us with novel ideas. Below, we will review the potential therapeutic targets for RA and provide a reference for subsequent research.

2. Cellular Mechanisms in RA

RA is a systemic immune disease causing synovial inflammation, joint swelling, and injury [16]. In the development of RA, the synovial membrane gradually becomes invasive, destroying cartilage and bone. The synovial intimal lining is composed of MLSs and FLSs in equal proportions. Usually, the synovial intimal lining is a thin and loose membrane-like structure with only one or two layers of cells. In comparison, RA intimal lining can be thick with 10–15 layers of cells [17]. MLSs and FLSs cells play an essential role in the development of RA. They mainly secrete a variety of cytokines to stimulate the occurrence of inflammation. In addition, the recruitment of macrophages, T cells, and B cells expands the synovial sublayer and promotes inflammation. The cellular mechanism in RA is shown in Figure 2.

2.1. Fibroblast-Like Synovial (FLSs)

In healthy synovial tissue, FLSs promote synovial fluid production, thus protecting cartilage. FLSs also maintain synovial ECM (extracellular matrix) by producing matrix components and ECM-degrading enzymes [18]. FLSs have beneficial effects on healthy tissues, but they also “commit indelible crimes” in the pathogenesis of RA. The source of the destruction of bone tissue and cartilage tissue is FLS-mediated. In RA, different immune cells produce a variety of cytokines and growth factors, leading to the production of inflammation and proliferation of FLSs, leading to the formation of pannus [18]. In addition, FLSs can also resist endoplasmic reticulum stress-induced apoptosis [19]. Metabolic changes of FLSs are also a feature of RA. Studies have shown that changes in the glucose metabolism pathway, lipid metabolism, and amino acid metabolism of RA FLSs can induce synovial hyperplasia and increase inflammation. Increased glycolysis in FLS was also a feature in determining RA. The Hypoxia-inducible factor 1α (HIF1α) is a transcription factor that can induce glycolysis, so targeting HIF1α has also been used as a treatment strategy for RA in recent years [20]. FLSs activate T cells and B cells by producing a variety of cytokines [21] and secreting various enzymes (MMP1, MMP13, MMP3, ADAMTS4, ADAMTS5, and so on) that promote the invasion of immune cells and aggravate inflammation [22].

2.2. Macrophage-Like Synovial (MLSs)

MLSs co-exist with FLSs in the synovium, but the function of MLSs is less diverse than that of FLSs [23]. There are not many relevant studies on MLSs, but MLSs are known to induce an inflammatory response during the development of RA [24]. MLS promotes RA development by secreting a variety of factors (oxygen species, nitric oxide intermediates, and matrix-degrading enzymes) [25] and can release a variety of cytokines to stimulate immune cells and FLSs [26].

2.3. T Cells

The association of human leukocyte antigen with RA patients reminds us of the important role of T cells in inducing immune responses. In general, type 1 T helper (Th1) and type 17 T helper (Th17) are thought to play a significant role in the pathogenesis of RA, but type 22 T helper cells (Th22) have also recently been shown to promote the progression of RA [27]. T cells were the primary immune infiltrating cells in RA patients. The important role of cytokines in the pathogenesis of RA has been extensively described [28]. Th1 cells and Th17 cells which can produce IL-1 and IL-17 have been proven to cause RA [29]. The role of IL-17 is highly valued in RA because it can induce a variety of cells (synovial cells, macrophages, etc.) to produce a large number of inflammatory factors (IL-1β, IL-6, TNFα), and it plays an important role in the production of chemokines (CXCL1, CXCL2, CXCL8, CCL2, CCL7) [30]. Treg cells proliferate in an inflammatory environment. In the joints and synovium of RA patients, the capacity of Treg cells is reduced. Forkhead box p3 (Foxp3), as an upstream regulator of Treg cells, can attenuate the inhibitory function of Treg cells under the induction of IL-1β and IL-6 [31,32]. Overall, T cells in RA (Th1, Th17, Treg, and Th22) have a complex interaction that leads to increased inflammation [27].

2.4. B Cells

The autoantibody production, antigen presentation, and cytokine secretion of B cells affect the progression of RA. There are various subsets of B cells, but data suggest that double negative (CD27IgD) and class-switched memory (CD27+IgD) are the key to RA pathogenesis [33]. This may be due to the fact that CD27+IgD is more likely to express RANKL, which induces RA, and in CD27IgD, miR-155 level increased, which is required for the production of autoantibodies by B cells [34,35]. CD21−/low CD27 IgD B cells have been detected in the peripheral blood and synovial fluid of RA patients, and the cells secrete RANKL even in the absence of stimulation, leading to bone destruction. Therefore, CD21−/low may be a potential target cell for the treatment of RA [36]. B cells act as antigen-presenting cells that can deliver antigens to CD4+ helper T cells, triggering an immune response. In RA, B cells play an increasingly important role in activating autoreactive T cells as inflammation worsens. In addition, in the synovial environment of RA patients, inflammatory factors and cytokines (TNF-α, IFN-γ, IL-6, IL-1β, IL-17, CCL20, RANKL, etc.) secreted by B cells promote bone destruction and inhibit bone formation. It also affects T cells and other cells in the synovial membrane, speeding up the progression of the inflammation [37,38,39]. However, the role of Regulatory B (Breg) cells in RA is opposite to that of B cells, which have an immunosuppressive effect and produce anti-inflammatory factors [40]. In addition, B cells are activated and produce autoantibodies in RA.

3. Signaling Pathways in RA

The development process of RA is associated with a variety of pathways, such as the NF-κB, MAPK, Wnt, JAK-STAT, PI3K-Akt, and so on. We have recently made a detailed review of the association between pathways in RA [15]. These pathways will be briefly introduced below and summarized in Figure 3. The main aim is to find new targets for the treatment of RA through these pathways.
NF-κB is one of the most common pathways in RA and is often associated with inflammatory responses. NF-κB is a dimer transcription factor associated with the production of several pro-inflammatory factors (TNF-α, IL-1β, and IL-6) in RA [41]. The production of ROS indirectly induces the activation of IkB kinase (IkK), resulting in the degradation of IkB and the induction of inflammation in the nucleus of NF-κB. An obvious sign of RA occurrence is the activation of NF-κB. NF-κB can cause synovial inflammation by prompting macrophages to release inflammatory factors. Many studies have found that the inhibition of the NF-κB signaling pathway can relieve inflammatory symptoms in rats with arthritis. AP1 can regulate cytokines, affect MMPs, NF-κB production, and synovial hyperplasia, and its expression is related to the severity of the disease [42]. ROS can activate the AP1 pathway, leading to the production of pro-inflammatory factors. In IL-18-induced FLS inflammation, Roflumilast can relieve the degree of oxidative stress in cells, reduce the expression of inflammatory factors, and inhibit the activation of NF-κB and AP1 [43]. IL-1β induces ROS production and AP1 activation in RASFs. cPLA2 has been shown to contribute to the development of arthritis. Activated AP1 binds to the promoter region of cPLA2, resulting in increased cPLA2 expression. While ho-1, which has antioxidant effects, attenuates AP-1 and cPLA2 expression, the antioxidant HO-1 can inhibit both AP1 and cPLA2 [43].
The MAPK family includes p38 kinases, ERK, and JNK, which play an important role in inflammation. Phosphorylated forms of MAPKs can induce the transcription and activation of cytokines in RA. ERK is involved in pannus formation, JNK regulates collagenase production in SFs, and p38 regulates MMP3 expression in fibroblasts [44]. MAPKs are closely related to the proliferation and apoptosis of FLS in RA and can promote the proliferation of FLS by activating the JAK-STAT pathway. Hydrogen, as a molecule found in nature, has been shown to neutralize OH and ONOO-. In CIA-induced arthritis, hydrogen was shown to reduce joint swelling in mice. In in vitro experiments, hydrogen can inhibit the proliferation of FLS and has an antioxidant effect, which is accompanied by the activation and inhibition of MAPK signaling pathways [45]. Sonic Hedgehog (SHH) signaling pathway is related to cell proliferation and migration, and its expression is increased in RA synovial tissues and RAFLS [46,47]. Further study found that SHH could activate an intracellular MAPK/ERK cascade [48].
The Wnt pathway plays a role in a variety of cancers and immune diseases and is related to various physiological processes, including cell proliferation and migration [49,50]. It mainly contains two pathways: β-catenin-dependent and β-catenin-independent [51]. It is well known that the Wnt pathway is highly expressed in synovial tissues of RA patients [52]. In addition, the Wnt//β-catenin pathway can promote the generation of bone cells and plays a positive role in human bone development [53]. In RA, the Wnt inhibitor DKK1 can promote pannus formation [49]. Additionally, a variety of miRNAs have been confirmed to regulate the pathogenesis of RA through the Wnt pathway. A recent study by our group shows that the transcription factor E2F1 can bind to the promoter region of Neuron navigator 2 (NAV2), activate the transcription of NAV2 [53], and regulate RA through the Wnt/β-catenin signaling pathway [53], and subsequent studies have found that NAV2 can regulate RA through SSH1L/Cofilin-1 signaling pathway [54]. This provides a new pathway target for the treatment of RA.
The Janus kinases (JAK) family includes Jak1, Jak2, Jak3, and tyrosine kinase (TYK) 2 [55]. STAT family includes STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6 and is involved in a variety of physiological processes in cells (proliferation, apoptosis, etc.) [56]. The JAK/stat pathway is associated with a variety of cytokines, and when the associated cytokines bind to their receptors, it means that the JAK/STAT signaling pathway is about to activate [56]. When activated, members of the JAK family phosphorylate some tyrosine residues to dock with signaling molecules [55]. STAT members are recruited to the JAK dimer and phosphorylated, separated from the receptor, translocated to the nucleus, and regulated the transcription of genes involved in cell physiological processes [57,58]. Additional good news for JAK inhibitors comes from a recent clinical study that found relief of pain in RA patients with JNK inhibitors [59].
Phosphatidylinositol 3 kinase (PI3K)-Akt plays an important role in cell physiology, regulating cell differentiation, proliferation, autophagy, and so on [60]. PI3K is a heterodimer composed of a p110 catalytic subunit and a p85 regulatory subunit [61]. PI3K can be activated by dimer conformation changes or by binding of Ras and p110. PI3K phosphorylates PIP2 to PIP3, thus binding with a variety of downstream effector molecules and affecting the physiological process of cells [62]. Protein kinase B (AKT) is an important downstream target in the PI3K pathway. After PIP3 recruits PDK1 and AKT to the cell membrane, PDK1 phosphorylates AKT1 to further activate downstream substrates (protein kinases, E3 ubiquitin ligases, regulators of small G proteins, metabolic enzymes, and transcription factors) [63]. In RA, the PI3K-AKT signaling pathway is associated with the expression of a variety of cytokines and participates in various pathological processes, such as pannus formation, cell proliferation, and migration [64]. Abnormal activation of PI3K/AKT pathway also stimulates the expression of HIF-1α and promotes angiogenesis. HIF-1 is involved in various cellular processes, such as glycolysis, cell growth, migration, and apoptosis. HIF-1 activates inflammatory cells and participates in pannus formation. In RA, ubiquitination is inhibited, leading to the accumulation of HIF-1α, which binds to the HRE promoter and regulates the expression of VEGF, ultimately leading to angiogenesis [65].
Based on different cellular mechanisms and targeting pathways in RA, a variety of target inhibitors have been applied in clinical practice (Table 1), but most of them have adverse reactions, even so, JAK inhibitors are still the pinup targets for RA drug development, and some new JAK inhibitors are in clinical trials. Meanwhile, many studies have focused on the formulation of new drugs that can alleviate RA. The extraction of active ingredients from plants has been a research hotspot in recent years, and this paper will summarize recent findings in Table 2.

4. Epigenetic Regulation in RA

The word ‘epigenetics’ was first used to describe heritable changes in gene function that occur without a change in the genetic coding. Nowadays, the word is used to characterize chromosomal structural changes that do not entail nucleotide sequence changes, regardless of whether the changes are strictly heritable [105]. DNA methylation and covalent histone modifications are examples of epigenetic changes. Histone modifications include (de)acetylation, (de)methylation, ubiquitination, and sumoylation. These changes control how accessible DNA is to transcription factors. An increasing amount of epigenetic enzymes are discovered to act as ‘writers’ or ‘erasers’ in the modifications of DNA and histones by attaching or removing certain functional groups, resulting in the regulation of gene expression. In addition, some proteins (‘readers’) that are able to recognize epigenetically modified sites also act as key players in the regulation of gene expression. We describe a schematic of epigenetics in Figure 4.
Epigenetics regulate the different levels of DNA and chromatin that accumulate in the nucleus of eukaryotic cells. DNA regulatory regions (such as promoters and enhancers) influence gene transcription by regulating the expression of transcription factors (TF), which is related to the state of chromatin structure. Euchromatin promotes the interaction between DNA-binding proteins and TF and increases gene transcription activity, while heterochromatin does the opposite [106]. Epigenetic modification mainly includes histone modification, DNA methylation, and non-coding RNA modification. Next, this paper will introduce the impact of RA from these three aspects. Potential targets are summarized in Table 3.

4.1. Histone Modification

Recent research has shown that histone modifications may be involved in the development and progression of RA. Histones are proteins that help package and organize DNA in the nucleus of cells, and their modification can affect gene expression and protein production. Histones can be modified by methylation, acetylation, phosphorylation, and poly ADP-ribosylation. Histone methylation, which occurs mainly in proteins containing SET domains, is the most stable modification method, and the modification sites are lysine and arginine. Studies have found that specific histone modifications, such as histone acetylation and methylation, are associated with increased inflammation and joint destruction in RA.
Our team found that in PDGF-induced FLS, the expression of Jumonji C family of histone demethylases (JMJD3) was increased through the Akt signaling pathway, and the proliferation and migration ability of FLS was weakened after inhibition or silence of JMJD3, and the symptoms of DBA/1 mice by collagen-induced arthritis (CIA) were alleviated [107]. At the same time, we also found that CSE/H2S can reduce the expression of JMJD3 by inhibiting transcription factor Sp-1 and alleviating arthritis [108]. GATA4 plays an important role in regulating cardiac function. Our team found that GATA4 was up-regulated in MH7A stimulated by IL-1β and could regulate the proliferation and migration of endothelial cells and blood vessel formation through the MAPK signaling pathway [109]. This conclusion may provide ideas for RA treatment.
Some studies have found that the downregulation of SIRT3 can promote the increase in ROS levels in mice and regulate oxidative stress [110]. After the reduction of the SIRT3 level, mitochondrial oxidative stress was aggravated, and osteoarthritis was induced in mice [111]. The role of SIRT4 in RA has not been studied, but SIRT4 can affect osteoarthritis through a variety of inflammatory factors, such as TNFα and IL-6, and the effect of SIRT4 on oxidative stress has also been proven the increased expression of SIRT4 can up-regulate SOD1, SOD2, and catalase [112].
Recent research has suggested that histone deacetylases (HDACs) may be involved in the development and progression of RA. Studies have shown that HDACs are increased in synovial tissues of patients with RA and that inhibiting HDACs can reduce inflammation responses [113,114]. HDAC1 is a key player in arthritis mediated by the T cell [115]. Research carried out by our group has proven that HDAC6 increased in the synovium tissues of adjuvant-induced arthritic rats [116].
HDAC inhibitors have been shown to decrease the production of pro-inflammatory cytokines, such as TNF-α and IL-6, and to increase the activity of regulatory T cells, which can help to suppress the immune response in RA [117,118]. Several HDAC inhibitors, such as HDAC6 inhibitors CKD-506 [119] and CKD-L [120] and HDAC6 inhibitor NK-HDAC1 [121], are currently being investigated as potential treatments for RA in pre-clinical studies. Additionally, further research is needed to understand further the role of HDACs in RA pathogenesis and the potential benefits of HDAC inhibitors in the treatment of RA.
The hydrogen sulfide (H2S) donor S-propargyl cysteine (SPRC/ZYZ-802) developed by our team alleviates the inflammatory response and inhibits HDAC6 and JMJD3 in RA models [108,116]; it might be a potential agent for RA treatment in the future.

4.2. DNA Methylation

Compared with other epigenetic modifications, DNA methylation has the advantage of stabilizing inheritance [122]. DNA methylation is the binding of a methyl group at the cytosine 5 carbon site of CpG dinucleotide in the genome through the action of DNA methyltransferase without changing the DNA sequence. CpG islands are the most common regions where DNA methylation occurs. DNA methylation is induced by DNMTs DNA methyltransferases (DNMTs), the most important of which is DNMT1. There are differences in DNA methylation between Rheumatoid arthritis synovial fibroblasts (RASFs) and Osteoarthrosis synovial fibroblasts (OASFs). RASF is hypomethylated and DNMT1 expression is low [123]. As early as 1986, DNA methyltransferase inhibitor 5-azacytidine was found to induce autoimmune-related symptoms [124]. Studies have shown that in PBMCs of RA patients, CpG methylation in the promoter region of IL-6 can regulate the pathogenesis of RA [125]. CD4+ T cells of RA patients show hypomethylation, and CD4+CD8+ T cells have been found to have significantly hypomethylated IFNγ promoters, which can produce higher levels of IFNγ [126,127]. In a recent study, the DNA methylation of T cells in RA patients was significantly different between synovium and blood, which provided a strong support for the discovery of new therapeutic targets by targeting T cells [128]. In general, hypermethylation of gene promoter regions usually prevents transcription factor binding, leading to transcriptional repression. The hypomethylation of genes is associated with increased gene expression, and a study found that DNA methylation levels of the Peptidylarginine deiminase type4 (PADI4) were significantly lower in RA patients compared to healthy controls. PADI4 is involved in the citrullination of proteins, which is thought to be a key process and a non-MHC genetic risk factor in the development of RA. The hypo-methylation of the PADI4 gene promoter region may lead to the increased expression of the gene, which in turn could contribute to the development and progression of RA [129].
In the PBMC isolated from RA patients, and the DNMTs inhibitor 5′-AzaC increased the expression of anti-inflammatory cytokine IL-10 [130]. Therefore, DNMTs inhibitors have been proposed as potential RA drugs. However, the relationship between DNA methylation and the occurrence and development of RA is complex and needs further research and exploration.

4.3. Micro RNA

MicroRNAs occupy an important position in the modification of non-coding RNAs, and their special property is that they are genome encoded. There are differences in miRNA expression in RA patients and various cells associated with RA. For example, in peripheral blood monocytes from patients with RA, a variety of miRNAs were detected to be upregulated (miR-16, miR-103a, miR-132, miR-145, miR-146a, and miR-155), and also down-regulated (miR-21, miR-125b, and miR548a), these may be related to T cell homeostasis [131]. The abnormal expression of miR-17 and miR-146a were found to be related to the imbalance of Treg cells in peripheral blood T cells [132]. In RASFs, a large number of miRNAs are involved in the regulation of pathways (Wnt, NF-κB, JAK/STAT). There are few studies on miRNA regulation by oxidative stress in RA. Studies have shown that particulate-matter-induced RA can produce ROS and activate the MAPK signaling pathway, thus downregulating miR137 [133]. Therefore, it can be speculated whether targeting miR137 can mediate ROS, thus providing new ideas for the treatment of RA. Mechanisms of some miRNA regulating RA have been listed in Table 3.
Targeting miRNAs has also emerged as a potential therapeutic strategy for RA. For example, inhibition of miR-155 has been shown to upregulate FOXO3 and reduce inflammation and proliferation of FLS [134]. Similarly, targeting miR-146a has been shown to reduce the invasion and migration of FLSs via the miR-146a/GATA6 axis [135,136].
Changes in miRNA expression levels have been proposed as potential biomarkers for predicting the therapeutic response to RA treatments. Studies have shown that drug treatment can modulate the expression of miRNAs in clinical patients with rheumatoid arthritis (RA). For example, methotrexate (MTX), a commonly used disease-modifying anti-rheumatic drug (DMARD), miR-16, miR-132, miR-22, miR-155, and miR-146a described as MTX-treatment response biomarkers [137]. MTX has been shown to cause increased marrow adiposity and reduced trabecular bone volume; it has been reported that MTX upregulates miR-6315 after treatment, which might be a vital cause of bone and marrow fat formation [138]. Similarly, statistically significant differences between miR-26b, miR-29, miR-451, and miR-522 were observed after olokizumab therapy [139].
Further studies and patients are needed to fully elucidate the role of miRNAs in RA treatment and to develop miRNA-based therapies for this disease.
Table 3. Potential epigenetic targets in RA.
Table 3. Potential epigenetic targets in RA.
TargetsEffectsReferences
JMJD3Regulating the proliferation and migration ability of FLS through the AKT signaling pathway
Regulating symptoms of CIA mice
[107,108]
GATA4Mediating the proliferation and migration ability of FLS through MAPK signaling pathway
Promoting angiogenesis
[109]
SIRT3Regulating of ROS levels in miceInducing osteoarthritis in mice
Inducing osteoarthritis in mice
[110,111]
SIRT4Regulating the expression of SOD1, SOD2, and catalase
Targeting osteoarthritis with inflammatory factors
[112]
HDAC1Knockdown relieves joint swelling and synovial inflammation in arthritic mice[115]
HDAC6Increase in the synovium tissues of adjuvant-induced arthritic rats[116]
miR-137Decreasing expression is due to ROS activation of MAPK in AIA rats[133]
miR-155Upregulating FOXO3 and reducing inflammation and proliferation of FLS[134]
miR-146aTargeting miR-146a reduces the invasion and migration of FLSs via the miR-146a/GATA6 axis[135]
NAV2Mediating the proliferation and migration of MH7A via Wnt/β-catenin signaling pathway[140,141]
miR-203Promoting the production of MMP1 and IL-6 induces RA[142]
miR-19Inducing inflammation through regulation of TLR2, IL-6, and MMP3 in FLS stimulated by TNFα
Having an anti-inflammatory effect in FLS stimulated by LPS by regulating IL-6 and IL-1β
[143,144,145]
miR-10aMediating the production of inflammatory factors through NF-κB in RA-FLS[146]
miR-124aReducing the proliferation of synovial cells
Relieving cartilage or bone destruction and the symptoms in AIA rats
[147]
miR-152Upregulating the expression of SFRP4 (negative regulator of Wnt signaling pathway) by targeting DNMT1, and reducing the proliferation of FLSs by inhibiting the activation of Wnt pathway[148]
miR-375Decreasing the expression of FZD8 (a member of Wnt pathway), inhibiting the expression of IL-6 and IL-8, and relieving the symptoms of AIA rats[149]

5. Conclusions and Perspective

RA, as a systemic inflammatory disease, is not fatal, but it harms patients’ physical and mental health. At present, there are a variety of therapeutic drugs and methods in clinical practice, but some of them have side effects, and about 50% of patients are not sensitive to the existing therapeutic targets, so it is difficult to achieve the ideal therapeutic effect. There are many factors in the development of RA, involving a variety of cells, cytokines, and signaling pathways. So, there is a great possibility of finding a better target.
In recent years, epigenetics has developed rapidly, providing a lot of ideas for finding new targets for RA treatment. With the rapid development of bioinformatics, the application of multiomics analysis has become more extensive and we can find potential new targets more efficiently. In recent decades, the rise in biological targeted drugs has provided multiple options for the treatment of RA. Although there are still many targets that have not been applied clinically, we believe that in the future, targeted drugs will provide better treatment, less cost, faster therapeutic effects, and fewer side effects for a large number of RA patients.
Our research group proved for the first time that Neuron Navigator 2 (NAV2) promotes the inflammatory response in RA, and we noticed that the transcription factor E2F1 can bind to the NAV2 promoter region to activate the transcription and expression of NAV2 [54,140,141]. Previous studies have demonstrated that NAV2 plays a key role in the development of the mammalian nervous system [150], and we thus propose that NAV2 may affect inflammatory pain during RA disease progression through neuronal navigation [151], which may provide a basis for alleviating the pain of RA patients. At the same time, we also found that the level of GATA4 increased in the synovium of RA patients. The transcription factor GATA4 is an important regulator of cardiac-differentiation-specific gene expression. We demonstrated for the first time that GATA4 plays a role in regulating VEGF in RA FLS, inducing cell migration/proliferation, and plays an important role in angiogenesis [109]. Our study also showed that the protein level of histone deacetylase HDAC6 was increased in the synovial tissue of arthritic rats [116]. In an animal model of RA, HDAC inhibitors could improve synovial inflammation and joint swelling and relieve RA symptoms [117,118]. At the same time, we also found that the expression of histone demethylase JMJD3 was increased in PDGF-induced FLS, after inhibition or silence of JMJD3, the migration, and proliferation of FLS receded [107].
The hydrogen sulfide (H2S) donor S-propargyl cysteine (SPRC/ZYZ-802) independently developed by our team alleviates the inflammatory response through the Nrf2-ARE signaling pathway [152], or the HDAC6/MyD88/NF-κB signaling pathway inhibited the expression of HDAC6 [116]. Meanwhile, we also found that CSE/H2S can reduce the expression of JMJD3 and reduce arthritis. These findings proposed that SPRC may be used as a potential drug for the treatment of rheumatoid arthritis. We have developed two types of H2S sustained-release donors [153,154] that solve the problem of the too-fast release of H2S in traditional formulations. ZYZ-802 is currently being submitted to the CFDA and FDA for clinical trial applications [15].
In conclusion, the study of epigenetics has opened up new avenues for the development of therapeutics for rheumatoid arthritis (RA). Epigenetic modifications have been shown to play a crucial role in the development and progression of RA, and drugs that target these modifications have shown promising results in animal models of the disease.
Furthermore, epigenetic changes are reversible, which means that epigenetic therapies have the potential to not only treat the symptoms of RA but also modify the disease course and prevent its progression. This contrasts to conventional RA therapies, which are often palliative and do not modify the underlying disease process.
However, there are also challenges that need to be addressed before epigenetic therapies can be widely used in the clinic. One of the major challenges is the need to identify specific epigenetic modifications that are involved in the pathogenesis of RA and to develop drugs that target these modifications with high specificity and efficacy.
HDAC inhibitors and DNA methyltransferase inhibitors are two types of epigenetic drugs that have been studied extensively for the treatment of RA. While these drugs have shown some efficacy, there is still a need for further research to determine their safety and effectiveness in humans.
MicroRNAs have also emerged as potential targets for RA therapy. These small non-coding RNAs can regulate gene expression and have been shown to be involved in the pathogenesis of RA. MicroRNAs have gained significant attention as potential therapeutic targets and diagnostic biomarkers for RA.
In addition to the potential use of epigenetic drugs, there is also growing interest in the role of the microbiome in the development of RA. Studies have suggested that changes in the gut microbiome could play a role in the development of RA, and targeting the microbiome with probiotics or fecal microbiota transplantation could be a potential therapeutic strategy for the disease [155,156].
Another area of research is the use of gene editing technologies, such as CRISPR-Cas9 [157,158,159], to modify the expression of specific genes involved in the pathogenesis of RA. While this approach is still in its early stages, it holds promise for the development of highly targeted and personalized therapies for RA.
Overall, although there is currently no cure for RA, effective treatment options are available that can help manage the symptoms and improve the quality of life for people with this condition. Drug development of new targets is expected to provide more options for patients who do not respond to current treatments. Ongoing research into the underlying causes of RA may lead to new and more effective treatments in the future. The study of epigenetics has provided new insights into the complex pathogenesis of RA and has opened up new possibilities for the development of targeted therapies. While more research is needed, the use of potential therapeutics for the treatment of RA holds great promise for improving the lives of millions of people affected by this chronic autoimmune disorder.

Author Contributions

Writing—Original draft preparation, M.Z. (Menglin Zhu).; Investigation, Q.D.; Project administration, Z.L. (Zhongxiao Lin), R.F.; Writing—Reviewing and Editing, F.Z., Z.L. (Zhaoyi Li), M.Z. (Mei Zhang), Y.Z.; Funding acquisition, Y.Z. All authors have read and agreed to the published version of the manuscript.

Funding

Macau Science and Technology Development fund (FDCT (0011/2020/A1, 0012/2021/AMJ, 003/2022/ALC, 0092/2022/A2)). The National Natural Science Foundation of China (Nos. 81973320).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Vesperini, V.; Lukas, C.; Fautrel, B.; Le Loet, X.; Rincheval, N.; Combe, B. Association of Tobacco Exposure and Reduction of Radiographic Progression in Early Rheumatoid Arthritis: Results from a French Multicenter Cohort. Arthritis Care Res. 2013, 65, 1899–1906. [Google Scholar] [CrossRef] [PubMed]
  2. Minihane, A.M.; Vinoy, S.; Russell, W.R.; Baka, A.; Roche, H.M.; Tuohy, K.M.; Teeling, J.L.; Blaak, E.E.; Fenech, M.; Vauzour, D.; et al. Low-grade inflammation, diet composition and health: Current research evidence and its translation. Br. J. Nutr. 2015, 114, 999–1012. [Google Scholar] [CrossRef]
  3. Qin, B.; Yang, M.; Fu, H.; Ma, N.; Wei, T.; Tang, Q.; Hu, Z.; Liang, Y.; Yang, Z.; Zhong, R. Body mass index and the risk of rheumatoid arthritis: A systematic review and dose-response meta-analysis. Arthritis Res. Ther. 2015, 17, 86. [Google Scholar] [CrossRef]
  4. Gioia, C.; Lucchino, B.; Tarsitano, M.G.; Iannuccelli, C.; Di Franco, M. Dietary Habits and Nutrition in Rheumatoid Arthritis: Can Diet Influence Disease Development and Clinical Manifestations? Nutrients 2020, 12, 1456. [Google Scholar] [CrossRef]
  5. Smith, M.D. The normal synovium. Open Rheumatol. J. 2011, 5, 100–106. [Google Scholar] [CrossRef] [PubMed]
  6. Walker, M.E.; Souza, P.R.; Colas, R.A.; Dalli, J. 13-Series resolvins mediate the leukocyte-platelet actions of atorvastatin and pravastatin in inflammatory arthritis. FASEB J. 2017, 31, 3636–3648. [Google Scholar] [CrossRef]
  7. Misharin, A.V.; Cuda, C.M.; Saber, R.; Turner, J.D.; Gierut, A.K.; Haines, G.K., 3rd; Berdnikovs, S.; Filer, A.; Clark, A.R.; Buckley, C.D.; et al. Nonclassical Ly6C(-) monocytes drive the development of inflammatory arthritis in mice. Cell Rep. 2014, 9, 591–604. [Google Scholar] [CrossRef]
  8. Thieblemont, N.; Wright, H.L.; Edwards, S.W.; Witko-Sarsat, V. Human neutrophils in auto-immunity. Semin. Immunol. 2016, 28, 159–173. [Google Scholar] [CrossRef] [PubMed]
  9. Filer, A.; Pitzalis, C.; Buckley, C.D. Targeting the stromal microenvironment in chronic inflammation. Curr. Opin. Pharmacol. 2006, 6, 393–400. [Google Scholar] [CrossRef]
  10. Wojcik, P.; Gegotek, A.; Zarkovic, N.; Skrzydlewska, E. Oxidative Stress and Lipid Mediators Modulate Immune Cell Functions in Autoimmune Diseases. Int. J. Mol. Sci. 2021, 22, 723. [Google Scholar] [CrossRef]
  11. Zhang, Q.; Peng, W.; Wei, S.; Wei, D.; Li, R.; Liu, J.; Peng, L.; Yang, S.; Gao, Y.; Wu, C.; et al. Guizhi-Shaoyao-Zhimu decoction possesses anti-arthritic effects on type II collagen-induced arthritis in rats via suppression of inflammatory reactions, inhibition of invasion & migration and induction of apoptosis in synovial fibroblasts. Biomed. Pharmacother. 2019, 118, 109367. [Google Scholar] [CrossRef]
  12. Zhang, Q.; Liu, J.; Zhang, M.; Wei, S.; Li, R.; Gao, Y.; Peng, W.; Wu, C. Apoptosis Induction of Fibroblast-Like Synoviocytes Is an Important Molecular-Mechanism for Herbal Medicine along with its Active Components in Treating Rheumatoid Arthritis. Biomolecules 2019, 9, 795. [Google Scholar] [CrossRef] [PubMed]
  13. Bolam, A.; Manandhar, D.S.; Shrestha, P.; Ellis, M.; Costello, A.M. The effects of postnatal health education for mothers on infant care and family planning practices in Nepal: A randomised controlled trial. BMJ 1998, 316, 805–811. [Google Scholar] [CrossRef] [PubMed]
  14. Abbasi, M.; Mousavi, M.J.; Jamalzehi, S.; Alimohammadi, R.; Bezvan, M.H.; Mohammadi, H.; Aslani, S. Strategies toward rheumatoid arthritis therapy; the old and the new. J. Cell Physiol. 2019, 234, 10018–10031. [Google Scholar] [CrossRef]
  15. Ding, Q.; Hu, W.; Wang, R.; Yang, Q.; Zhu, M.; Li, M.; Cai, J.; Rose, P.; Mao, J.; Zhu, Y.Z. Signaling pathways in rheumatoid arthritis: Implications for targeted therapy. Signal Transduct. Target. Ther. 2023, 8, 68. [Google Scholar] [CrossRef]
  16. Arnett, F.C.; Edworthy, S.M.; Bloch, D.A.; McShane, D.J.; Fries, J.F.; Cooper, N.S.; Healey, L.A.; Kaplan, S.R.; Liang, M.H.; Luthra, H.S.; et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988, 31, 315–324. [Google Scholar] [CrossRef]
  17. Filer, A. The fibroblast as a therapeutic target in rheumatoid arthritis. Curr. Opin. Pharmacol. 2013, 13, 413–419. [Google Scholar] [CrossRef] [PubMed]
  18. Nygaard, G.; Firestein, G.S. Restoring synovial homeostasis in rheumatoid arthritis by targeting fibroblast-like synoviocytes. Nat. Rev. Rheumatol. 2020, 16, 316–333. [Google Scholar] [CrossRef]
  19. Shin, Y.J.; Han, S.H.; Kim, D.S.; Lee, G.H.; Yoo, W.H.; Kang, Y.M.; Choi, J.Y.; Lee, Y.C.; Park, S.J.; Jeong, S.K.; et al. Autophagy induction and CHOP under-expression promotes survival of fibroblasts from rheumatoid arthritis patients under endoplasmic reticulum stress. Arthritis Res. Ther. 2010, 12, R19. [Google Scholar] [CrossRef]
  20. Hua, S.; Dias, T.H. Hypoxia-Inducible Factor (HIF) as a Target for Novel Therapies in Rheumatoid Arthritis. Front. Pharmacol. 2016, 7, 184. [Google Scholar] [CrossRef]
  21. Tsaltskan, V.; Firestein, G.S. Targeting fibroblast-like synoviocytes in rheumatoid arthritis. Curr. Opin. Pharmacol. 2022, 67, 102304. [Google Scholar] [CrossRef]
  22. Wu, Z.; Ma, D.; Yang, H.; Gao, J.; Zhang, G.; Xu, K.; Zhang, L. Fibroblast-like synoviocytes in rheumatoid arthritis: Surface markers and phenotypes. Int. Immunopharmacol. 2021, 93, 107392. [Google Scholar] [CrossRef]
  23. Bartok, B.; Firestein, G.S. Fibroblast-like synoviocytes: Key effector cells in rheumatoid arthritis. Immunol. Rev. 2010, 233, 233–255. [Google Scholar] [CrossRef] [PubMed]
  24. Tu, J.; Hong, W.; Zhang, P.; Wang, X.; Korner, H.; Wei, W. Ontology and Function of Fibroblast-Like and Macrophage-Like Synoviocytes: How Do They Talk to Each Other and Can They Be Targeted for Rheumatoid Arthritis Therapy? Front. Immunol. 2018, 9, 1467. [Google Scholar] [CrossRef] [PubMed]
  25. McInnes, I.B.; Schett, G. Cytokines in the pathogenesis of rheumatoid arthritis. Nat. Rev. Immunol. 2007, 7, 429–442. [Google Scholar] [CrossRef]
  26. Burger, D.; Dayer, J.M. The role of human T-lymphocyte-monocyte contact in inflammation and tissue destruction. Arthritis Res. 2002, 4 (Suppl. 3), S169–S176. [Google Scholar] [CrossRef]
  27. Mellado, M.; Martinez-Munoz, L.; Cascio, G.; Lucas, P.; Pablos, J.L.; Rodriguez-Frade, J.M. T Cell Migration in Rheumatoid Arthritis. Front. Immunol. 2015, 6, 384. [Google Scholar] [CrossRef] [PubMed]
  28. Kondo, N.; Kuroda, T.; Kobayashi, D. Cytokine Networks in the Pathogenesis of Rheumatoid Arthritis. Int. J. Mol. Sci. 2021, 22, 10922. [Google Scholar] [CrossRef]
  29. Jang, S.; Kwon, E.J.; Lee, J.J. Rheumatoid Arthritis: Pathogenic Roles of Diverse Immune Cells. Int. J. Mol. Sci. 2022, 23, 905. [Google Scholar] [CrossRef]
  30. Onishi, R.M.; Gaffen, S.L. Interleukin-17 and its target genes: Mechanisms of interleukin-17 function in disease. Immunology 2010, 129, 311–321. [Google Scholar] [CrossRef] [PubMed]
  31. Jiang, Q.; Yang, G.; Liu, Q.; Wang, S.; Cui, D. Function and Role of Regulatory T Cells in Rheumatoid Arthritis. Front. Immunol. 2021, 12, 626193. [Google Scholar] [CrossRef]
  32. Li, X.; Zheng, Y. Regulatory T cell identity: Formation and maintenance. Trends Immunol. 2015, 36, 344–353. [Google Scholar] [CrossRef] [PubMed]
  33. Floudas, A.; Canavan, M.; McGarry, T.; Mullan, R.; Nagpal, S.; Veale, D.J.; Fearon, U. ACPA Status Correlates with Differential Immune Profile in Patients with Rheumatoid Arthritis. Cells 2021, 10, 647. [Google Scholar] [CrossRef] [PubMed]
  34. Meednu, N.; Zhang, H.; Owen, T.; Sun, W.; Wang, V.; Cistrone, C.; Rangel-Moreno, J.; Xing, L.; Anolik, J.H. Production of RANKL by Memory B Cells: A Link Between B Cells and Bone Erosion in Rheumatoid Arthritis. Arthritis Rheumatol. 2016, 68, 805–816. [Google Scholar] [CrossRef]
  35. Filipowicz, W.; Bhattacharyya, S.N.; Sonenberg, N. Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight? Nat. Rev. Genet. 2008, 9, 102–114. [Google Scholar] [CrossRef] [PubMed]
  36. Thorarinsdottir, K.; Camponeschi, A.; Jonsson, C.; Granhagen Onnheim, K.; Nilsson, J.; Forslind, K.; Visentini, M.; Jacobsson, L.; Martensson, I.L.; Gjertsson, I. CD21(-/low) B cells associate with joint damage in rheumatoid arthritis patients. Scand. J. Immunol. 2019, 90, e12792. [Google Scholar] [CrossRef] [PubMed]
  37. Yanaba, K.; Bouaziz, J.D.; Haas, K.M.; Poe, J.C.; Fujimoto, M.; Tedder, T.F. A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses. Immunity 2008, 28, 639–650. [Google Scholar] [CrossRef]
  38. Gomez-Puerta, J.A.; Celis, R.; Hernandez, M.V.; Ruiz-Esquide, V.; Ramirez, J.; Haro, I.; Canete, J.D.; Sanmarti, R. Differences in synovial fluid cytokine levels but not in synovial tissue cell infiltrate between anti-citrullinated peptide/protein antibody-positive and -negative rheumatoid arthritis patients. Arthritis Res. Ther. 2013, 15, R182. [Google Scholar] [CrossRef]
  39. Sun, W.; Meednu, N.; Rosenberg, A.; Rangel-Moreno, J.; Wang, V.; Glanzman, J.; Owen, T.; Zhou, X.; Zhang, H.; Boyce, B.F.; et al. B cells inhibit bone formation in rheumatoid arthritis by suppressing osteoblast differentiation. Nat. Commun. 2018, 9, 5127. [Google Scholar] [CrossRef] [PubMed]
  40. Wu, F.; Gao, J.; Kang, J.; Wang, X.; Niu, Q.; Liu, J.; Zhang, L. B Cells in Rheumatoid Arthritis: Pathogenic Mechanisms and Treatment Prospects. Front. Immunol. 2021, 12, 750753. [Google Scholar] [CrossRef]
  41. Nishimura, R.; Hata, K.; Takahata, Y.; Murakami, T.; Nakamura, E.; Ohkawa, M.; Ruengsinpinya, L. Role of Signal Transduction Pathways and Transcription Factors in Cartilage and Joint Diseases. Int. J. Mol. Sci. 2020, 21, 1340. [Google Scholar] [CrossRef] [PubMed]
  42. Asahara, H.; Fujisawa, K.; Kobata, T.; Hasunuma, T.; Maeda, T.; Asanuma, M.; Ogawa, N.; Inoue, H.; Sumida, T.; Nishioka, K. Direct evidence of high DNA binding activity of transcription factor AP-1 in rheumatoid arthritis synovium. Arthritis Rheum. 1997, 40, 912–918. [Google Scholar] [CrossRef] [PubMed]
  43. Chi, P.L.; Chen, Y.W.; Hsiao, L.D.; Chen, Y.L.; Yang, C.M. Heme oxygenase 1 attenuates interleukin-1beta-induced cytosolic phospholipase A2 expression via a decrease in NADPH oxidase/reactive oxygen species/activator protein 1 activation in rheumatoid arthritis synovial fibroblasts. Arthritis Rheum. 2012, 64, 2114–2125. [Google Scholar] [CrossRef]
  44. Thalhamer, T.; McGrath, M.A.; Harnett, M.M. MAPKs and their relevance to arthritis and inflammation. Rheumatology 2008, 47, 409–414. [Google Scholar] [CrossRef] [PubMed]
  45. Meng, J.; Yu, P.; Jiang, H.; Yuan, T.; Liu, N.C.; Tong, J.; Chen, H.Y.; Bao, N.R.; Zhao, J.N. Molecular hydrogen decelerates rheumatoid arthritis progression through inhibition of oxidative stress. Am. J. Transl. Res. 2016, 8, 4472–4477. [Google Scholar]
  46. Wang, M.; Zhu, S.; Peng, W.; Li, Q.; Li, Z.; Luo, M.; Feng, X.; Lin, Z.; Huang, J. Sonic hedgehog signaling drives proliferation of synoviocytes in rheumatoid arthritis: A possible novel therapeutic target. J. Immunol. Res. 2014, 2014, 401903. [Google Scholar] [CrossRef]
  47. Skoda, A.M.; Simovic, D.; Karin, V.; Kardum, V.; Vranic, S.; Serman, L. The role of the Hedgehog signaling pathway in cancer: A comprehensive review. Bosn. J. Basic Med. Sci. 2018, 18, 8–20. [Google Scholar] [CrossRef]
  48. Liu, F.; Feng, X.X.; Zhu, S.L.; Huang, H.Y.; Chen, Y.D.; Pan, Y.F.; June, R.R.; Zheng, S.G.; Huang, J.L. Sonic Hedgehog Signaling Pathway Mediates Proliferation and Migration of Fibroblast-Like Synoviocytes in Rheumatoid Arthritis via MAPK/ERK Signaling Pathway. Front. Immunol. 2018, 9, 2847. [Google Scholar] [CrossRef]
  49. Cici, D.; Corrado, A.; Rotondo, C.; Cantatore, F.P. Wnt Signaling and Biological Therapy in Rheumatoid Arthritis and Spondyloarthritis. Int. J. Mol. Sci. 2019, 20, 5552. [Google Scholar] [CrossRef] [PubMed]
  50. Maruotti, N.; Corrado, A.; Neve, A.; Cantatore, F.P. Systemic effects of Wnt signaling. J. Cell. Physiol. 2013, 228, 1428–1432. [Google Scholar] [CrossRef]
  51. Sen, M. Wnt signalling in rheumatoid arthritis. Rheumatology 2005, 44, 708–713. [Google Scholar] [CrossRef]
  52. Sen, M.; Reifert, J.; Lauterbach, K.; Wolf, V.; Rubin, J.S.; Corr, M.; Carson, D.A. Regulation of fibronectin and metalloproteinase expression by Wnt signaling in rheumatoid arthritis synoviocytes. Arthritis Rheum. 2002, 46, 2867–2877. [Google Scholar] [CrossRef]
  53. Tao, S.S.; Cao, F.; Sam, N.B.; Li, H.M.; Feng, Y.T.; Ni, J.; Wang, P.; Li, X.M.; Pan, H.F. Dickkopf-1 as a promising therapeutic target for autoimmune diseases. Clin. Immunol. 2022, 245, 109156. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, R.; Cai, J.; Chen, K.; Zhu, M.; Li, Z.; Liu, H.; Liu, T.; Mao, J.; Ding, Q.; Zhu, Y.Z. STAT3-NAV2 axis as a new therapeutic target for rheumatoid arthritis via activating SSH1L/Cofilin-1 signaling pathway. Signal Transduct. Target. Ther. 2022, 7, 209. [Google Scholar] [CrossRef] [PubMed]
  55. Kiu, H.; Nicholson, S.E. Biology and significance of the JAK/STAT signalling pathways. Growth Factors 2012, 30, 88–106. [Google Scholar] [CrossRef] [PubMed]
  56. Baldini, C.; Moriconi, F.R.; Galimberti, S.; Libby, P.; De Caterina, R. The JAK-STAT pathway: An emerging target for cardiovascular disease in rheumatoid arthritis and myeloproliferative neoplasms. Eur. Heart J. 2021, 42, 4389–4400. [Google Scholar] [CrossRef]
  57. Villarino, A.V.; Kanno, Y.; O’Shea, J.J. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat. Immunol. 2017, 18, 374–384. [Google Scholar] [CrossRef]
  58. Delgoffe, G.M.; Vignali, D.A. STAT heterodimers in immunity: A mixed message or a unique signal? JAKSTAT 2013, 2, e23060. [Google Scholar] [CrossRef]
  59. Simon, L.S.; Taylor, P.C.; Choy, E.H.; Sebba, A.; Quebe, A.; Knopp, K.L.; Porreca, F. The Jak/STAT pathway: A focus on pain in rheumatoid arthritis. Semin. Arthritis Rheum. 2021, 51, 278–284. [Google Scholar] [CrossRef]
  60. Sun, K.; Luo, J.; Guo, J.; Yao, X.; Jing, X.; Guo, F. The PI3K/AKT/mTOR signaling pathway in osteoarthritis: A narrative review. Osteoarthr. Cartil. 2020, 28, 400–409. [Google Scholar] [CrossRef]
  61. Ito, Y.; Hart, J.R.; Vogt, P.K. Isoform-specific activities of the regulatory subunits of phosphatidylinositol 3-kinases—Potentially novel therapeutic targets. Expert Opin. Ther. Targets 2018, 22, 869–877. [Google Scholar] [CrossRef]
  62. Ersahin, T.; Tuncbag, N.; Cetin-Atalay, R. The PI3K/AKT/mTOR interactive pathway. Mol. Biosyst. 2015, 11, 1946–1954. [Google Scholar] [CrossRef]
  63. Manning, B.D.; Toker, A. AKT/PKB Signaling: Navigating the Network. Cell 2017, 169, 381–405. [Google Scholar] [CrossRef] [PubMed]
  64. Li, G.Q.; Zhang, Y.; Liu, D.; Qian, Y.Y.; Zhang, H.; Guo, S.Y.; Sunagawa, M.; Hisamitsu, T.; Liu, Y.Q. PI3 kinase/Akt/HIF-1alpha pathway is associated with hypoxia-induced epithelial-mesenchymal transition in fibroblast-like synoviocytes of rheumatoid arthritis. Mol. Cell. Biochem. 2013, 372, 221–231. [Google Scholar] [CrossRef] [PubMed]
  65. Park, S.Y.; Lee, S.W.; Kim, H.Y.; Lee, W.S.; Hong, K.W.; Kim, C.D. HMGB1 induces angiogenesis in rheumatoid arthritis via HIF-1alpha activation. Eur. J. Immunol. 2015, 45, 1216–1227. [Google Scholar] [CrossRef] [PubMed]
  66. Zhao, S.; Mysler, E.; Moots, R.J. Etanercept for the treatment of rheumatoid arthritis. Immunotherapy 2018, 10, 433–445. [Google Scholar] [CrossRef]
  67. Feldman, M.; Taylor, P.; Paleolog, E.; Brennan, F.M.; Maini, R.N. Anti-TNF alpha therapy is useful in rheumatoid arthritis and Crohn’s disease: Analysis of the mechanism of action predicts utility in other diseases. Transplant. Proc. 1998, 30, 4126–4127. [Google Scholar] [CrossRef]
  68. Gholami, A.; Azizpoor, J.; Aflaki, E.; Rezaee, M.; Keshavarz, K. Cost-Effectiveness Analysis of Biopharmaceuticals for Treating Rheumatoid Arthritis: Infliximab, Adalimumab, and Etanercept. BioMed Res. Int. 2021, 2021, 4450162. [Google Scholar] [CrossRef]
  69. Cvetkovic, R.S.; Scott, L.J. Adalimumab: A review of its use in adult patients with rheumatoid arthritis. BioDrugs 2006, 20, 293–311. [Google Scholar] [CrossRef]
  70. Zhao, S.; Chadwick, L.; Mysler, E.; Moots, R.J. Review of Biosimilar Trials and Data on Adalimumab in Rheumatoid Arthritis. Curr. Rheumatol. Rep. 2018, 20, 57. [Google Scholar] [CrossRef]
  71. Cohen, M.D.; Keystone, E.C. Intravenous golimumab in rheumatoid arthritis. Expert Rev. Clin. Immunol. 2014, 10, 823–830. [Google Scholar] [CrossRef]
  72. Markatseli, T.E.; Papagoras, C.; Nikoli, A.; Voulgari, P.V.; Drosos, A.A. Drosos. Certolizumab for rheumatoid arthritis. Clin. Exp. Rheumatol. 2014, 32, 415–423. [Google Scholar] [PubMed]
  73. Mok, C.C. Rituximab for the treatment of rheumatoid arthritis: An update. Drug Des. Dev. Ther. 2013, 8, 87–100. [Google Scholar] [CrossRef]
  74. Blair, H.A.; Deeks, E.D. Abatacept: A Review in Rheumatoid Arthritis. Drugs 2017, 77, 1221–1233. [Google Scholar] [CrossRef]
  75. Scott, L.J. Tocilizumab: A Review in Rheumatoid Arthritis. Drugs 2017, 77, 1865–1879. [Google Scholar] [CrossRef]
  76. Pelechas, E.; Voulgari, P.V.; Drosos, A.A. Sirukumab: A promising therapy for rheumatoid arthritis. Expert Opin. Biol. Ther. 2017, 17, 755–763. [Google Scholar] [CrossRef] [PubMed]
  77. Shaw, S.; Bourne, T.; Meier, C.; Carrington, B.; Gelinas, R.; Henry, A.; Popplewell, A.; Adams, R.; Baker, T.; Rapecki, S.; et al. Discovery and characterization of olokizumab: A humanized antibody targeting interleukin-6 and neutralizing gp130-signaling. MAbs 2014, 6, 774–782. [Google Scholar] [CrossRef] [PubMed]
  78. Serio, I.; Tovoli, F. Rheumatoid arthritis: New monoclonal antibodies. Drugs Today 2018, 54, 219–230. [Google Scholar] [CrossRef]
  79. Bauer, E.; Lucier, J.; Furst, D.E. Brodalumab -an IL-17RA monoclonal antibody for psoriasis and psoriatic arthritis. Expert Opin. Biol. Ther. 2015, 15, 883–893. [Google Scholar] [CrossRef]
  80. Dhillon, S. Tofacitinib: A Review in Rheumatoid Arthritis. Drugs 2017, 77, 1987–2001. [Google Scholar] [CrossRef]
  81. Baricitinib for rheumatoid arthritis. Aust. Prescr. 2019, 42, 34–35. [CrossRef]
  82. Serhal, L.; Edwards, C.J. Upadacitinib for the treatment of rheumatoid arthritis. Expert Rev. Clin. Immunol. 2019, 15, 13–25. [Google Scholar] [CrossRef] [PubMed]
  83. Tanaka, Y.; Kavanaugh, A.; Wicklund, J.; McInnes, I.B. Filgotinib, a novel JAK1-preferential inhibitor for the treatment of rheumatoid arthritis: An overview from clinical trials. Mod. Rheumatol. 2022, 32, 1–11. [Google Scholar] [CrossRef] [PubMed]
  84. Qiu, Q.; Feng, Q.; Tan, X.; Guo, M. JAK3-selective inhibitor peficitinib for the treatment of rheumatoid arthritis. Expert Rev. Clin. Pharmacol. 2019, 12, 547–554. [Google Scholar] [CrossRef] [PubMed]
  85. Zhao, Y.; Zhang, L.; Ding, Y.; Tao, X.; Ji, C.; Dong, X.; Lu, J.; Wu, L.; Wang, R.; Lu, Q.; et al. Efficacy and Safety of SHR0302, a Highly Selective Janus Kinase 1 Inhibitor, in Patients with Moderate to Severe Atopic Dermatitis: A Phase II Randomized Clinical Trial. Am. J. Clin. Dermatol. 2021, 22, 877–889. [Google Scholar] [CrossRef] [PubMed]
  86. Genovese, M.C.; van Vollenhoven, R.F.; Pacheco-Tena, C.; Zhang, Y.; Kinnman, N. VX-509 (Decernotinib), an Oral Selective JAK-3 Inhibitor, in Combination with Methotrexate in Patients With Rheumatoid Arthritis. Arthritis Rheumatol. 2016, 68, 46–55. [Google Scholar] [CrossRef]
  87. Peng, S.; Hu, C.; Liu, X.; Lei, L.; He, G.; Xiong, C.; Wu, W. Rhoifolin regulates oxidative stress and proinflammatory cytokine levels in Freund’s adjuvant-induced rheumatoid arthritis via inhibition of NF-kappaB. Braz. J. Med. Biol. Res. 2020, 53, e9489. [Google Scholar] [CrossRef]
  88. Puppala, E.R.; Jain, S.; Saha, P.; Rachamalla, M.; Np, S.; Yalamarthi, S.S.; Abubakar, M.; Chaudhary, A.; Chamundeswari, D.; Usn, M.; et al. Perillyl alcohol attenuates rheumatoid arthritis via regulating TLR4/NF-kappaB and Keap1/Nrf2 signaling pathways: A comprehensive study onin-vitro and in-vivo experimental models. Phytomedicine 2022, 97, 153926. [Google Scholar] [CrossRef]
  89. Wang, G.; Xie, X.; Yuan, L.; Qiu, J.; Duan, W.; Xu, B.; Chen, X. Resveratrol ameliorates rheumatoid arthritis via activation of SIRT1-Nrf2 signaling pathway. Biofactors 2020, 46, 441–453. [Google Scholar] [CrossRef]
  90. Zhang, Y.; Wang, G.; Wang, T.; Cao, W.; Zhang, L.; Chen, X. Nrf2-Keap1 pathway-mediated effects of resveratrol on oxidative stress and apoptosis in hydrogen peroxide-treated rheumatoid arthritis fibroblast-like synoviocytes. Ann. N. Y. Acad. Sci. 2019, 1457, 166–178. [Google Scholar] [CrossRef]
  91. Xia, Z.B.; Yuan, Y.J.; Zhang, Q.H.; Li, H.; Dai, J.L.; Min, J.K. Salvianolic Acid B Suppresses Inflammatory Mediator Levels by Downregulating NF-kappaB in a Rat Model of Rheumatoid Arthritis. Med. Sci. Monit. 2018, 24, 2524–2532. [Google Scholar] [CrossRef]
  92. Huang, Y.; Guo, L.; Chitti, R.; Sreeharsha, N.; Mishra, A.; Gubbiyappa, S.K.; Singh, Y. Wogonin ameliorate complete Freund’s adjuvant induced rheumatoid arthritis via targeting NF-kappaB/MAPK signaling pathway. Biofactors 2020, 46, 283–291. [Google Scholar] [CrossRef] [PubMed]
  93. Shen, Y.; Fan, X.; Qu, Y.; Tang, M.; Huang, Y.; Peng, Y.; Fu, Q. Magnoflorine attenuates inflammatory responses in RA by regulating the PI3K/Akt/NF-kappaB and Keap1-Nrf2/HO-1 signalling pathways in vivo and in vitro. Phytomedicine 2022, 104, 154339. [Google Scholar] [CrossRef] [PubMed]
  94. Cai, J.; Zhang, L.C.; Zhao, R.J.; Pu, L.M.; Chen, K.Y.; Nasim, A.A.; Leung, E.L.; Fan, X.X. Chelerythrine ameliorates rheumatoid arthritis by modulating the AMPK/mTOR/ULK-1 signaling pathway. Phytomedicine 2022, 104, 154140. [Google Scholar] [CrossRef] [PubMed]
  95. Bai, J.; Xie, N.; Hou, Y.; Chen, X.; Hu, Y.; Zhang, Y.; Meng, X.; Wang, X.; Tang, C. The enhanced mitochondrial dysfunction by cantleyoside confines inflammatory response and promotes apoptosis of human HFLS-RA cell line via AMPK/Sirt 1/NF-kappaB pathway activation. Biomed. Pharmacother. 2022, 149, 112847. [Google Scholar] [CrossRef]
  96. Wu, Z.M.; Xiang, Y.R.; Zhu, X.B.; Shi, X.D.; Chen, S.; Wan, X.; Guo, J. Icariin represses the inflammatory responses and survival of rheumatoid arthritis fibroblast-like synoviocytes by regulating the TRIB1/TLR2/NF-kB pathway. Int. Immunopharmacol. 2022, 110, 108991. [Google Scholar] [CrossRef]
  97. Wang, X.H.; Dai, C.; Wang, J.; Liu, R.; Li, L.; Yin, Z.S. Therapeutic effect of neohesperidin on TNF-alpha-stimulated human rheumatoid arthritis fibroblast-like synoviocytes. Chin. J. Nat. Med. 2021, 19, 741–749. [Google Scholar] [CrossRef] [PubMed]
  98. Ahmad, S.; Mittal, S.; Gulia, R.; Alam, K.; Saha, T.K.; Arif, Z.; Nafees, K.A.; Al-Shaghdali, K.; Ahmad, S. Therapeutic role of hesperidin in collagen-induced rheumatoid arthritis through antiglycation and antioxidant activities. Cell Biochem. Funct. 2022, 40, 473–480. [Google Scholar] [CrossRef] [PubMed]
  99. Jing, M.; Yang, J.; Zhang, L.; Liu, J.; Xu, S.; Wang, M.; Zhang, L.; Sun, Y.; Yan, W.; Hou, G.; et al. Celastrol inhibits rheumatoid arthritis through the ROS-NF-kappaB-NLRP3 inflammasome axis. Int. Immunopharmacol. 2021, 98, 107879. [Google Scholar] [CrossRef]
  100. Yang, J.; Liu, J.; Li, J.; Jing, M.; Zhang, L.; Sun, M.; Wang, Q.; Sun, H.; Hou, G.; Wang, C.; et al. Celastrol inhibits rheumatoid arthritis by inducing autophagy via inhibition of the PI3K/AKT/mTOR signaling pathway. Int. Immunopharmacol. 2022, 112, 109241. [Google Scholar] [CrossRef]
  101. Villa, T.; Kim, M.; Oh, S. Fangchinoline Has an Anti-Arthritic Effect in Two Animal Models and in IL-1beta-Stimulated Human FLS Cells. Biomol. Ther. 2020, 28, 414–422. [Google Scholar] [CrossRef]
  102. Liao, K.; Su, X.; Lei, K.; Liu, Z.; Lu, L.; Wu, Q.; Pan, H.; Huang, Q.; Zhao, Y.; Wang, M.; et al. Sinomenine protects bone from destruction to ameliorate arthritis via activating p62(Thr269/Ser272)-Keap1-Nrf2 feedback loop. Biomed. Pharmacother. 2021, 135, 111195. [Google Scholar] [CrossRef] [PubMed]
  103. Dong, X.; Zhang, Q.; Zeng, F.; Cai, M.; Ding, D. The protective effect of gentisic acid on rheumatoid arthritis via the RAF/ERK signaling pathway. J. Orthop. Surg. Res. 2022, 17, 109. [Google Scholar] [CrossRef]
  104. Xu, Z.; Shang, W.; Zhao, Z.; Zhang, B.; Liu, C.; Cai, H. Curcumin alleviates rheumatoid arthritis progression through the phosphatidylinositol 3-kinase/protein kinase B pathway: An in vitro and in vivo study. Bioengineered 2022, 13, 12899–12911. [Google Scholar] [CrossRef] [PubMed]
  105. Klein, K.; Gay, S. Epigenetics in rheumatoid arthritis. Curr. Opin. Rheumatol. 2015, 27, 76–82. [Google Scholar] [CrossRef] [PubMed]
  106. Nemtsova, M.V.; Zaletaev, D.V.; Bure, I.V.; Mikhaylenko, D.S.; Kuznetsova, E.B.; Alekseeva, E.A.; Beloukhova, M.I.; Deviatkin, A.A.; Lukashev, A.N.; Zamyatnin, A.A., Jr. Epigenetic Changes in the Pathogenesis of Rheumatoid Arthritis. Front. Genet. 2019, 10, 570. [Google Scholar] [CrossRef]
  107. Jia, W.; Wu, W.; Yang, D.; Xiao, C.; Su, Z.; Huang, Z.; Li, Z.; Qin, M.; Huang, M.; Liu, S.; et al. Histone demethylase JMJD3 regulates fibroblast-like synoviocyte-mediated proliferation and joint destruction in rheumatoid arthritis. FASEB J. 2018, 32, 4031–4042. [Google Scholar] [CrossRef]
  108. Wu, W.; Qin, M.; Jia, W.; Huang, Z.; Li, Z.; Yang, D.; Huang, M.; Xiao, C.; Long, F.; Mao, J.; et al. Cystathionine-γ-lyase ameliorates the histone demethylase JMJD3-mediated autoimmune response in rheumatoid arthritis. Cell. Mol. Immunol. 2019, 16, 694–705. [Google Scholar] [CrossRef] [PubMed]
  109. Jia, W.; Wu, W.; Yang, D.; Xiao, C.; Huang, M.; Long, F.; Su, Z.; Qin, M.; Liu, X.; Zhu, Y.Z. GATA4 regulates angiogenesis and persistence of inflammation in rheumatoid arthritis. Cell Death Dis. 2018, 9, 503. [Google Scholar] [CrossRef]
  110. Jing, E.; Emanuelli, B.; Hirschey, M.D.; Boucher, J.; Lee, K.Y.; Lombard, D.; Verdin, E.M.; Kahn, C.R. Sirtuin-3 (Sirt3) regulates skeletal muscle metabolism and insulin signaling via altered mitochondrial oxidation and reactive oxygen species production. Proc. Natl. Acad. Sci. USA 2011, 108, 14608–14613. [Google Scholar] [CrossRef]
  111. Tseng, A.H.; Shieh, S.S.; Wang, D.L. SIRT3 deacetylates FOXO3 to protect mitochondria against oxidative damage. Free. Radic. Biol. Med. 2013, 63, 222–234. [Google Scholar] [CrossRef] [PubMed]
  112. Lang, A.; Grether-Beck, S.; Singh, M.; Kuck, F.; Jakob, S.; Kefalas, A.; Altinoluk-Hambuchen, S.; Graffmann, N.; Schneider, M.; Lindecke, A.; et al. MicroRNA-15b regulates mitochondrial ROS production and the senescence-associated secretory phenotype through sirtuin 4/SIRT4. Aging 2016, 8, 484–505. [Google Scholar] [CrossRef] [PubMed]
  113. Angiolilli, C.; Grabiec, A.M.; Ferguson, B.S.; Ospelt, C.; Malvar Fernandez, B.; van Es, I.E.; van Baarsen, L.G.; Gay, S.; McKinsey, T.A.; Tak, P.P.; et al. Inflammatory cytokines epigenetically regulate rheumatoid arthritis fibroblast-like synoviocyte activation by suppressing HDAC5 expression. Ann. Rheum. Dis. 2016, 75, 430–438. [Google Scholar] [CrossRef]
  114. Park, J.K.; Shon, S.; Yoo, H.J.; Suh, D.H.; Bae, D.; Shin, J.; Jun, J.H.; Ha, N.; Song, H.; Choi, Y.I.; et al. Inhibition of histone deacetylase 6 suppresses inflammatory responses and invasiveness of fibroblast-like-synoviocytes in inflammatory arthritis. Arthritis Res. Ther. 2021, 23, 177. [Google Scholar] [CrossRef] [PubMed]
  115. Goschl, L.; Preglej, T.; Boucheron, N.; Saferding, V.; Muller, L.; Platzer, A.; Hirahara, K.; Shih, H.Y.; Backlund, J.; Matthias, P.; et al. Histone deacetylase 1 (HDAC1): A key player of T cell-mediated arthritis. J. Autoimmun. 2020, 108, 102379. [Google Scholar] [CrossRef]
  116. Li, M.; Hu, W.; Wang, R.; Li, Z.; Yu, Y.; Zhuo, Y.; Zhang, Y.; Wang, Z.; Qiu, Y.; Chen, K.; et al. Sp1 S-Sulfhydration Induced by Hydrogen Sulfide Inhibits Inflammation via HDAC6/MyD88/NF-κB Signaling Pathway in Adjuvant-Induced Arthritis. Antioxidants 2022, 11, 732. [Google Scholar] [CrossRef]
  117. Chung, Y.L.; Lee, M.Y.; Wang, A.J.; Yao, L.F. A therapeutic strategy uses histone deacetylase inhibitors to modulate the expression of genes involved in the pathogenesis of rheumatoid arthritis. Mol. Ther. 2003, 8, 707–717. [Google Scholar] [CrossRef]
  118. Nishida, K.; Komiyama, T.; Miyazawa, S.; Shen, Z.N.; Furumatsu, T.; Doi, H.; Yoshida, A.; Yamana, J.; Yamamura, M.; Ninomiya, Y.; et al. Histone deacetylase inhibitor suppression of autoantibody-mediated arthritis in mice via regulation of p16INK4a and p21(WAF1/Cip1) expression. Arthritis Rheum. 2004, 50, 3365–3376. [Google Scholar] [CrossRef]
  119. Park, J.K.; Jang, Y.J.; Oh, B.R.; Shin, J.; Bae, D.; Ha, N.; Choi, Y.I.; Youn, G.S.; Park, J.; Lee, E.Y.; et al. Therapeutic potential of CKD-506, a novel selective histone deacetylase 6 inhibitor, in a murine model of rheumatoid arthritis. Arthritis Res. Ther. 2020, 22, 176. [Google Scholar] [CrossRef]
  120. Oh, B.R.; Suh, D.H.; Bae, D.; Ha, N.; Choi, Y.I.; Yoo, H.J.; Park, J.K.; Lee, E.Y.; Lee, E.B.; Song, Y.W. Therapeutic effect of a novel histone deacetylase 6 inhibitor, CKD-L, on collagen-induced arthritis in vivo and regulatory T cells in rheumatoid arthritis in vitro. Arthritis Res. Ther. 2017, 19, 154. [Google Scholar] [CrossRef]
  121. Li, M.; Liu, X.; Sun, X.; Wang, Z.; Guo, W.; Hu, F.; Yao, H.; Cao, X.; Jin, J.; Wang, P.G.; et al. Therapeutic effects of NK-HDAC-1, a novel histone deacetylase inhibitor, on collagen-induced arthritis through the induction of apoptosis of fibroblast-like synoviocytes. Inflammation 2013, 36, 888–896. [Google Scholar] [CrossRef] [PubMed]
  122. Ciechomska, M.; Roszkowski, L.; Maslinski, W. DNA Methylation as a Future Therapeutic and Diagnostic Target in Rheumatoid Arthritis. Cells 2019, 8, 953. [Google Scholar] [CrossRef]
  123. Karouzakis, E.; Gay, R.E.; Michel, B.A.; Gay, S.; Neidhart, M. DNA hypomethylation in rheumatoid arthritis synovial fibroblasts. Arthritis Rheum. 2009, 60, 3613–3622. [Google Scholar] [CrossRef] [PubMed]
  124. Richardson, B. Effect of an inhibitor of DNA methylation on T cells. II. 5-Azacytidine induces self-reactivity in antigen-specific T4+ cells. Hum. Immunol. 1986, 17, 456–470. [Google Scholar] [CrossRef] [PubMed]
  125. Nile, C.J.; Read, R.C.; Akil, M.; Duff, G.W.; Wilson, A.G. Methylation status of a single CpG site in the IL6 promoter is related to IL6 messenger RNA levels and rheumatoid arthritis. Arthritis Rheum. 2008, 58, 2686–2693. [Google Scholar] [CrossRef]
  126. Richardson, B.; Scheinbart, L.; Strahler, J.; Gross, L.; Hanash, S.; Johnson, M. Evidence for impaired T cell DNA methylation in systemic lupus erythematosus and rheumatoid arthritis. Arthritis Rheum. 1990, 33, 1665–1673. [Google Scholar] [CrossRef] [PubMed]
  127. Pieper, J.; Johansson, S.; Snir, O.; Linton, L.; Rieck, M.; Buckner, J.H.; Winqvist, O.; van Vollenhoven, R.; Malmstrom, V. Peripheral and site-specific CD4+ CD28null T cells from rheumatoid arthritis patients show distinct characteristics. Scand. J. Immunol. 2014, 79, 149–155. [Google Scholar] [CrossRef]
  128. Ai, R.; Boyle, D.L.; Wang, W.; Firestein, G.S. Distinct DNA Methylation Patterns of Rheumatoid Arthritis Peripheral Blood and Synovial Tissue T Cells. ACR Open Rheumatol. 2021, 3, 127–132. [Google Scholar] [CrossRef]
  129. Kolarz, B.; Ciesla, M.; Dryglewska, M.; Majdan, M. Peptidyl Arginine Deiminase Type 4 Gene Promoter Hypo-Methylation in Rheumatoid Arthritis. J. Clin. Med. 2020, 9, 2049. [Google Scholar] [CrossRef]
  130. Fu, L.H.; Ma, C.L.; Cong, B.; Li, S.J.; Chen, H.Y.; Zhang, J.G. Hypomethylation of proximal CpG motif of interleukin-10 promoter regulates its expression in human rheumatoid arthritis. Acta Pharmacol. Sin. 2011, 32, 1373–1380. [Google Scholar] [CrossRef]
  131. Evangelatos, G.; Fragoulis, G.E.; Koulouri, V.; Lambrou, G.I. MicroRNAs in rheumatoid arthritis: From pathogenesis to clinical impact. Autoimmun. Rev. 2019, 18, 102391. [Google Scholar] [CrossRef] [PubMed]
  132. Wang, L.; Wang, C.; Jia, X.; Yu, J. Circulating Exosomal miR-17 Inhibits the Induction of Regulatory T Cells via Suppressing TGFBR II Expression in Rheumatoid Arthritis. Cell. Physiol. Biochem. 2018, 50, 1754–1763. [Google Scholar] [CrossRef]
  133. Tsai, M.H.; Chi, M.C.; Hsu, J.F.; Lee, I.T.; Lin, K.M.; Fang, M.L.; Lee, M.H.; Lee, C.W.; Liu, J.F. Urban Particulate Matter Enhances ROS/IL-6/COX-II Production by Inhibiting MicroRNA-137 in Synovial Fibroblast of Rheumatoid Arthritis. Cells 2020, 9, 1378. [Google Scholar] [CrossRef] [PubMed]
  134. Liu, N.; Feng, X.; Wang, W.; Zhao, X.; Li, X. Paeonol protects against TNF-α-induced proliferation and cytokine release of rheumatoid arthritis fibroblast-like synoviocytes by upregulating FOXO3 through inhibition of miR-155 expression. Inflamm. Res. 2017, 66, 603–610. [Google Scholar] [CrossRef]
  135. Zhao, J.; Chen, B.; Peng, X.; Wang, C.; Wang, K.; Han, F.; Xu, J. Quercetin suppresses migration and invasion by targeting miR-146a/GATA6 axis in fibroblast-like synoviocytes of rheumatoid arthritis. Immunopharmacol. Immunotoxicol. 2020, 42, 221–227. [Google Scholar] [CrossRef]
  136. Pauley, K.M.; Cha, S. miRNA-146a in rheumatoid arthritis: A new therapeutic strategy. Immunotherapy 2011, 3, 829–831. [Google Scholar] [CrossRef] [PubMed]
  137. Mucientes, A.; Lisbona, J.M.; Mena-Vázquez, N.; Ruiz-Limón, P.; Manrique-Arija, S.; Fernández-Nebro, A. miRNA-Mediated Epigenetic Regulation of Treatment Response in RA Patients-A Systematic Review. Int. J. Mol. Sci. 2022, 23, 12989. [Google Scholar] [CrossRef]
  138. Zhang, Y.; Liu, L.; Pillman, K.A.; Hayball, J.; Su, Y.W.; Xian, C.J. Differentially expressed miRNAs in bone after methotrexate treatment. J. Cell. Physiol. 2022, 237, 965–982. [Google Scholar] [CrossRef]
  139. Bure, I.V.; Mikhaylenko, D.S.; Kuznetsova, E.B.; Alekseeva, E.A.; Bondareva, K.I.; Kalinkin, A.I.; Lukashev, A.N.; Tarasov, V.V.; Zamyatnin, A.A., Jr.; Nemtsova, M.V. Analysis of miRNA Expression in Patients with Rheumatoid Arthritis during Olokizumab Treatment. J. Pers. Med. 2020, 10, 205. [Google Scholar] [CrossRef]
  140. Wang, R.; Li, M.; Ding, Q.; Cai, J.; Yu, Y.; Liu, X.; Mao, J.; Zhu, Y.Z. Neuron navigator 2 is a novel mediator of rheumatoid arthritis. Cell. Mol. Immunol. 2021, 18, 2288–2289. [Google Scholar] [CrossRef]
  141. Wang, R.; Li, M.; Wu, W.; Qiu, Y.; Hu, W.; Li, Z.; Wang, Z.; Yu, Y.; Liao, J.; Sun, W.; et al. NAV2 positively modulates inflammatory response of fibroblast-like synoviocytes through activating Wnt/β-catenin signaling pathway in rheumatoid arthritis. Clin. Transl. Med. 2021, 11, e376. [Google Scholar] [CrossRef] [PubMed]
  142. Stanczyk, J.; Ospelt, C.; Karouzakis, E.; Filer, A.; Raza, K.; Kolling, C.; Gay, R.; Buckley, C.D.; Tak, P.P.; Gay, S.; et al. Altered expression of microRNA-203 in rheumatoid arthritis synovial fibroblasts and its role in fibroblast activation. Arthritis Rheum. 2011, 63, 373–381. [Google Scholar] [CrossRef] [PubMed]
  143. Philippe, L.; Alsaleh, G.; Suffert, G.; Meyer, A.; Georgel, P.; Sibilia, J.; Wachsmann, D.; Pfeffer, S. TLR2 expression is regulated by microRNA miR-19 in rheumatoid fibroblast-like synoviocytes. J. Immunol. 2012, 188, 454–461. [Google Scholar] [CrossRef]
  144. Gantier, M.P.; Stunden, H.J.; McCoy, C.E.; Behlke, M.A.; Wang, D.; Kaparakis-Liaskos, M.; Sarvestani, S.T.; Yang, Y.H.; Xu, D.; Corr, S.C.; et al. A miR-19 regulon that controls NF-κB signaling. Nucleic Acids Res. 2012, 40, 8048–8058. [Google Scholar] [CrossRef] [PubMed]
  145. Philippe, L.; Alsaleh, G.; Pichot, A.; Ostermann, E.; Zuber, G.; Frisch, B.; Sibilia, J.; Pfeffer, S.; Bahram, S.; Wachsmann, D.; et al. MiR-20a regulates ASK1 expression and TLR4-dependent cytokine release in rheumatoid fibroblast-like synoviocytes. Ann. Rheum. Dis. 2013, 72, 1071–1079. [Google Scholar] [CrossRef] [PubMed]
  146. Mu, N.; Gu, J.; Huang, T.; Zhang, C.; Shu, Z.; Li, M.; Hao, Q.; Li, W.; Zhang, W.; Zhao, J.; et al. A novel NF-κB/YY1/microRNA-10a regulatory circuit in fibroblast-like synoviocytes regulates inflammation in rheumatoid arthritis. Sci. Rep. 2016, 6, 20059. [Google Scholar] [CrossRef]
  147. Nakamachi, Y.; Ohnuma, K.; Uto, K.; Noguchi, Y.; Saegusa, J.; Kawano, S. MicroRNA-124 inhibits the progression of adjuvant-induced arthritis in rats. Ann. Rheum. Dis. 2016, 75, 601–608. [Google Scholar] [CrossRef]
  148. Miao, C.G.; Yang, Y.Y.; He, X.; Huang, C.; Huang, Y.; Qin, D.; Du, C.L.; Li, J. MicroRNA-152 modulates the canonical Wnt pathway activation by targeting DNA methyltransferase 1 in arthritic rat model. Biochimie 2014, 106, 149–156. [Google Scholar] [CrossRef]
  149. Miao, C.G.; Shi, W.J.; Xiong, Y.Y.; Yu, H.; Zhang, X.L.; Qin, M.S.; Du, C.L.; Song, T.W.; Li, J. miR-375 regulates the canonical Wnt pathway through FZD8 silencing in arthritis synovial fibroblasts. Immunol. Lett. 2015, 164, 1–10. [Google Scholar] [CrossRef]
  150. McNeill, E.M.; Roos, K.P.; Moechars, D.; Clagett-Dame, M. Nav2 is necessary for cranial nerve development and blood pressure regulation. Neural Dev. 2010, 5, 6. [Google Scholar] [CrossRef]
  151. Chakrabarti, S.; Hore, Z.; Pattison, L.A.; Lalnunhlimi, S.; Bhebhe, C.N.; Callejo, G.; Bulmer, D.C.; Taams, L.S.; Denk, F.; Smith, E.S.J. Sensitization of knee-innervating sensory neurons by tumor necrosis factor-α-activated fibroblast-like synoviocytes: An in vitro, coculture model of inflammatory pain. Pain 2020, 161, 2129–2141. [Google Scholar] [CrossRef]
  152. Wu, W.J.; Jia, W.W.; Liu, X.H.; Pan, L.L.; Zhang, Q.Y.; Yang, D.; Shen, X.Y.; Liu, L.; Zhu, Y.Z. S-propargyl-cysteine attenuates inflammatory response in rheumatoid arthritis by modulating the Nrf2-ARE signaling pathway. Redox Biol. 2016, 10, 157–167. [Google Scholar] [CrossRef] [PubMed]
  153. Yu, Y.; Wang, Z.; Yang, Q.; Ding, Q.; Wang, R.; Li, Z.; Fang, Y.; Liao, J.; Qi, W.; Chen, K.; et al. A novel dendritic mesoporous silica based sustained hydrogen sulfide donor for the alleviation of adjuvant-induced inflammation in rats. Drug Deliv. 2021, 28, 1031–1042. [Google Scholar] [CrossRef] [PubMed]
  154. Yu, Y.; Wang, Z.; Ding, Q.; Yu, X.; Yang, Q.; Wang, R.; Fang, Y.; Qi, W.; Liao, J.; Hu, W.; et al. The Preparation of a Novel Poly(Lactic Acid)-Based Sustained H(2)S Releasing Microsphere for Rheumatoid Arthritis Alleviation. Pharmaceutics 2021, 13, 742. [Google Scholar] [CrossRef] [PubMed]
  155. Yu, D.; Du, J.; Pu, X.; Zheng, L.; Chen, S.; Wang, N.; Li, J.; Chen, S.; Pan, S.; Shen, B. The Gut Microbiome and Metabolites Are Altered and Interrelated in Patients with Rheumatoid Arthritis. Front. Cell. Infect. Microbiol. 2021, 11, 763507. [Google Scholar] [CrossRef]
  156. Bergot, A.S.; Giri, R.; Thomas, R. The microbiome and rheumatoid arthritis. Best Pr. Res. Clin. Rheumatol. 2019, 33, 101497. [Google Scholar] [CrossRef]
  157. Shaw, A.M.; Qasem, A.; Naser, S.A. Modulation of PTPN2/22 Function by Spermidine in CRISPR-Cas9-Edited T-Cells Associated with Crohn’s Disease and Rheumatoid Arthritis. Int. J. Mol. Sci. 2021, 22, 8883. [Google Scholar] [CrossRef]
  158. Ding, J.; Orozco, G. Identification of rheumatoid arthritis causal genes using functional genomics. Scand. J. Immunol. 2019, 89, e12753. [Google Scholar] [CrossRef]
  159. Lee, M.H.; Shin, J.I.; Yang, J.W.; Lee, K.H.; Cha, D.H.; Hong, J.B.; Park, Y.; Choi, E.; Tizaoui, K.; Koyanagi, A.; et al. Genome Editing Using CRISPR-Cas9 and Autoimmune Diseases: A Comprehensive Review. Int. J. Mol. Sci. 2022, 23, 1337. [Google Scholar] [CrossRef]
Figure 1. The structure of the joint and its change in rheumatoid arthritis.
Figure 1. The structure of the joint and its change in rheumatoid arthritis.
Biomolecules 13 00766 g001
Figure 2. Cellular mechanisms in rheumatoid arthritis. The pathogenesis of RA is regulated by the interaction between FLS, MLS, and immune cells.
Figure 2. Cellular mechanisms in rheumatoid arthritis. The pathogenesis of RA is regulated by the interaction between FLS, MLS, and immune cells.
Biomolecules 13 00766 g002
Figure 3. The main signaling pathway in rheumatoid arthritis. NF-κB, MAPK, PI3K-AKT, Wnt, and JAK-STAT are the classic pathways in RA.
Figure 3. The main signaling pathway in rheumatoid arthritis. NF-κB, MAPK, PI3K-AKT, Wnt, and JAK-STAT are the classic pathways in RA.
Biomolecules 13 00766 g003
Figure 4. Epigenetic modification and promising new targets in rheumatoid arthritis. Histone modifications, DNA methylation, and Non-coding RNA modifications are common in epigenetic studies of RA.
Figure 4. Epigenetic modification and promising new targets in rheumatoid arthritis. Histone modifications, DNA methylation, and Non-coding RNA modifications are common in epigenetic studies of RA.
Biomolecules 13 00766 g004
Table 1. Biological agents for the treatment of RA.
Table 1. Biological agents for the treatment of RA.
Biological AgentsTarget CellTarget FactorMechanismsStatusReferences
Etanercept (ENT) TNF-αIn binding to TNFα and TNFβ, it has a more remarkable ability than endogenous soluble TNFRs to prevent the proinflammatory reactionApproved[66]
Infliximab (IFX) TNF-αIt binds to the target receptor of TNFα and inhibits the production of inflammatory factorsApproved[67,68]
Adalimumab (ADA) TNF-αIt specifically binds to TNFα and blocks its interaction with TNF receptors on p55 and p75 cellsApproved[69,70]
Golimumab TNF-αLike Infliximab, it has a strong binding force on soluble and transmembrane TNFα, resulting in TNFα neutralizationApproved[71]
Certolizumab pegol TNF-αIt is a human antibody consisting of an antigen-binding fragment (Fab ‘) of an anti-TNF-α monoclonal antibodyApproved[72]
RituximabB cells It removes all B cells except the pre-B cells and plasma cellsApproved[73]
Abatacept (CTLA4-Ig)T cells By binding to CD80 and CD86 on the surface of B cells, it inhibits co-stimulation and activation of T cells, leading to the downregulation of inflammatory mediatorsApproved[74]
Tocilizumab IL-6It inhibits IL-6-mediated inflammatory responses, induces/expanses Breg cells, and down-regulates the expression of pro-inflammatory factors.Approved[75]
Sirukumab IL-6It binds IL-6R and blocks its binding with IL-6 molecules to inhibit the IL-6 signaling pathway to slow the diseaseApproved[76]
Olokizumab IL-6It recognizes the “site 3” site of IL-6, thus preventing the binding of IL-6 to gp130 and achieving the purpose of inhibiting the IL-6 pathwayApproved[77]
Clazakizumab IL-6It targets soluble and membrane-bound IL-6 receptors to prevent IL-6 receptor complex formation and inhibit IL-6 signalingApproved[78]
Brodalumab IL-17It leads to the disruption of the IL-17 pathway by blocking the activities of 17A, 17E, and 17FApproved[79]
Tofacitinib JAKIt inhibits STAT phosphorylation and activation through JAK1/JAK3 dimer-mediated signaling and inhibits B lymphocyte activation and differentiationApproved[80]
Baricitinib JAKIt selectively inhibits JAK1 and JAK2Approved[81]
Upadacitinib JAKIt selectively inhibits JAK1 and effectively inhibits STAT3 phosphorylation induced by IL-6 and IFNγApproved[82]
Filgotinib JAKIt causes the selective inhibition of JAK1 and reduces the pro-inflammatory responseApproved[83]
Peficitinib JAKIt is a JAK3 inhibitor, restraining the activation of pro-inflammatory cytokine signalingApproved[84]
SHR0302 JAKIts highly selective inhibition of JAK1 suppresses the inflammatory responseClinical trial phase 3 (NCT04333771)[85]
VX-509 JAKIt causes selective JAK3 inhibition and mitigates the inflammatory responseClinical trial phase 2/3 (NCT01830985)[86]
Table 2. Natural compounds for potential treatment of RA.
Table 2. Natural compounds for potential treatment of RA.
ExtractSourceIn VivoIn VitroPotential PathwayReferences
RhoifolinA flavanone extracted from Rhus succedaneaAlleviating the symptoms of CIA mice, and improving cytokines and oxidation indexes NF-κB[87]
Perillyl alcoholA monoterpene separated and collected from the essential oils of citronella, lavandin, peppermint, spearmint, celery seeds, and a few different plantsImproving the levels of MDA, GSH, NO, IL-1β, IL-10, IL-6, TNFα, and PGE2 in CIA miceProtecting LPS-induced RAW 264.7 activation and improving inflammatory and oxidative markersTLR4/NF-κB and Keap1/Nrf2[88]
ResveratrolA natural polyphenol found in grapes and Polygonum cuspidatumAttenuating paw swelling and decreasing serum antioxidant enzyme levels in AA ratsActivating antioxidant pathways and inhibiting the proliferation of FLSNrf2-ARE[89]
Reducing H2O2-induced mitochondrial ROS productionNrf2–Keap1[90]
Salvianolic acidA major phytoconstituent of the plant Radix Salvia miltiorrhiza.Alleviating oxidative stress state in mice and antagonizes free radicals [91]
wogoninA flavonoid contained in the Scutellaria Baicalensis Georgi rootDecreasing the content of lipid peroxides in CFA rats [92]
MagnoflorineA main component purified from Clematis manshurica Rupr.Decreasing inflammatory responses in AIA ratsDecreasing the proliferation, migration, invasion, and reactive oxygen species levels in IL-1β-treated MH7API3K/Akt/NF-κBKeap1-Nrf2/HO-1[93]
ChelerythrineA benzo phenanthridine alkaloid isolated from the root of Papaveraceae.Protecting AIA rats from inflammation and bone damage.Inhibiting the migration and colony-formation of the HFLS-RA, increasing the intracellular level of ROS, promoting apoptosisAMPK/mTOR/ULK-1[94]
CantleyosideA iridoid glycosides in P. hookeri Leading to mitochondrial damage by Ca2+ overload and ROS release, promotes apoptosis and inhibits proliferation of HFLS-RAAMPK/Sirt 1/NF-κB[95]
IcariinA prenylated flavonol glycoside isolated from Epimedium Interfering with cell cycle progression, increasing intracellular ROS level, inducing cell apoptosis, and inhibiting the proliferation of FLS [96]
NeohesperidinA flavone compound isolated from various dietary sources Anti-migration, anti-invasion, anti-oxidative, and apoptosis-induced in RA-FLSMAPK[97]
HesperidinA flavanone present in large quantities in citrus fruitsReducing inflammation in CIA rats, regulating adenosine nucleotide and nucleoside hydrolases, reducing intracellular ROS, attenuating the apoptotic process of bone marrow cells [98]
CelastrolA quinone-methylated triterpenoid extracted from Tripterygium wilfordiiReducing the degree of joint swelling, arthritis index score, inflammatory cell infiltration and synovial hyperplasiaInhibiting the activation of NLRP3 inflammasome and the production of ROS induced by LPS and ATP in THP-1 cells. [99]
Reducing the expression of inflammatory factors in the serum of CIA-induced RA mice, alleviate cartilage tissue damage and inflammatory invasion in miceInhibiting FLSs proliferation and enhancing autophagyPI3K/AKT/mTOR[100]
FangchinolineAn alkaloid can be extracted from the roots of Stephaniae tetrandraImproving behavioral parameters and inflammatory signs in C/K rats and CIA mouseDecreasing the production of inflammatory cytokines and ROS in HFLSMAPK
NF-κB
[101]
Sinomenine Alleviating symptoms in CAIA mice by protecting their joints from destructionInhibiting the secretion of IL-6 and IL-33 and ROS production in RASFs, thereby mediating the protective effect against bone destructionKeap1-Nrf2[102]
Gentiolic acidAn active component in the whole plant or root of Gaultheria leucocarpa Bl.va. crenulata (Kurz) t.z.HsuReducing the expression of inflammatory factors in serum of CIA rats and relieving the symptoms of RAInhibiting proliferation and invasion of MH7A, and the apoptosis was promotedRAF/ERK[103]
CurcuminA tetraterpenoid obtained from Curcuma longaInhibiting the expression of pro-inflammatory cytokines TNF-α, IL-6, and IL-17 in CIA-stimulated mice, relieving hind paw edema in RA miceReducing TNFα-induced MH7A proliferation and migration ability and promoting cell apoptosis.PI3K/AKT[104]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhu, M.; Ding, Q.; Lin, Z.; Fu, R.; Zhang, F.; Li, Z.; Zhang, M.; Zhu, Y. New Targets and Strategies for Rheumatoid Arthritis: From Signal Transduction to Epigenetic Aspect. Biomolecules 2023, 13, 766. https://doi.org/10.3390/biom13050766

AMA Style

Zhu M, Ding Q, Lin Z, Fu R, Zhang F, Li Z, Zhang M, Zhu Y. New Targets and Strategies for Rheumatoid Arthritis: From Signal Transduction to Epigenetic Aspect. Biomolecules. 2023; 13(5):766. https://doi.org/10.3390/biom13050766

Chicago/Turabian Style

Zhu, Menglin, Qian Ding, Zhongxiao Lin, Rong Fu, Fuyuan Zhang, Zhaoyi Li, Mei Zhang, and Yizhun Zhu. 2023. "New Targets and Strategies for Rheumatoid Arthritis: From Signal Transduction to Epigenetic Aspect" Biomolecules 13, no. 5: 766. https://doi.org/10.3390/biom13050766

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop