Next Article in Journal
Effect of Nigella sativa on Selected Gastrointestinal Diseases
Previous Article in Journal
Using Odontoblasts Derived from Dog Endometrial Stem Cells Encapsulated in Fibrin Gel Associated with BMP-2 in a Rat Pulp-Capping Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

Rheumatoid Arthritis and Reactive Oxygen Species: A Review

1
Division of Orthopedic Surgery, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
2
Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8510, Japan
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2023, 45(4), 3000-3015; https://doi.org/10.3390/cimb45040197
Submission received: 9 February 2023 / Revised: 30 March 2023 / Accepted: 31 March 2023 / Published: 3 April 2023

Abstract

:
Rheumatoid arthritis (RA) is a chronic, systemic inflammatory disease that causes progressive joint damage and can lead to lifelong disability. Numerous studies support the hypothesis that reactive oxygen species (ROS) are associated with RA pathogenesis. Recent advances have clarified the anti-inflammatory effect of antioxidants and their roles in RA alleviation. In addition, several important signaling pathway components, such as nuclear factor kappa B, activator-protein-1, nuclear factor (erythroid-derived 2)-like 2/kelch-like associated protein, signal transducer and activator of transcription 3, and mitogen-activated protein kinases, including c-Jun N-terminal kinase, have been identified to be associated with RA. In this paper, we outline the ROS generation process and relevant oxidative markers, thereby providing evidence of the association between oxidative stress and RA pathogenesis. Furthermore, we describe various therapeutic targets in several prominent signaling pathways for improving RA disease activity and its hyper oxidative state. Finally, we reviewed natural foods, phytochemicals, chemical compounds with antioxidant properties and the association of microbiota with RA pathogenesis.

1. Introduction

Rheumatoid arthritis (RA) is a chronic, systemic inflammatory disease that causes progressive joint damage and can lead to lifelong disability [1]. RA is characterized by synovial inflammatory cell infiltration, synovial hyperplasia, angiogenesis, and cartilage damage, which in turn can lead to bone degradation [2]. Recent data have demonstrated that bone and cartilage degradation in RA are due to an increase in metalloproteinases (MMPs) and serin proteases [3]. Many studies have reported that circulating neutrophils show an aberrant, activated phenotype in RA, characterized by delayed apoptosis and the increased production of reactive oxygen species (ROS) and cytokines, resulting in bone and joint damage [4,5,6,7].
Numerous studies have reported an association between RA pathogenesis and ROS [8,9,10]. For example, circulating neutrophils in RA patients can generate superoxide anions (O2−), unlike those in healthy controls [11]. Moreover, the levels of catalase and ceruloplasmin were remarkably elevated in the synovial fluid of RA patients compared to those of controls, suggesting that antioxidant activity was enhanced in RA pathogenesis in response to inflammation (Figure 1) [12].
The overproduction of nitric oxide (NO) contributes to the pathogenesis of chronic arthritis [13]. In collagen-induced rodent arthritis models, increased levels of nitrite/nitrate in the plasma [14,15] and synovial fluid [16] and a high expression of inducible nitric oxide synthase (NOS) in proliferating synovium [14] and chondrocytes [17] have been reported. Increased circulating levels of nitrate/nitrite have been detected in arthritis patients [18], and the synovial tissues of RA patients were characterized by high iNOS expression [19,20] and enhanced NO production [14].
In this review, the relationship between ROS and RA is summarized. ROS are briefly described, and the association between antioxidants and RA treatment is reviewed by focusing on neutrophils and the autophagy of synovial fibroblasts in RA. Redox signaling in RA is also discussed. Finally, natural food or antioxidants with the potential for improving RA disease severity and novel therapeutic targets are enumerated.

2. Oxidative Stress and Markers

There are two methods for measuring oxidative DNA damage. Steady-state damage can be measured when DNA is isolated from human cells or tissues and analyzed for base damage products. Several DNA base damage products, such as nucleoside 8-hydroxy-deoxyguanosine (8-OH-dG), 8-hydroxy-adenine, and 7-methyl-8-hydroxyguanine, are excreted in human urine [21,22,23]. The most used marker is 8-OH-dG, which is usually measured using high-performance liquid chromatography. The level of 8-OH-dG in urine is probably not influenced by the diet, as nucleotides are not absorbed from the gut. In RA patients, 8-OH-dG level was significantly decreased by methotrexate or TNF-inhibitors, such as infliximab and etanercept [24,25,26].
Biologics are widely used in the clinical setting in patients with RA; however, side effects are a concern. If other targets, such as antioxidants or ROS-inhibitory or antioxidant-promoting agents, are induced, the clinical symptoms of RA, the number of tender joints, arthralgias, joint swellings, and osteoarthritic joint disorders, are reduced. Physical activity, such as knee flexion, can also be improved. Commonly used oxidative stress markers include 8-hydroxy-2-deoxyguanosine (8-OHdG), thiobarbituric acid reactive substances, malondialdehyde (MDA), isoprostane (IsoPs) and its metabolites, and allantoin [27,28] (Table 1), as well as advanced glycation end product (AGE) [29]. IsoPs is the isomer of prostaglandin, produced from polyunsaturated fatty acids involved in the peroxidation of phospholipid membranes by free radical or oxidative stress [30]. Allantoin is the final metabolite in the non-enzymatic oxidation of uric acid. Although a small number of cases were reported, allantoin levels were significantly higher in RA patients than in healthy subjects. Uric acid acts as a scavenger of free radicals, and allantoin increases reactivity, suggesting the involvement of free radicals in RA pathophysiology [31].
NADPH oxidase is the most important generator of ROS in the vasculature [32,33]. It comprises membrane-bound gp91 (phox) and gp22(phox) and cellular solute subunits, such as p47(phox), p67 (phox), and the small GTPase Rac. Neutrophil cytosolic factor-1 (NCF-1), also known as p47 phox, which is an essential subunit of NADPH oxidase 2 (NOX2), an enzyme that promotes oxidative stress. The activation of NOX2 first occurs in the cytoplasm. The extent of oxidative stress is derived from the production of superoxides, which is called oxidative disruption oxidative burst. Genetic polymorphisms in the genes coding for the Nox2 complex have become a hot topic in human immune diseases. ROS derived from the NCF1 and NOX2 complex are important regulators of rheumatoid arthritis, multiple sclerosis, psoriasis, psoriatic arthritis, gout, lupus, and other chronic inflammatory diseases [34]. Table 1 summarizes the oxidative stress markers that are available (Table 1) [24,25,26,35,36,37,38,39,40,41,42,43].
Table 1. Markers of oxidative stress.
Table 1. Markers of oxidative stress.
ProcessMarkersSamplesExplanationsReferences
Nucleic acid oxidation8-hydroxy-2′-deoxyguanosineUrine, serum, tissueGuanine receives oxidative stress and 8-OHdG is produced by oxidation of carbon and excreted in urine.[24,25,26,35]
4-hydroxynonenal (CloneHNEJ-2)TissueRepresentative oxidative stress product generated at the late stage of lipid peroxidation[36,37]
Lipid peroxidation15-Isoprostane F2tUrine, serum, tissueOxidized phospholipids by free radicals[38]
Malonedialdehyde (MDA)Urine, serum, tissueMDA derives from polyunsaturated lipid acid and reacts to thiobarbitulic acid (TBA) and is detected spectroscopically.[39]
Lipid peroxidase (LPO)Serum, tissueLPO is detected by the methylene blue/hemoglobin method.[40]
Oxidized-LDLSerum, tissueGenerated free radicals oxidizes lipids in LDL and oxidized LDL is formed.[41]
Glucose oxidationPentosidineUrine, serumMain advanced glycation end products (AGEs)
Glycated albumin is a glycation product of albumin.
[24]
DithyrosineUrine, serum, tissueDityrosine is a tyrosine dimer that is formed by the oxidation of tyrosine.
[42]
NO stressNitrotyrosine (NT)Serum, tissueNT is formed by the nitration of protein-bound and free tyrosine residues by reactive peroxinitrite molecules.[43]

3. Neutrophils in RA and ROS

Neutrophils are differentiated cells. In the absence of inflammation, they circulate in the blood for 24–48 h until they return to the bone marrow, leading to apoptosis [44]. In the presence of inflammation, neutrophil apoptosis is delayed by inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-alpha) and granulocyte macrophage colony-stimulating factor (GM-CSF).
The hypoxic environment in RA synovial joints also plays a key role in delaying neutrophil apoptosis by increasing MCL1 expression [45]. Hypoxia can also delay apoptosis via the stabilization of hypoxia-inducible factor-1-alpha (HIF1-α) and the activation of nuclear factor-kappa B (NF-κB) [46]. Furthermore, hypoxia regulates neutrophil retention at the sites of inflammation, thereby prolonging inflammation [47].

4. Autophagy and ROS in RA-SF

Autophagy is involved in the transformation of RA-SF, and microRNA is one of these. MicroRNA (miRNA) is a small non-coding RNA consisting of 18 to 25 nucleotides in length [48]. By downregulating the mRNA translocation of downstream target genes, miRNA suppresses gene expression. miRNAs are essential for cell proliferation, apoptosis, oxidative stress, and immune response. In particular, miR-19 [49], miR-21 [50], MiR-27a [51], and MiR-29a have been reported to be involved in RA pathogenesis. In addition, MiR-650 was significantly less expressed in RASF than in normal cells, whereas AKT2 was highly expressed. The downregulation of MiR650 or the upregulation of AKT2 increased RASF proliferation, migration, and erosion and suppressed apoptosis [52]. miR-126 was associated with PI3KR2 as a target gene, and its overexpression suppressed P13KR2 expression, promoted RASF proliferation, and suppressed apoptosis; thus, miR-126 is a candidate for the predictive biomarker of RA [53]. miR-218-5p was highly expressed in RASFs. Its inhibition severely suppressed the production of oxidative stress and promoted SOD [54]. Collectively, the knockdown of miR-218-5p increased KLF9 expression through the downregulation of the JAK2/STAT3 signaling pathway, suggesting that it may be a potential therapeutic target for controlling RASF growth, apoptosis, and oxidative stress [54].
The myeloid-specific deletion of the gene encoding IRE1α protected mice from inflammatory arthritis, and the IRE1α-specific inhibitor 4u8c attenuated joint inflammation in mice [55]. Recently, oxidative stress was reported to be associated with autophagy/ER stress in the pathogenesis of RA [34,56,57]. Thus, the IRE1/JNK pathway might be a therapeutic target for regulating oxidative stress in RA.

5. Main Transcriptional Factors Associated with ROS

5.1. NF-κB

NF-κB was the first eukaryotic transcription factor shown to respond directly to oxidative stress [58]. It plays a key role in the regulation of numerous genes involved in immune and inflammatory processes [59]. TNFα, IL-1, phorbol ester, lipopolysaccharide, and UV radiation potently activate NF-κB in intact cells. The H2O2 exposure of several types of cells rapidly induced NF-κB activation, indicating that H2O2 might be a mediator of prooxidant-induced NF-κB activation.

5.2. AP-1

Activator protein-1 (AP-1) is a transcription factor for regulating collagen genes, TNFα, IL8, IL9, IL3, IFNγ, adhesion molecules related to the formation of atherosclerotic plaques, and genes involved in the cell division cycle [60]. AP-1 activity is induced in response to certain metals in the presence of H2O2 and several cytokines and other physical and chemical stressors.

6. Redox Signaling

Redox signaling refers to a regulatory process in which the signal is transduced through redox reactions [61]. NF-κB, hypoxia-inducible factor-1 (HIF-1), AP-1, and Nrf2 are redox-sensitive transcriptional factors. They are closely involved in the pathogenesis of RA. NF-κB is crucial for the maturation of immune cells and the production of TNFα and MMPs. TNFα and MMPs aggravate RA. HIF-1 is induced by inflammatory cytokines and needed for angiogenesis and pannus formation in RA. AP-1 and IL-1 beta affect the gene expression and activity of each other, which results in an orchestrated cross-talk. AP-1 also regulates MMP production and synovial hyperplasia in RA [61].
Nrf2 triggers the first line of homeostatic response against endogenous deviations in redox metabolism, proteostasis, and inflammation [62]. Nrf2 deficiency worsened disease activity in experimental arthritis models, whereas its activation exhibited immunoregulatory and anti-inflammatory effects. Thus, the pharmacologic regulation of Nrf2 has gained increasing interest as a strategy to target ROS [63].
In THP-1 monocytes/macrophage cells, light-emitting diode irradiation at 630 nm significantly reduced ROS levels and inhibited the expression of TNFα and IL-1β mRNA. Lastly, the level of phosphorylated NF-κB was significantly reduced, whereas that of its inhibitor, Nrf2, was slightly upregulated [64].

7. New Therapeutic Targets

Poly-(ADP-ribose) polymerase-1 (PARP-1) is a member of the PARP enzyme family, consisting of PARP-1 and several other additional poly-(ADP-ribosylating) enzymes [65]. Peroxynitrite-dependent cell necrosis is partially mediated by a complex process involving DNA damage and the activation of the DNA repair enzyme PARP-1 [66]. PARP-1 detects and transmits the signals of DNA strand breaks induced by various genotoxic insults and oxidants (hydrogen peroxide and peroxynitrite) and free radicals (mainly carbonate or hydroxyradicals) [67,68]. A significant increase in DNA strand breaks in peripheral mononuclear cells was observed in RA patients compared with that in healthy subjects [69,70]. PARP was highly expressed in the joint tissues of collagen-induced arthritis rodent models. Following peroxynitrite formation blockage by selective iNOS inhibitors or the suppression of genetic iNOS, PARP activation was blocked [14]. Several PARP-1 inhibitors, such as nicotinamide [71], 5-inodo-6-amino-1,2-benzopyrone [72], and PJ-34 [73], were previously used.
Scavenging NO might be an alternative strategy for treating inflammatory disorders. Yeo et al. [74] developed a NO-responsive macro-sized hydrogel by incorporating a NO-cleavable cross-linker. The NO-scavenging nanosized hydrogel (NO-Scv gel) reduced inflammation levels by scavenging NO in vitro. Furthermore, the NO-Scv gel suppressed RA onset in a mouse RA model compared with the dexamethasone treatment.
Mateen et al. [75] identified the association of inflammatory cytokines with 25-hydroxy vitamin D (25-OH-D) and ROS in RA patients. The level of 25-OH-D in RA patients was 11.07 ± 4.81 ng/mL, which was significantly reduced compared to that in healthy controls (37.88 ± 9.78 ng/mL). Another study using the DCF-DA method showed that the ROS levels in RA patients were significantly increased compared to those of healthy controls [76].

8. Natural Foods, Phytochemicals, and Chemical Compounds with Antioxidant Properties and the Association of Microbiota with RA Pathogenesis

Once organisms are exposed to free radicals, a series of defense mechanisms are activated, one of which is represented by antioxidants. The most prevalent enzymatic antioxidants are superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT). Nonenzymatic antioxidants, such as ascorbic acid (vitamin C), α-tocophenol (vitamin E), glutathione (GSH), carotenoids, flavonoids, and other antioxidants, also play an important role.
Various interventions have been performed for the dietary pattern disease activity of rheumatoid arthritis [77,78].
Private diet eliminating meat, gluten, and lactose for 3 months in 40 patients with RA significantly decreased in pain, DAS28 scores, CRP level, and the overall state of physical and mental health [79].
A dietary intake of vitamins C and E, zinc, magnesium, copper, and selenium was introduced in 87 RA patients. Of these dietary components, vitamin C intake was related to decreased IL-1b, zinc intake was related to decreased IL-2, and magnesium intake was related to decreased levels of IL-1b and IL-2. In addition, vitamin E and copper intake increased catalase (an enzyme largely involved with anti-inflammatory pathways) expression [80].
The dietary inflammatory index (DII) was assessed for patients with RA and control (case–control study. The mean DII score was higher in the RA patients compared with control cases (0.66 vs. −0.58, p = 0.002). Higher DII scores were significantly correlated with higher CRP, TNF-alpha, DAS-28 scores, and the number of tender joints [81].
The evaluation of diet quality is also important. Participants with lower diet quality showed significantly higher pain and ESR scores [82]. The anti-inflammatory diet in rheumatoid arthritis diet (ADIRA) for 44 patients with RA demonstrated a significant decrease (p = 0.012) in DAS28-ESR as a randomized controlled test [83]. In addition, poor diet quality, as defined by Swedish National Food agency (diet with a low intake of fish, shellfish, whole grain, fruit, and vegetables and a high intake of sausages and sweets), was associated with higher CRP (p = 0.044) and ESR (p = 0.002) levels in patients with RA [84].
The Mediterranean diet (MD) is effective for the decrease in inflammation for patients with RA. Patients with high adherence to the MD showed a significantly lower CRP (p = 0.037) and DAS28 (p = 0.034) than the 40 patients with low or moderate adherence to MD. A healthier gut microbiota status was detected in the high adherence group [85].
Diurnal fasting for 1 month is also effective. A significant decrease in visual analogue pain scores, tender and swollen joints (p = 0.02), DAS-28 (p = 0.003), and ESR was observed [86]. RA patients who continued the diurnal fasting of Ramadan demonstrated a significant improvement in DAS28-CRP (p = 0.001) and DAS28-ESR (p < 0.001) compared with patients who did not participate in the fasting [87].
In anthropometric findings, body mass index (BMI) in patients with RA was correlated with CRP (r = 0.36, p < 0.01) and ESR (r = 0.31, p < 0.01). Asymmetric dimethylarginine (ADMA) is a naturally occurring chemical found in blood plasma. ADMA was associated with increased BMI and disease RA activity. A higher intake of protein was correlated to higher CRP and ESR [88].
Supplementation with high fiber 30 g bars daily for 15 days and 30 days were administered to 10 healthy controls and 29 patients with RA. Increased anti-inflammatory short-chain fatty acids was detected (p < 0.001). Proarthritic cytokines concentrations, such as MCP-1, IL-18, and IL-33, were decreased and the Firmicutes-to-Bacteroides ratio, one of markers of gut microbiota, were decreased (p < 0.05) [89].
Naïve human CD4+ T cells were cultured in 10, 20, 40, and 60 mM NaCl solution for 3 days. NaCl aggravated arthritis by affecting Th17 differentiation [90]. A low sodium diet, as defined as less than 5 μg/day for 3 weeks, significantly reduced the serum levels of transforming growth factor-beta (TGFβ) and IL-9 [91].
The efficacy of flavonoids, PUFAs, and probiotics in the disease activity of RA has also been reported.
Black barberry extract (1000 mg/day for 12 weeks) intervention significantly decreased IL-17 levels and increased IL-10 [92]. The trial of Brazilian propolis (508.5 mg daily for 24 weeks) did not show a significant difference in DAS28-ESR, CRP, simplified disease activity index, or clinical disease activity index [93]. Cinnamon powder (500 mg daily for 8 weeks) showed a significant decrease in the serum levels of CRP and TNF-alpha. It also showed a significant decrease in DAS-28, visual analogue scale, and the tender and swollen joints count (p < 0.001) [94]. Cinnamaldehyde and eugenol on peripheral blood mononuclear cells showed significant dose-dependent decreases in TNF-alpha and IL-6 (p < 0.05), ameliorated reactive oxygen species formation, biomolecular oxidation, and antioxidant defense response (p < 0.05) [95].
Curcumin nanomicelle (120 mg daily for 12 weeks) demonstrated no significant decrease in the DAS-28, tender joint count, swollen joint count, and ESR after intervention [96].
However, increased doses of curcumin (500 mg daily for 8 weeks) showed a significant decrease in insulin resistance, ESR, CRP, triglycerides, weight, body mass index, and the waist circumference of RA patients (p < 0.05) [97].
Associated with a human cell line study, 3′3-diindolylmethane inhibited proliferation, migration, and the invasion of RA fibroblast-like synoviocytes in vitro and significantly decreased TNF-alpha-induced increases in the mRNA levels of MMP-2, MMP-3, MMP-8, and MMP-9, as well as the proinflammatory cytokines, such as IL-6, IL-8, and IL-1b [98].
In addition, polyphenolic extract from extra virgin olive oil inhibited IL-1b-induced MMPs, TNF-a, and IL-6 production (p < 0.001). IL-1b-induced MAPK phosphorylation and nuclear factor kappa B (NF-kB) activation were also significantly decreased (p < 0.001) [99].
Garlic (1000 mg daily for 8 weeks) significantly decreased CRP, TNF-alpha, swollen joint count, pain intensity, tender joint count, DAS-28, and fatigue [100].
Pomegranate extract (500 mg daily for 8 weeks) showed a significant decrease in DAS-28 (p < 0.001), pain intensity (p = 0.03), and health assessment score (HAQ) (p = 0.007). Moreover, glutathione peroxidase concentrations significantly increased (p < 0.001), but resulted in no significant difference in MMP-3 and CRP levels between intervention and control groups [101].
Saffron supplementation (100 mg/day for 12 weeks) significantly decreased tender joint count and swollen joint count, pain intensity, and DAS-28. CRP, TNF-alpha, interferon-gamma, and malonedialdehyde (MDA) were also significantly decreased by saffron supplementation [102].
Sesamin supplementation (200 mg/day for 6 weeks) significantly decreased the serum levels of hyaluronidase, MMP-3, CRP, TNF-alpha, and cyclooxygenase-2 and decreased the tender joint count and severity of pain [103].
The inverse relationship with erythrocyte levels of the n-6 PUFA linoleic acid was detected as a risk of RA development (odds ratio 0.29, 95% CI; 0.12–0.75, p < 0.01) in patients with RA [104].
Flaxseed supplements (30 g per day for 12 weeks) significantly decreased DAS-28 scores, pain severity, morning stiffness, and feelings of disease, compared to regular diet group [105].
In a prospective case–control study, omega-3 PUFA consumption was inversely significantly associated with omega-6, and the omega-6:omega-3 ratio was directly associated with unacceptable and refractory pain [106]. The dietary recall of average weekly servings of fish had significantly lower DAS-28-CRP scores when compared to RA patients who never ate fish or ate it less than once per month [107]. Fish oil n-3 fatty acids (3 g) and reduced-calorie cranberry juice (500 mL daily) was supplemented as a prospective control study. The fish oil-only group showed improvements in DAS28-CRP and adiponectin, but when consuming fish oil supplements together with cranberry juice, significantly reductions in ESR, CRP, DAS28-CRP, adiponectin, and IL-6 levels compared to controls were observed [108]. Notably, five studies have investigated the role of probiotics. Lactobacillus acidophilus (L. acidophilus), Lactobacillus casei (L. casei), Lactococcus lactis, Bifidobacterium (B.) lactis, and Bifidobacterium (B.) bifidum consumption for 60 days improved inflammatory profiles with reductions in white blood cell count, TNF-alpha, and IL-6 levels, but resulted in no significant difference in IL-10 levels, adiponectin, CRP, ESR, ferritin, or DAS-28 [109].
L. acidophilus, L. casei, and B. bifidum dophilus consumption for 8 weeks decreased serum CRP and improved DAS-28 but resulted in no improvement in oxidative stress levels [110]. Synbiotic supplement intake for 8 weeks significantly reduced CRP, DAS-28 scores, and plasma nitric oxide levels [111]. As seen in an in vitro study, recombinant B. bifidum significantly increased IL-10 levels and inhibited levels of IL-6, IL-8, and TNF-alpha to a higher degree than those from food grade bacteria [112]. B. longum significantly inhibited Th17 cells and IL-17-related genes, as well as several proinflammatory mediators [113]. Regarding herbal studies, Nigella sativa extract (500 mg twice daily for 2 months) significantly decreased CRP levels and DAS28 scores [114]. Stachys schtschegleevii tea (2.4 g daily for 8 weeks) markedly reduced DAS-28 scores and serum MMP-3 levels [115]. Xinfeng supplement (three pills, three times a day for 2 months) significantly decreased the DAS-28 score, ESR, and CRP levels [116].
Regarding the effect of anti-oxidants/ROS scavengers, N-acetyl cysteine (600 mg twice daily for 12 weeks) significantly reduced MDA, NO, and total thiol groups compared to the control [117]. Two other reports for serum silicon levels in patients with RA and vitamin K1 (10 mg daily for 8 weeks) showed controversial results [118,119]. Vitamin A (retinol) detoxifies H2O2 through ascorbate peroxidase [63]. Vitamin E (tocopherol and tocotrienols) guards against and detoxifies the products of membrane lipid peroxidation. Vitamin C (ascorbic acid) directly scavenges ROS or indirectly supports in the synthesis of extracellular matrix proteins, such as collagen [120]. Selenium exerted antioxidant effects in patients with RA [121]. It is a conjugate factor of selenoenzymes, such as TrxRs and GPx, and showed anti-inflammatory activity by suppressing the NF-κB cascade [122]. Regarding natural products, Humulus japonicus (HJ), a widely used herbal medicine in Asia with antioxidative effects, significantly decreased the gross arthritic scores and paw swelling in a collagen-induced arthritic mouse model. HJ also significantly inhibited the expression of IL-6 both in vivo and in vitro. The mechanism underlying HJ effects was regulated by STAT3 phosphoylation [123].
Monotropein, an iridoid glycoside isolated from the roots of Morinda officinalis [124], ameliorated H2O2-induced inflammation in human umbilical vein endothelial cells via NF-κB/AP-1 signaling [125]. In addition, it protected against oxidative stress in osteoblasts via Akt/mTOR-mediated autophagy [126]. Perillyl alcohol is a monoterpene that shows anti-inflammatory and antioxidative properties and can be extracted from widely available essential oils. In RAW264.7 cells, the lipopolysaccharide-induced elevation of IL-1b, IL-6, and TNFα levels was completely inhibited by perillyl alcohol. It also inhibited ROS and nitrite levels via the NF-κB and Nrf2 signaling pathways [127]. Moreover, elastr8ol, a quinone-methylated triterpenoid extracted from Tripterygium wilfordii, is used to treat RA. Celastrol-inhibited ROS levels in vitro and attenuated collagen induced arthritis via the NF-κB signaling pathway [128,129]. Genistein, an isoflavone derivative found in soy, decreased the TNFα-induced production of IL-1b, IL-6, and IL-8 in MH7A cells. It also induced NF-κB translocation by TNFα and the phosphorylation of IkB kinase-α/β and IkBα. TNFα-induced adenosine monophosphate-activated protein kinase inhibition was prominently inhibited by genistein [130]. Once IL-21 binds to its receptor, ROS are produced, JAK1/STAT3 signaling is activated, and targeted inflammatory cytokines, such as TNFα, IL-6, MMP-3, and MMP-13, in MH7A fibroblast-like synoviocytes are produced, resulting in the degradation of the extracellular matrix. Nobiletin, a derivative of citrus fruit, attenuated the development of osteoarthritis and inhibited the production of proinflammatory cytokines. Nobiletin potently inhibited the IL21-induced production of ROS by inhibiting the phosphorylation of JAK/STAT3 [131]. Regarding Ayurvedic foods, Gugglipid [132] suppressed collagen degradation and reactive oxygen species in an arthritis mouse model. Kalpaamruthaa also had a suppressive effect on arthritis models [133]. N-acetylcysteine (NAC) is a cysteine prodrug that indirectly activates cysteine-glutamate antiporters [134]. MH7A cells, which are rheumatoid synovial fibroblast-like cell lines, are recombinant cells in which the SV40 T antigen is incorporated into synovial cells collected from RA patients. The amount of ROS produced by MH7A cells was significantly increased at 1 h following the addition of hydrogen peroxide but was significantly decreased following the administration of NAC for 24 h. Therefore, NAC reduced the amount of ROS produced and exerted antioxidant effects in MH7A cells [135]. NAC has antioxidant and detoxifying effects and is a drug clinically prescribed for acetaminophen poisoning and is known to decrease cytokine activity [136]. Nrf2 is the main regulator of the oxidative stress response system [137]. It is normally bound to Keap1; however, when oxidative stress is induced, the conjugated factor p62 is phosphorylated. Nrf2 dissociates from Keap1 and is translocated from the cytoplasm to the nucleus to bind to the oxidative stress response site, promoting the expression of detoxifying enzymes, the antioxidant protein heme oxygenase 1, and anti-inflammatory enzymes. At 24 h after the administration of 1000 μM NAC, the levels of Nrf2 and phosphorylated p62 significantly increased. In a previous clinical study, Nrf2 mRNA and protein levels in the RA synovia were compared to those in the OA synovia. Nrf2 mRNA levels were significantly correlated with the preoperative d-ROM value, suggesting that increased Nrf2 mRNA expression reflects an upregulation of antioxidant capacity in response to high oxidative stresses in RA patients [138]. When cells were observed under a confocal laser scanning microscope, Nrf2, which showed green staining in the untreated cells, was localized in the cytoplasm; however, after NAC administration, Nrf2 was translocated to the nucleus, where it exerted an antioxidant effect. Furthermore, MMP-3 protein levels were significantly reduced by the administration of NAC for 24 h, and JNK phosphorylation was significantly suppressed 3 h after NAC administration. The ROS increase associated with the addition of H2O2 was significantly reduced by the administration of the JNK inhibitor SP600125 [139], similar to the IL-6 concentration in the MH7A cell supernatant. These findings suggest that the JNK pathway plays an important role in the pathway of oxidative stress and IL-6 suppression [135]. One clinical prospective study showed that 600 mg NAC treatment (twice a day) for 8 weeks significantly improved the RA disease progression and serum IL-17 concentration compared to those of the control group [140].
Conversely, another report showed that NAC oral administration for 12 weeks only had partial effects on the global health parameter, visual analogue scale, and health assessment questionnaire but not DAS28 [141]. Thus, the clinical effect of NAC for the treatment of RA patients remains controversial. Recently, novel thiol-amides, NAC-amide (AD4/NACA), and thioredoxin mimetics (TXM-peptides) have been tested for neurodegenerative disorders [142]. The AD4 compound was effective at blocking cocaine-seeking behaviors [143]. Nevertheless, further in vivo and in vitro investigations of the effect of NAC-amide and thioredoxin mimetics in RA are required. The gut microbiota influences metabolic and immune homeostasis. Oxidative stress, such as the generation of ROS, is the main trigger that directly influences the microbial pattern of human microbiota [144]. Obesity itself is associated with the presence of inflammation, oxidative stress, and mitochondrial dysfunction. Furthermore, these circumstances may develop neurodegenerative diseases, such as Alzheimer disease and Parkinson’s disease (PD) [145].
Obesity and neurodegenerative diseases, such as PD, also show dysbiosis (microbiota) [146], and the improvement of dietary pattern (short-chain fatty acid intake) restores this dysregulation pattern [147]. Studies associated with gut microbiota demonstrate that an expansion and/or decrease in bacterial groups is a primary feature in RA compared to the control [148].
The metagenomic shotgun sequencing and a metagenome-wide association study of the fecal, dental, and salivary samples from a cohort of individuals with RA and healthy controls were performed. The results showed that Heaemophilus spp. were depleted and Lactobacillus salivarius was over-represented in patients with RA at all three sites. In very active RA, Lactobacillus salivarius increased [149]. Disease-modifying anti-rheumatic drug (DMARD) treatments partially restore a healthy microbiome because DMARDs improve metagenomic linkage groups (MLGs) in dental and salivary sites after DMARD treatment in patients with RA [149]. Diet is the main environmental factor influencing gut microbiota. The whole dietary pattern of the Mediterranean diet possibly acts as a therapeutic approach by modulating gut microbiota [150]. Red meat and salt are suspected to have harmful effects when controlling the disease activity of RA [151].
In addition, PUFAs, vitamin D, and probiotics supplementation demonstrated protective effects regarding RA development by improving the environment of gut microbiota. Healthy lifestyle and nutrition are encouraged for patients with RA [151].

9. Conclusions

We described the role of ROS in the pathogenesis of rheumatoid arthritis. ROS are elevated in the serum of patients with RA. The biomarkers of oxidative stress were demonstrated. Oxidative stress was reported to be associated with autophagy/ER stress in the pathogenesis of RA. In RA, the main regulator of redox signaling is the Nrf2/Keap1 pathway. New therapeutic targets and natural food or phytochemicals with the potential for improving the severity of RA were described. Dietary patterns, Mediterranean diet (MD), flavonoids, PUFAs, probiotics, herbals, and antioxidants are useful for decreasing RA disease severity.
Recently, there has been progress in the dysregulation of gut microbiota in patients with RA, and the improvement of the environment of gut microbiota by diet would be a target for future research.

Author Contributions

Conceptualization, N.K.; methodology, N.K., T.K. and M.O.; validation, N.K.; data curation, N.K., T.K. and M.O.; writing—original draft preparation, writing—Review and editing, supervision, N.K.; visualization; T.K. and M.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board of Niigata University (protocol code: 2018-0377 and date of approval: 25 July 2019).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to acknowledge the support of Rika Kakutani (Division of Orthopedic Surgery, Niigata University) in collecting and reviewing the reference articles and drawing Figure 1, and Akiko Yoshi (Department of Neurosurgery, Brain Research Institute, Niigata University) for providing exclusive support with the experiments.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Smolen, J.S.; Aletaha, D.; McInnes, I.B. Rheumatoid arthritis. Lancet 2016, 388, 2023–2038. [Google Scholar] [CrossRef] [PubMed]
  2. McInnes, I.B.; Schett, G. The Pathogenesis of Rheumatoid Arthritis. N. Engl. J. Med. 2011, 365, 2205–2219. [Google Scholar] [CrossRef] [Green Version]
  3. Tossetta, G.; Fantone, S.; Licini, C.; Marzioni, D.; Mattioli-Belmonte, M. The multifaced role of HtrA1 in the development of joint and skeletal disorders. Bone 2022, 157, 116350. [Google Scholar] [CrossRef] [PubMed]
  4. Wright, H.L.; Lyon, M.; Chapman, E.A.; Moots, R.J.; Edwards, S.W. Rheumatoid Arthritis Synovial Fluid Neutrophils Drive Inflammation Through Production of Chemokines, Reactive Oxygen Species, and Neutrophil Extracellular Traps. Front. Immunol. 2021, 11, 584116. [Google Scholar] [CrossRef]
  5. Trofimenko, A.S.; Mozgovaya, E.E.; Bedina, S.A.; Spasov, A.A. Ambiguities in Neutrophil Extracellular Traps. Ongoing Concepts and Potential Biomarkers for Rheumatoid Arthritis: A Narrative Review. Curr. Rheumatol. Rev. 2021, 17, 283–293. [Google Scholar] [CrossRef] [PubMed]
  6. Lee, K.H.; Kronbichler, A.; Park, D.D.-Y.; Park, Y.; Moon, H.; Kim, H.; Choi, J.H.; Choi, Y.; Shim, S.; Lyu, I.S.; et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: A comprehensive review. Autoimmun. Rev. 2017, 16, 1160–1173. [Google Scholar] [CrossRef]
  7. Mirshafiey, A.; Mohsenzadegan, M. The role of reactive oxygen species in immunopathogenesis of rheumatoid arthritis. Iran. J. Allergy Asthma Immunol. 2008, 7, 195–202. [Google Scholar]
  8. Veselinovic, M.; Barudzic, N.; Vuletic, M.; Zivkovic, V.; Tomic-Lucic, A.; Djuric, D.; Jakovljevic, V. Oxidative stress in rheumatoid arthritis patients: Relationship to diseases activity. Mol. Cell Biochem. 2014, 391, 225–232. [Google Scholar] [CrossRef]
  9. Smallwood, M.J.; Nissim, A.; Knight, A.R.; Whiteman, M.; Haigh, R.; Winyard, P.G. Oxidative stress in autoimmune rheumatic diseases. Free Radic. Biol. Med. 2018, 125, 3–14. [Google Scholar] [CrossRef]
  10. Phull, A.-R.; Nasir, B.; Haq, I.U.; Kim, S.J. Oxidative stress, consequences and ROS mediated cellular signaling in rheumatoid arthritis. Chem. Biol. Interact. 2018, 281, 121–136. [Google Scholar] [CrossRef]
  11. Eggleton, P.; Wang, L.; Penhallow, J.; Crawford, N.; Brown, K.A. Differences in oxidative response of subpopulations of neutrophils from healthy subjects and patients with rheumatoid arthritis. Ann. Rheum. Dis. 1995, 54, 916–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Biemond, P.; Swaak, A.J.G.; Koster, J.F. Protective factors against oxygen free radicals and hydrogen peroxide in rheumatoid arthritis synovial fluid. Arthritis Rheum. 1984, 27, 760–765. [Google Scholar] [CrossRef] [Green Version]
  13. Henrotin, Y.E.; Bruckner, P.; Pujol, J.-P. The role of reactive oxygen species in homeostasis and degradation of cartilage. Osteoarthr. Cartil. 2003, 11, 747–755. [Google Scholar] [CrossRef] [Green Version]
  14. Connor, J.R.; Manning, P.T.; Settle, S.L.; Moore, W.M.; Jerome, G.M.; Webber, R.; Tjoeng, F.; Currie, M.G. Suppression of adjuvant-induced arthritis by selective inhibition of inducible nitric oxide synthase. Eur. J. Pharmacol. 1995, 273, 15–24. [Google Scholar] [CrossRef] [PubMed]
  15. Cuzzocrea, S.; Chatterjee, P.K.; Mazzon, E.; McDonald, M.C.; Dugo, L.; di Paola, R.; Serraino, I.; Britti, D.; Caputi, A.P.; Thiemermann, C. Beneficial effects of GW274150, a novel, potent and selective inhibitor of iNOS activity, in a rodent model of collagen-induced arthritis. Eur. J. Pharmacol. 2002, 453, 119–129. [Google Scholar] [CrossRef]
  16. Stefanovic-Racic, M.; Meyers, K.; Meschter, C.; Coffey, J.W.; Hoffman, R.A.; Evans, C.H. Comparison of the nitric oxide synthase inhibitors methylarginine and aminoguanidine as prophylactic and therapeutic agents in rat adjuvant arthritis. J. Rheumatol. 1995, 22, 1922–1928. [Google Scholar] [PubMed]
  17. Yonekura, Y.; Koshiishi, I.; Yamada, K.-I.; Mori, A.; Uchida, S.; Nakamura, T.; Utsumi, H. Association between the expression of inducible nitric oxide synthase by chondrocytes and its nitric oxide-generating activity in adjuvant arthritis in rats. Nitric Oxide 2003, 8, 164–169. [Google Scholar] [CrossRef]
  18. Farrell, A.J.; Blake, D.R.; Palmer, R.M.; Moncada, S. Increased concentrations of nitrite in synovial fluid and serum samples suggest increased nitric oxide synthesis in rheumatic diseases. Ann. Rheum. Dis. 1992, 51, 1219–1222. [Google Scholar] [CrossRef] [Green Version]
  19. McInnes, I.; Leung, B.; Field, M.D.; Wei, X.; Huang, F.; Sturrock, R.D.; Kinninmonth, A.W.G.; Weidner, J.R.; Mumford, R.A.; Liew, F. Production of nitric oxide in the synovial membrane of rheumatoid and osteoarthritis patients. J. Exp. Med. 1996, 184, 1519–1524. [Google Scholar] [CrossRef]
  20. Sakurai, H.; Kohsaka, H.; Liu, M.F.; Higashiyama, H.; Hirata, Y.; Kanno, K.; Saito, I.; Miyasaka, N. Nitric oxide production and inducible nitric oxide synthase expression in inflammatory arthritides. J. Clin. Investig. 1995, 96, 2357–2363. [Google Scholar] [CrossRef] [Green Version]
  21. Shigenaga, M.K.; Gimeno, C.J.; Ames, B.N. Urinary 8-hydroxy-2′-deoxyguanosine as a biological marker of in vivo oxidative DNA damage. Proc. Natl. Acad. Sci. USA 1989, 86, 9697–9701. [Google Scholar] [CrossRef] [Green Version]
  22. Loft, S.; Fischer-Nielsen, A.; Jeding, I.B.; Vistisen, K.; Poulsen, H.E. 8-hydroxydeoxyguanosine as a urinary biomarker of oxidative DNA damage. J. Toxicol. Environ. Health 1993, 40, 391–404. [Google Scholar] [CrossRef] [PubMed]
  23. Stillwell, W.G.; Xu, H.X.; Adkins, J.A.; Wishnok, J.S.; Tannenbaum, S.R. Analysis of methylated and oxidized purines in urine by capillary gas chromatography-mass spectrometry. Chem. Res. Toxicol. 1989, 2, 94–99. [Google Scholar] [CrossRef] [PubMed]
  24. Kageyama, Y.; Takahashi, M.; Nagafusa, T.; Torikai, E.; Nagano, A. Methotrexate reduces the levels of pentosidine and 8-hydroxy-deoxy guanosine in patients with rheumatoid arthritis. Mod. Rheumatol. 2007, 17, 398–402. [Google Scholar] [CrossRef] [PubMed]
  25. Kageyama, Y.; Takahashi, M.; Ichikawa, T.; Torikai, E.; Nagano, A. Reduction of oxidative stress marker levels by anti-TNF-alpha antibody, infliximab, in patients with rheumatoid arthritis. Clin. Exp. Rheumatol. 2008, 26, 73–80. [Google Scholar]
  26. Kageyama, Y.; Takahashi, M.; Nagafusa, T.; Torikai, E.; Nagano, A. Etanercept reduces the oxidative stress marker levels in patients with rheumatoid arthritis. Rheumatol. Int. 2008, 28, 245–251. [Google Scholar] [CrossRef]
  27. Czerska, M.; Mikołajewska, K.; Zieliński, M.; Gromadzińska, J.; Wąsowicz, W. Today’s oxidative stress markers. Med. Pract. 2015, 66, 393–405. [Google Scholar] [CrossRef]
  28. Yoshikawa, T.; Naito, Y. What is oxidative stress? Jpn. Med. Assoc. J. 2002, 45, 271–276. [Google Scholar]
  29. Monu; Agnihotri, P.; Biswas, S. AGE/Non-AGE Glycation: An Important Event in Rheumatoid Arthritis Pathophysiology. Inflammation 2022, 45, 477–496. [Google Scholar] [CrossRef]
  30. Crankshaw, D.; Rangachari, P. Isoprostanes: More than just mere markers. Mol. Cell Biochem. 2003, 253, 125–130. [Google Scholar] [CrossRef]
  31. Yardim-Akaydin, S.; Sepici, A.; Ozkan, Y.; Torun, M.; Şimşek, B.; Sepici, V. Oxidation of Uric Acid in Rheumatoid Arthritis: Is Allantoin a Marker of Oxidative Stress? Free Radic. Res. 2004, 38, 623–628. [Google Scholar] [CrossRef] [PubMed]
  32. Manea, A. NADPH oxidase-derived reactive oxygen species: Involvement in vascular physiology and pathology. Cell Tissue Res. 2010, 342, 325–339. [Google Scholar] [CrossRef] [PubMed]
  33. Montezano, A.C.; Touyz, R.M. Reactive oxygen species and endothelial function—Role of nitric oxide synthase uncoupling and Nox family nicotinamide adenine dinucleotide phosphate oxidases. Basic Clin. Pharmacol. Toxicol. 2012, 110, 87–94. [Google Scholar] [CrossRef] [PubMed]
  34. Holmdahl, R.; Sareila, O.; Olsson, L.M.; Bäckdahl, L.; Wing, K. Ncf1 polymorphism reveals oxidative regulation of autoimmune chronic inflammation. Immunol. Rev. 2016, 269, 228–247. [Google Scholar] [CrossRef]
  35. Shiihara, T.; Kato, M.; Ichiyama, T.; Takahashi, Y.; Tanuma, N.; Miyata, R.; Hayasaka, K. Acute encephalopathy with refractory status epilepticus: Bilateral mesial temporal and claustral lesions, associated with a peripheral marker of oxidative DNA damage. J. Neurol. Sci. 2006, 250, 159–161. [Google Scholar] [CrossRef]
  36. Tanaka, T.; Nishiyama, Y.; Okada, K.; Hirota, K.; Matsui, M.; Yodoi, J.; Hiai, H.; Toyokuni, S. Induction and nuclear translocation of thioredoxin by oxidative damage in the mouse kidney: Independence of tubular necrosis inductionand sulfthydryl depletion. Lab. Investig. 1997, 77, 145–155. [Google Scholar]
  37. Toyokuni, S.; Miyake, N.; Hiai, H.; Hagiwara, M.; Kawakishi, S.; Osawa, T.; Uchida, K. The monoclonal antibody specific for the 4-hydroxy-2-nonenal histidine adduct. FEBS Lett. 1995, 359, 189–191. [Google Scholar] [CrossRef] [Green Version]
  38. Morrow, J.D.; Zackert, W.E.; Yang, J.P.; Kurhts, E.H.; Callewaert, D.; Dworski, R.; Kanai, K.; Taber, D.; Moore, K.; Oates, J.A.; et al. Quantification of the Major Urinary Metabolite of 15-F2t-Isoprostane (8-iso-PGF2α) by a Stable Isotope Dilution Mass Spectrometric Assay. Anal. Biochem. 1999, 269, 326–331. [Google Scholar] [CrossRef]
  39. Koike, M.; Nojiri, H.; Kanazawa, H.; Yamaguchi, H.; Miyagawa, K.; Nagura, N.; Banno, S.; Iwase, Y.; Kurosawa, H.; Kaneko, K. Superoxide dismutase activity is significantly lower in end-stage osteoarthritic cartilage than non-osteoarthritic cartilage. PLoS ONE 2018, 13, e0203944. [Google Scholar] [CrossRef]
  40. Wang, M.; Liehr, J. Identification of fatty acid hydroperoxide cofactors in the cytochrome P450-mediated oxidation of estrogens to quinone metabolites. Role and balance of lipid peroxides during estrogen-induced carcinogenesis. J. Biol. Chem. 1994, 269, 284–291. [Google Scholar] [CrossRef]
  41. Koubaa, N.; Nakbi, A.; Smaoui, M.; Abid, N.; Chaaba, R.; Abid, M.; Hammami, M. Hyperhomocysteinemia and elevated ox-LDL in Tunisian type 2 diabetic patients: Role of genetic and dietary factors. Clin. Biochem. 2007, 40, 1007–1014. [Google Scholar] [CrossRef] [PubMed]
  42. Kato, Y.; Wu, X.; Naito, M.; Nomura, H.; Kitamoto, N.; Osawa, T. Immunohistochemical detection of protein dityrosine in atherosclerotic lesion of apo-E-deficient mice using a novel monoclonal antibody. Biochem. Biophys. Res. Comun. 2000, 275, 11–15. [Google Scholar] [CrossRef]
  43. Souza, J.M.; Peluffo, G.; Radi, R. Protein tyrosine nitration-functional alteration or just a biomarker? Free Rad. Biol. Med. 2008, 45, 357–366. [Google Scholar] [CrossRef]
  44. McCracken, J.M.; Allen, L.-A.H. Regulation of Human Neutrophil Apoptosis and Lifespan in Health and Disease. J. Cell Death 2014, 7, 15–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Cross, A.; Barnes, T.; Bucknall, R.C.; Edwards, S.W.; Moots, R.J. Neutrophil apoptosis in rheumatoid arthritis is regulated by local oxygen tensions within joints. J. Leukoc. Biol. 2006, 80, 521–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Walmsley, S.R.; Print, C.; Farahi, N.; Peyssonnaux, C.; Johnson, R.S.; Cramer, T.; Sobolewski, A.; Condliffe, A.M.; Cowburn, A.S.; Johnson, N.; et al. Hypoxia-induced neutrophil survival is mediated by HIF-1α–dependent NF-κB activity. J. Exp. Med. 2005, 201, 105–115. [Google Scholar] [CrossRef] [Green Version]
  47. Elks, P.; Van Eeden, F.J.; Dixon, G.; Wang, X.; Reyes-Aldasoro, C.C.; Ingham, P.W.; Whyte, M.K.B.; Walmsley, S.; Renshaw, S.A. Activation of hypoxia-inducible factor-1α (Hif-1α) delays inflammation resolution by reducing neutrophil apoptosis and reverse migration in a zebrafish inflammation model. Blood 2011, 118, 712–722. [Google Scholar] [CrossRef] [Green Version]
  48. Rupaimoole, R.; Slack, F.J. Microma therapeutics: Towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 2016, 16, 203–222. [Google Scholar] [CrossRef]
  49. Li, Z.; Cai, J.; Cao, X. MiR-19 suppresses fibroblast-like synoviocytes cytokine release by targeting toll like receptor 2 in rheumatoid arthritis. Am. J. Transl. Res. 2016, 8, 5512–5518. [Google Scholar]
  50. Jin, S.; Chen, H.; Li, Y.; Zhong, H.; Sun, W.; Wang, J.; Zhang, T.; Ma, J.; Yan, S.; Zhang, J.; et al. Maresin 1 improves the Treg/Th17 imbalance in rheumatoid arthritis through miR-21. Ann. Rheum. Dis. 2018, 77, 1644–1652. [Google Scholar] [CrossRef]
  51. Shi, D.-L.; Shi, G.-R.; Xie, J.; Du, X.-Z.; Yang, H. MicroRNA-27a Inhibits Cell Migration and Invasion of Fibroblast-Like Synoviocytes by Targeting Follistatin-Like Protein 1 in Rheumatoid Arthritis. Mol. Cells 2016, 39, 611–618. [Google Scholar] [CrossRef] [PubMed]
  52. Xu, X.; Chen, H.; Zhang, Q.; Xu, J.; Shi, Q.; Wang, M. MiR-650 inhibits proliferation, migration and invasion of rheumatoid arthritis synovial fibroblasts by targeting AKT2. Biomed. Pharmacother. 2017, 88, 535–541. [Google Scholar] [CrossRef] [PubMed]
  53. Qu, Y.; Wu, J.; Deng, J.-X.; Zhang, Y.-P.; Liang, W.-Y.; Jiang, Z.-L.; Yu, Q.-H.; Li, J. MicroRNA-126 affects rheumatoid arthritis synovial fibroblast proliferation and apoptosis by targeting PIK3R2 and regulating PI3K-AKT signal pathway. Oncotarget 2016, 7, 74217–74226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Chen, M.; Li, M.; Zhang, N.; Sun, W.; Wang, H.; Wei, W. Mechanism of Mir-218-5P in Autophagy, Apoptosis and Oxidative Stress in Rheumatoid Arthritis Synovial Fibroblasts is Mediated by Klf9 and Jak/Stat3 Pathways. J. Investig. Med. 2021, 69, 824–832. [Google Scholar] [CrossRef]
  55. Qiu, Q.; Zheng, Z.; Chang, L.; Zhao, Y.-S.; Tan, C.; Dandekar, A.; Zhang, Z.; Lin, Z.; Gui, M.; Li, X.; et al. Toll-like receptor-mediated IRE1α activation as a therapeutic target for inflammatory arthritis. EMBO J. 2013, 32, 2477–2490. [Google Scholar] [CrossRef] [Green Version]
  56. Kundu, S.; Ghosh, P.; Datta, S.; Ghosh, A.; Chattopadhyay, S.; Chatterjee, M. Oxidative stress as a potential biomarker for determining disease activity in patients with Rheumatoid Arthritis. Free Radic. Res. 2012, 46, 1482–1489. [Google Scholar] [CrossRef]
  57. Scherz-Shouval, R.; Shvets, E.; Fass, E.; Shorer, H.; Gil, L.; Elazar, Z. Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J. 2007, 26, 1749–1760. [Google Scholar] [CrossRef] [Green Version]
  58. Schreck, R.; Rieber, P.; Baeuerle, P. Reactive oxygen intermediates as apparently widely used messengers in the activation of the NF-kappa B transcription factor and HIV-1. EMBO J. 1991, 10, 2247–2258. [Google Scholar] [CrossRef]
  59. Rhee, S.G. Redox signaling: Hydrogen peroxide as intracellular messenger. Exp. Mol. Med. 1999, 31, 53–59. [Google Scholar] [CrossRef] [Green Version]
  60. Sukkar, S.G.; Rossi, E. Oxidative stress and nutritional prevention in autoimmune rheumatic diseases. Autoimmun. Rev. 2004, 3, 199–206. [Google Scholar] [CrossRef]
  61. Le Rossignol, S.; Ketheesan, N.; Haleagrahara, N. Redox-sensitive transcription factors play a significant role in the development of rheumatoid arthritis. Int. Rev. Immunol. 2018, 37, 129–143. [Google Scholar] [CrossRef] [PubMed]
  62. Robledinos-Antón, N.; Fernández-Ginés, R.; Manda, G.; Cuadrado, A. Activators and Inhibitors of NRF2: A Review of Their Potential for Clinical Development. Oxidative Med. Cell Longev. 2019, 2019, 9372182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lee, W.-L.; Huang, J.-Y.; Shyur, L.-F. Phytoagents for Cancer Management: Regulation of Nucleic Acid Oxidation, ROS, and Related Mechanisms. Oxidative Med. Cell. Longev. 2013, 2013, 925804. [Google Scholar] [CrossRef] [Green Version]
  64. Li, Y.; Wei, S.; Zhang, K.; Fang, Y.; Liu, H.; Jin, Z.; Guo, Q.; He, J.; Song, W.; Zhang, F. The inflammation and reactive oxygen species regulated by Nrf2 and NF-κB signaling pathways in 630-nm light-emitting diode irradiation treated THP-1 monocytes/macrophages. Lasers Med. Sci. 2021, 36, 1411–1419. [Google Scholar] [CrossRef] [PubMed]
  65. García, S.; Conde, C. The Role of Poly(ADP-ribose) Polymerase-1 in Rheumatoid Arthritis. Mediat. Inflamm. 2015, 2015, 837250. [Google Scholar] [CrossRef] [Green Version]
  66. Szabó, C.; Zingarelli, B.; O’Connor, M.; Salzman, A.L. DNA strand breakage, activation of poly (ADP-ribose) synthetase, and cellular energy depletion are involved in the cytotoxicity of macrophages and smooth muscle cells exposed to peroxynitrite. Proc. Natl. Acad. Sci. USA 1996, 93, 1753–1758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Szabó, C. Poly(ADP-ribose) polymerase activation by reactive nitrogen species—Relevance for the pathogenesis of inflammation. Nitric Oxide 2006, 14, 169–179. [Google Scholar] [CrossRef]
  68. Alemasova, E.E.; Lavrik, O.I. Poly(ADP-ribosyl)ation by PARP1: Reaction mechanism and regulatory proteins. Nucleic Acids Res. 2019, 47, 3811–3827. [Google Scholar] [CrossRef] [Green Version]
  69. Bhusate, L.L.; Herbert, K.E.; Scott, D.L.; Perrett, D. Increased DNA strand breaks in mononuclear cells from patients with rheumatoid arthritis. Ann. Rheum. Dis. 1992, 51, 8–12. [Google Scholar] [CrossRef] [Green Version]
  70. Souliotis, V.L.; Vlachogiannis, N.; Pappa, M.; Argyriou, A.; Sfikakis, P.P. DNA damage accumulation, defective chromatin organization and deficient DNA repair capacity in patients with rheumatoid arthritis. Clin. Immunol. 2019, 203, 28–36. [Google Scholar] [CrossRef]
  71. Song, S.B.; Park, J.S.; Chung, G.J.; Lee, I.H.; Hwang, E.S. Diverse therapeutic efficacies and more diverse mechanisms of nicotinamide. Metabolomics 2019, 15, 137. [Google Scholar] [CrossRef]
  72. Szabó, C.; Virág, L.; Cuzzocrea, S.; Scott, G.S.; Hake, P.; O’Connor, M.P.; Zingarelli, B.; Salzman, A.; Kun, E. Protection against peroxynitrite-induced fibroblast injury and arthritis development by inhibition of poly(ADP-ribose) synthase. Proc. Natl. Acad. Sci. USA 1998, 95, 3867–3872. [Google Scholar] [CrossRef] [Green Version]
  73. Mabley, J.G.; Jagtap, P.; Perretti, M.; Getting, S.J.; Salzman, A.L.; Virág, L.; Szabó, É.; Soriano, F.G.; Liaudet, L.; Abdelkarim, G.E.; et al. Anti-inflammatory effects of a novel, potent inhibitor of poly (ADP-ribose) polymerase. Inflamm. Res. 2001, 50, 561–569. [Google Scholar] [CrossRef] [PubMed]
  74. Yeo, J.; Lee, Y.M.; Lee, J.; Park, D.; Kim, K.; Kim, J.; Park, J.; Kim, W.J. Nitric Oxide-Scavenging Nanogel for Treating Rheumatoid Arthritis. Nano Lett. 2019, 19, 6716–6724. [Google Scholar] [CrossRef]
  75. Mateen, S.; Moin, S.; Shahzad, S.; Khan, A.Q. Level of inflammatory cytokines in rheumatoid arthritis patients: Correlation with 25-hydroxy vitamin D and reactive oxygen species. PLoS ONE 2017, 12, e0178879. [Google Scholar] [CrossRef] [PubMed]
  76. Keller, A.; Mohamed, A.; Dröse, S.; Brandt, U.; Fleming, I.; Brandes, R. Analysis of Dichlorodihydrofluorescein and Dihydrocalcein as Probes for the Detection of Intracellular Reactive Oxygen Species. Free Radic. Res. 2004, 38, 1257–1267. [Google Scholar] [CrossRef]
  77. Lanspa, M.; Kothe, B.; Pereira, M.R.; Kesselman, M.M.; Petrosky, S.N. A Systematic Review of Nutritional Interventions on Key Cytokine Pathways in Rheumatoid Arthritis and Its Implications for Comorbid Depression: Is a More Comprehensive Approach Required? Cureus 2022, 14, e28031. [Google Scholar] [CrossRef] [PubMed]
  78. Marquez, A.M.; Evans, C.; Bolston, K.; Kesselman, M. Nutritional interventions and supplementation for rheumatoid arthritis patients: A systematic review for clinical application, Part 1: Dieting. Curr. Rheumatol. Res. 2020, 1, 17–26. [Google Scholar] [CrossRef]
  79. Guagnano, M.T.; D’ Angelo, C.; Caniglia, D.; Di Giovanni, P.; Celletti, E.; Sabatini, E.; Speranza, L.; Bucci, M.; Cipollone, F.; Paganelli, R. Improvement of inflammation and pain after three months’ exclusion diet in rheumatoid arthritis patients. Nutrients 2021, 13, 3535. [Google Scholar] [CrossRef]
  80. Arablou, T.; Aryaeian, N.; Djalali, M.; Shahram, F.; Rasouli, L. Association between dietary intake of some antioxidant micronutrients with some inflammatory and antioxidant markers in active Rheumatoid Arthritis patients. Int. J. Vitam. Nutr. Res. 2019, 89, 238–245. [Google Scholar] [CrossRef]
  81. Tandorost, A.; Kheirouri, S.; Moludi, J.; Seyedmardani, S. Association of Dietary Inflammatory Index (DII) with disease activity and inflammatory cytokines in the patients with rheumatoid arthritis. Int. J. Clin. Pract. 2021, 75, e14792. [Google Scholar] [CrossRef] [PubMed]
  82. McGarrity-Yoder, M.E.; Insel, K.C.; Crane, T.E.; Pace, T.W. Diet quality and disease activity in rheumatoid arthritis. Nutr. Health 2021, 28, 581–590. [Google Scholar] [CrossRef] [PubMed]
  83. Vadell, A.K.; Bärebring, L.; Hulander, E.; Gjertsson, I.; Lindqvist, H.M.; Winkvist, A. Anti-inflammatory Diet in Rheumatoid Arthritis (ADIRA)—A randomized, controlled crossover trial indicating effects on disease activity. Am. J. Clin. Nutr. 2020, 111, 1203–1213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Bärebring, L.; Winkvist, A.; Gjertsson, I.; Lindqvist, H.M. Poor Dietary Quality Is Associated with Increased Inflammation in Swedish Patients with Rheumatoid Arthritis. Nutrients 2018, 10, 1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Diamanti, A.P.; Panebianco, C.; Salerno, G.; Di Rosa, R.; Salemi, S.; Sorgi, M.; Meneguzzi, G.; Mariani, M.; Rai, A.; Iacono, D.; et al. Impact of Mediterranean Diet on Disease Activity and Gut Microbiota Composition of Rheumatoid Arthritis Patients. Microorganisms 2020, 8, 1989. [Google Scholar] [CrossRef]
  86. Assar, S.; Almasi, P.; Almasi, A. Effect of Islamic fasting on the severity of rheumatoid arthritis. JNFH 2020, 8, 28–33. [Google Scholar] [CrossRef]
  87. Ben Nessib, D.; Maatallah, K.; Ferjani, H.; Kaffel, D.; Hamdi, W. Impact of Ramadan diurnal intermittent fasting on rheumatic diseases. Clin. Rheumatol. 2020, 39, 2433–2440. [Google Scholar] [CrossRef]
  88. Naghashian, F.; Hosseinzadeh-Attar, M.J.; Akhlaghi, M.; Yekaninejad, M.S.; Aryaeian, N.; Derakhshanian, H. The relationship between anthrometric status and rheumatoid arthritis. Exploring the role of nesfatin and asymmetric dimethylarginine. Acta Rheumatol. Port. 2019, 44, 126–131. [Google Scholar]
  89. Dürholz, K.; Hofmann, J.; Iljazovic, A.; Häger, J.; Lucas, S.; Sarter, K.; Strowig, T.; Bang, H.; Rech, J.; Schett, G.; et al. Dietary Short-Term Fiber Interventions in Arthritis Patients Increase Systemic SCFA Levels and Regulate Inflammation. Nutrients 2020, 12, 3207. [Google Scholar] [CrossRef]
  90. Jung, S.M.; Kim, Y.; Kim, J.; Jung, H.; Yi, H.; Rim, Y.A.; Park, N.; Kwok, S.-K.; Park, S.-H.; Ju, J.H. Sodium Chloride Aggravates Arthritis via Th17 Polarization. Yonsei Med. J. 2019, 60, 88–97. [Google Scholar] [CrossRef]
  91. Scrivo, R.; Massaro, L.; Barbati, C.; Vomero, M.; Ceccarelli, F.; Spinelli, F.R.; Riccieri, V.; Spagnoli, A.; Alessandri, C.; Desideri, G.; et al. The role of dietary sodium intake on the modulation of T helper 17 cells and regulatory T cells in patients with rheumatoid arthritis and systemic lupus erythematosus. PLoS ONE 2017, 12, e0184449. [Google Scholar] [CrossRef]
  92. Ayaeian, N.; Hadidi, M.; Mahmoudi, M.; Asgari, M.; Hezaveh, Z.S.; Sadehi, S.K. The effect of black barberry hydroalcoholic extract on immune mediators in patients with active rheumatoid arthritis: A randomized, double-blind, controlled clinical trial. Phytother. Res. 2021, 35, 1062–1068. [Google Scholar] [CrossRef]
  93. Matsumoto, Y.; Takahashi, K.; Sugioka, Y.; Inui, K.; Okano, T.; Mandai, K.; Yamada, Y.; Shintani, A.; Koike, T. Double-blinded randomized controlled trial to reveal the effects of Brazilian propolis intake on rheumatoid arthritis disease activity index; BeeDAI. PLoS ONE 2021, 16, e0252357. [Google Scholar] [CrossRef] [PubMed]
  94. Shishehbor, F.; Safar, M.R.; Rajaei, E.; Haghighizadeh, M.H. Cinnamon consumption improves clinical symptoms and inflammatory markers in women with rheumatoid arthritis. J. Am. Coll. Nutr. 2018, 37, 685–690. [Google Scholar] [CrossRef]
  95. Mateen, S.; Rehman, T.; Shahzad, S.; Naeem, S.S.; Faizy, A.F.; Khan, A.Q.; Khan, M.S.; Husain, F.M.; Moin, S. Anti-oxidant and anti-inflammatory effects of cinnamaldehyde and eugenol on mononuclear cells of rheumatoid arthritis patients. Eur. J. Pharmacol. 2019, 852, 14–24. [Google Scholar] [CrossRef] [PubMed]
  96. Javadi, M.; Haghighian, H.K.; Goodarzy, S.; Abbasi, M.; Nassiri-Asl, M. Effect of curcumin nanomicelle on the clinical symptoms of patients with rheumatoid arthritis: A randomized, double-blind, controlled trial. Int. J. Rheum. Dis. 2019, 22, 1857–1862. [Google Scholar] [CrossRef]
  97. Pourhabibi-Zarandi, F.; Rafraf, M.; Zayeni, H.; Asghari-Jafarabadi, M.; Ebrahimi, A. Effects of curcumin supplementation on metabolic parameters, inflammatory factors and obesity values in women with rheumatoid arthritis: A randomized, double-blind, placebo-controlled clinical trial. Phytother. Res. 2022, 36, 1797–1806. [Google Scholar] [CrossRef]
  98. Du, H.; Zhang, X.; Zeng, Y.; Huang, X.; Chen, H.; Wang, S.; Wu, J.; Li, Q.; Zhu, W.; Li, H.; et al. A Novel Phytochemical, DIM, Inhibits Proliferation, Migration, Invasion and TNF-α Induced Inflammatory Cytokine Production of Synovial Fibroblasts From Rheumatoid Arthritis Patients by Targeting MAPK and AKT/mTOR Signal Pathway. Front. Immunol. 2019, 10, 1620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Rosillo, M.; Alarcón-De-La-Lastra, C.; Castejón, M.L.; Montoya, T.; Cejudo-Guillén, M.; Sánchez-Hidalgo, M. Polyphenolic extract from extra virgin olive oil inhibits the inflammatory response in IL-1β-activated synovial fibroblasts. Br. J. Nutr. 2019, 121, 55–62. [Google Scholar] [CrossRef]
  100. Moosavian, S.P.; Paknahad, Z.; Habibagahi, Z.; Maracy, M. The effects of garlic (Allium sativum) supplementation on inflammatory biomarkers, fatigue, and clinical symptoms in patients with active rheumatoid arthritis: A randomized, double-blind, placebo-controlled trial. Phytother. Res. 2020, 34, 2953–2962. [Google Scholar] [CrossRef] [PubMed]
  101. Ghavipour, M.; Sotoudeh, G.; Tavakoli, E.; Mowla, K.; Hasanzadeh, J.; Mazloom, Z. Pomegranate extract alleviates disease activity and some blood biomarkers of inflammation and oxidative stress in Rheumatoid Arthritis patients. Eur. J. Clin. Nutr. 2017, 71, 92–96. [Google Scholar] [CrossRef]
  102. Hamidi, Z.; Aryaeian, N.; Abolghasemi, J.; Shirani, F.; Hadidi, M.; Fallah, S.; Moradi, N. The effect of saffron supplement on clinical outcomes and metabolic profiles in patients with active rheumatoid arthritis: A randomized, double-blind, placebo-controlled clinical trial. Phytother. Res. 2020, 34, 1650–1658. [Google Scholar] [CrossRef]
  103. Helli, B.; Shahi, M.M.; Mowla, K.; Jalali, M.T.; Haghighian, H.K. A randomized, triple-blind, placebo-controlled clinical trial, evaluating the sesamin supplement effects on proteolytic enzymes, inflammatory markers, and clinical indices in women with rheumatoid arthritis. Phytother. Res. 2019, 33, 2421–2428. [Google Scholar] [CrossRef] [PubMed]
  104. De Pablo, P.; Romaguera, D.; Fisk, H.L.; Calder, P.C.; Quirke, A.-M.; Cartwright, A.J.; Panico, S.; Mattiello, A.; Gavrila, D.; Navarro, C.; et al. High erythrocyte levels of the n-6 polyunsaturated fatty acid linoleic acid are associated with lower risk of subsequent rheumatoid arthritis in a southern European nested case–control study. Ann. Rheum. Dis. 2018, 77, 981–987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Ghaseminasab-Parizi, M.; Nazarinia, M.-A.; Akhlaghi, M. The effect of flaxseed with or without anti-inflammatory diet in patients with rheumatoid arthritis, a randomized controlled trial. Eur. J. Nutr. 2022, 61, 1377–1389. [Google Scholar] [CrossRef] [PubMed]
  106. Lourdudoss, C.; Di Giuseppe, D.; Wolk, A.; Westerlind, H.; Klareskog, L.; Alfredsson, L.; Van Vollenhoven, R.F.; Lampa, J. Dietary Intake of Polyunsaturated Fatty Acids and Pain in Spite of Inflammatory Control Among Methotrexate-Treated Early Rheumatoid Arthritis Patients. Arthritis Care Res. 2018, 70, 205–212. [Google Scholar] [CrossRef] [PubMed]
  107. Tedeschi, S.K.; Bathon, J.M.; Giles, J.T.; Lin, T.-C.; Yoshida, K.; Solomon, D.H. Relationship Between Fish Consumption and Disease Activity in Rheumatoid Arthritis. Arthritis Care Res. 2018, 70, 327–332. [Google Scholar] [CrossRef] [Green Version]
  108. Fatel, E.C.; Rosa, F.T.; Alfieri, D.F.; Flauzino, T.; Scavuzzi, B.M.; Lozovoy, M.A.; Iriyoda, T.M.; Simão, A.N.; Dichi, I. Beneficial effects of fish oil and cranberry juice on disease activity and inflammatory biomarkers in people with rheumatoid arthritis. Nutrition 2021, 86, 111183. [Google Scholar] [CrossRef]
  109. Cannarella, L.A.T.; Mari, N.L.; Alcântara, C.C.; Iryioda, T.M.V.; Costa, N.T.; Oliveira, S.R.; Lozovoy, M.A.B.; Reiche, E.M.V.; Dichi, I.; Simão, A.N.C. Mixture of probiotics reduces inflammatory biomarkers and improves the oxidative/nitrosative profile in people with rheumatoid arthritis. Nutrition 2021, 89, 111282. [Google Scholar] [CrossRef]
  110. Zamani, B.; Golkar, H.K.; Farshbaf, S.; Emadi-Baygi, M.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akhavan, R.; Taghizadeh, M.; Memarzadeh, M.R.; Asemi, Z. Clinical and metabolic response to probiotic supplementation in patients with rheu-matoid arthritis: A randomized, double-blind, placebo-controlled trial. Int. J. Rheum. Dis. 2016, 19, 869–879. [Google Scholar] [CrossRef]
  111. Zamani, B.; Farshbaf, S.; Golkar, H.K.; Bahmani, F.; Asemi, Z. Synbiotic supplementation and the effects on clinical and metabolic responses in patients with rheumatoid arthritis: A randomized, double-blind, placebo-controlled trial. Br. J. Nutr. 2017, 117, 1095–1102. [Google Scholar] [CrossRef] [PubMed]
  112. Hong, N.; Ku, S.; Yuk, K.; Johnston, T.V.; Ji, G.E.; Park, M.S. Production of biologically active human interleukin-10 by Bifidobacterium bifidum BGN4. Microb. Cell Factories 2021, 20, 16. [Google Scholar] [CrossRef] [PubMed]
  113. Jeong, Y.; Jhun, J.; Lee, S.-Y.; Na, H.S.; Choi, J.; Cho, K.-H.; Lee, S.Y.; Lee, A.R.; Park, S.-J.; You, H.J.; et al. Therapeutic Potential of a Novel Bifidobacterium Identified Through Microbiome Profiling of RA Patients with Different RF Levels. Front. Immunol. 2021, 12, 736196. [Google Scholar] [CrossRef] [PubMed]
  114. Kheirouri, S.; Hadi, V.; Alizadeh, M. Immunomodulatory Effect of Nigella sativa Oil on T Lymphocytes in Patients with Rheumatoid Arthritis. Immunol. Investig. 2016, 45, 271–283. [Google Scholar] [CrossRef] [PubMed]
  115. Mirtaheri, E.; Khabbazi, A.; Nazemiyeh, H.; Ebrahimi, A.-A.; Hajalilou, M.; Novin, Z.S.; Pirouzpanah, S. Stachys schtschegleevii tea, matrix metalloproteinase, and disease severity in female rheumatoid arthritis patients: A randomized controlled clinical trial. Clin. Rheumatol. 2022, 41, 1033–1044. [Google Scholar] [CrossRef] [PubMed]
  116. Sun, Y.; Liu, J.; Wan, L. Effect of Xinfeng Capsule on Improving Pulmonary Function in Rheumatoid Arthritis Patients. Zhongguo Zhong xi yi jie he za zhi Zhongguo Zhongxiyi jiehe zazhi Chin. J. Integr. Tradit. West. Med. 2016, 36, 814–820. (In Chinese) [Google Scholar]
  117. Hashemi, G.; Mirjalili, M.; Basiri, Z.; Tahamoli-Roudsari, A.; Kheiripour, N.; Shahdoust, M.; Ranjbar, A.; Mehrpooya, M.; Ataei, S. A Pilot Study to Evaluate the Effects of Oral N-Acetyl Cysteine on Inflammatory and Oxidative Stress Biomarkers in Rheumatoid Arthritis. Curr. Rheumatol. Rev. 2019, 15, 246–253. [Google Scholar] [CrossRef]
  118. Prescha, A.; Zabłocka-Słowińska, K.; Płaczkowska, S.; Gorczyca, D.; Łuczak, A.; Grajeta, H. Silicon intake and plasma level and their relationships with systemic redox and inflammatory markers in rheumatoid arthritis patients. Adv. Clin. Exp. Med. 2019, 28, 1485–1494. [Google Scholar] [CrossRef] [Green Version]
  119. Shishavan, N.G.; Gargari, B.P.; Jafarabadi, M.A.; Kolahi, S.; Haggifar, S.; Noroozi, S. Vitamin K1 Supplementation Did Not Alter Inflammatory Markers and Clinical Status in Patients with Rheumatoid Arthritis. Int. J. Vitam. Nutr. Res. 2018, 88, 251–257. [Google Scholar] [CrossRef]
  120. Lepetsos, P.; Papavassiliou, A.G. ROS/oxidative stress signaling in osteoarthritis. Biochim. Biophys. Acta Mol. Basis Dis. 2016, 1862, 576–591. [Google Scholar] [CrossRef]
  121. Qamar, N.; John, P.; Bhatti, A. Emerging role of selenium in treatment of rheumatoid arthritis: An insight on its antioxidant properties. J. Trace Elem. Med. Biol. 2021, 66, 126737. [Google Scholar] [CrossRef] [PubMed]
  122. Bhattacharjee, A.; Basu, A.; Ghosh, P.; Biswas, J.; Bhattacharya, S. Protective effect of Selenium nanoparticle against cyclophosphamide induced hepatotoxicity and genotoxicity in Swiss albino mice. J. Biomater. Appl. 2014, 29, 303–317. [Google Scholar] [CrossRef] [PubMed]
  123. Kang, E.; Kim, H.J.; Choi, J.H.; Noh, J.; Kim, J.; Lee, I.B.; Choi, Y.; Choi, D.; An, J.; Oh, W.K.; et al. Humulus japonicus extract ameliorates collagen-induced arthritis in mice through regulation of overall articular inflammation. Int. J. Mol. Med. 2020, 45, 417–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Zhang, Z.; Zhang, Q.; Yang, H.; Liu, W.; Zhang, N.; Qin, L.; Xin, H. Monotropein isolated from the roots of Morinda officinalis increases osteoblastic bone formation and prevents bone loss in ovariectomized mice. Fitoterapia 2016, 110, 166–172. [Google Scholar] [CrossRef] [PubMed]
  125. Jiang, F.; Xu, X.; Li, W.; Xia, K.; Wang, L.; Yang, X. Monotropein alleviates H2O2-induced inflammation, oxidative stress and apoptosis via NF-κB/AP-1 signaling. Mol. Med. Rep. 2020, 22, 4828–4836. [Google Scholar] [CrossRef]
  126. Shi, Y.; Liu, X.-Y.; Jiang, Y.-P.; Zhang, J.-B.; Zhang, Q.-Y.; Wang, N.-N.; Xin, H.-L. Monotropein attenuates oxidative stress via Akt/mTOR-mediated autophagy in osteoblast cells. Biomed. Pharmacother. 2020, 121, 109566. [Google Scholar] [CrossRef]
  127. Puppala, E.R.; Jain, S.; Saha, P.; Rachamalla, M.; Np, S.; Yalamarthi, S.S.; Abubakar; Chaudhary, A.; Chamundeswari, D.; Usn, M.; et al. Perillyl alcohol attenuates rheumatoid arthritis via regulating TLR4/NF-κB and Keap1/Nrf2 signaling pathways: A comprehensive study onin-vitro and in-vivo experimental models. Phytomedicine 2022, 97, 153926. [Google Scholar] [CrossRef]
  128. An, L.; Li, Z.; Shi, L.; Wang, L.; Wang, Y.; Jin, L.; Shuai, X.; Li, J. Inflammation-Targeted Celastrol Nanodrug Attenuates Collagen-Induced Arthritis through NF-κB and Notch1 Pathways. Nano Lett. 2020, 20, 7728–7736. [Google Scholar] [CrossRef]
  129. 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-κB-NLRP3 inflammasome axis. Int. Immunopharmacol. 2021, 98, 107879. [Google Scholar] [CrossRef]
  130. Li, J.; Liu, R.; Zhang, P.; Li, J.; Yue, Y.; Hu, Y.; Cheng, W.; Pan, X. Genistein suppresses tumor necrosis factor α-induced inflammation via modulating reactive oxygen species/Akt/nuclear factor ΚB and adenosine monophosphate-activated protein kinase signal pathways in human synoviocyte MH7A cells. Drug Des. Dev. Ther. 2014, 8, 315–323. [Google Scholar] [CrossRef] [Green Version]
  131. Liu, Z.; Guo, S.; Dong, Q. Nobiletin suppresses IL-21/IL-21 receptor-mediated inflammatory response in MH7A fibroblast-like synoviocytes (FLS): An implication in rheumatoid arthritis. Eur. J. Pharmacol. 2020, 875, 172939. [Google Scholar] [CrossRef] [PubMed]
  132. Sundaram, M.S.; Neog, M.K.; Rasool, M.; Kumar, G.S.; Hemshekhar, M.; Kemparaju, K.; Girish, K.S. Guggulipid ameliorates adjuvant-induced arthritis and liver oxidative damage by suppressing inflammatory and oxidative stress mediators. Phytomedicine 2019, 64, 152924. [Google Scholar] [CrossRef] [PubMed]
  133. Mythilypriya, R.; Shanthi, P.; Sachdanandam, P. Restorative and synergistic efficacy of Kalpaamruthaa, a modified Siddha preparation, on an altered antioxidant status in adjuvant induced arthritic rat model. Chem. Biol. Interact. 2007, 168, 193–202. [Google Scholar] [CrossRef]
  134. Holdiness, M.R. Clinical Pharmacokinetics of N-Acetylcysteine. Clin. Pharmacokinet. 1991, 20, 123–134. [Google Scholar] [CrossRef]
  135. Kanai, T.; Kondo, N.; Okada, M.; Sano, H.; Okumura, G.; Kijima, Y.; Ogose, A.; Kawashima, H.; Endo, N. The JNK pathway represents a novel target in the treatment of rheumatoid arthritis through the suppression of MMP-3. J. Orthop. Surg. Res. 2020, 15, 87. [Google Scholar] [CrossRef] [Green Version]
  136. Pena, L.R.; Hill, D.B.; McClain, C.J. Treatment with Glutathione Precursor Decreases Cytokine Activity. J. Parenter. Enter. Nutr. 1999, 23, 1–6. [Google Scholar] [CrossRef]
  137. Bellezza, I.; Giambanco, I.; Minelli, A.; Donato, R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim. Biophys. Acta Mol. Cell Res. 2018, 1865, 721–733. [Google Scholar] [CrossRef]
  138. Kondo, N.; Netsu, T.; Arai, K.; Kanai, T.; Sano, H.; Kijima, Y.; Okumura, G.; Fujisawa, J.; Endo, N. Nuclear factor-erythroid 2-related 2 mRNA and protein are highly expressed in the synovium of patients with rheumatoid arthritis. Internet J. Rheumatol. Clin. Immunol. 2017, 5, OA4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Bennett, B.L.; Sasaki, D.T.; Murray, B.W.; O’Leary, E.C.; Sakata, S.T.; Xu, W.; Leisten, J.C.; Motiwala, A.; Pierce, S.; Satoh, Y.; et al. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc. Natl. Acad. Sci. USA 2001, 98, 13681–13686. [Google Scholar] [CrossRef] [Green Version]
  140. Jamali, F.; Ahmadzadeh, A.; Sahraei, Z.; Salamzadeh, J. Study of the Effects of N-acetylcysteine on Inflammatory Biomarkers and Disease Activity Score in Patients with Rheumatoid Arthritis. Iran. J. Allergy, Asthma Immunol. 2021, 20, 574–583. [Google Scholar] [CrossRef] [PubMed]
  141. Batooei, M.; Tahamoli-Roudsari, A.; Basiri, Z.; Yasrebifar, F.; Shahdoust, M.; Eshraghi, A.; Mehrpooya, M.; Ataei, S. Evaluating the Effect of Oral N-acetylcysteine as an Adjuvant Treatment on Clinical Outcomes of Patients with Rheumatoid Arthritis: A Randomized, Double Blind Clinical Trial. Rev. Recent Clin. Trials 2018, 13, 132–138. [Google Scholar] [CrossRef] [PubMed]
  142. Atlas, D. Emerging therapeutic opportunities of novel thiol-amides, NAC-amide (AD4/NACA) and thioredoxin mimetics (TXM-Peptides) for neurodegenerative-related disorders. Free Radic. Biol. Med. 2021, 176, 120–141. [Google Scholar] [CrossRef] [PubMed]
  143. Jastrzębska, J.; Frankowska, M.; Filip, M.; Atlas, D. N-acetylcysteine amide (AD4) reduces cocaine-induced reinstatement. Psychopharmacology 2016, 233, 3437–3448. [Google Scholar] [CrossRef] [PubMed]
  144. Vamanu, E. Polyphenolic Nutraceuticals to Combat Oxidative Stress Through Microbiota Modulation. Front. Pharmacol. 2019, 10, 492. [Google Scholar] [CrossRef] [Green Version]
  145. Mazon, J.N.; de Mello, A.H.; Ferreira, G.K.; Rezin, G.T. The impact of obesity on neurodegenerative diseases. Life Sci. 2017, 182, 22–28. [Google Scholar] [CrossRef]
  146. Vamanu, E.; Rai, S. The Link between Obesity, Microbiota Dysbiosis, and Neurodegenerative Pathogenesis. Diseases 2021, 9, 45. [Google Scholar] [CrossRef]
  147. Uyar, G.Ö.; Yildiran, H. A nutritional approach to microbiota in Parkinson’s disease. Biosci. Microbiota Food Health 2019, 38, 115–127. [Google Scholar] [CrossRef] [Green Version]
  148. Horta-Baas, G.; Romero-Figueroa, M.D.S.; Montiel-Jarquín, A.J.; Pizano-Zárate, M.L.; García-Mena, J.; Ramírez-Durán, N. Intestinal Dysbiosis and Rheumatoid Arthritis: A Link between Gut Microbiota and the Pathogenesis of Rheumatoid Arthritis. J. Immunol. Res. 2017, 2017, 4835189. [Google Scholar] [CrossRef] [Green Version]
  149. Zhang, X.; Zhang, D.; Jia, H.; Feng, Q.; Wang, D.; Liang, D.; Wu, X.; Li, J.; Tang, L.; Li, Y.; et al. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat. Med. 2015, 21, 895–905. [Google Scholar] [CrossRef]
  150. Dourado, E.; Ferro, M.; Guerreiro, C.S.; Fonseca, J. Diet as a Modulator of Intestinal Microbiota in Rheumatoid Arthritis. Nutrients 2020, 12, 3504. [Google Scholar] [CrossRef]
  151. 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] [PubMed]
Figure 1. Bone and joint damage and the generation of reactive oxygen species in an inflamed joint affected by rheumatoid arthritis. Abbreviations: RA, rheumatoid arthritis; GSH, reduced glutathione; GSSH, oxidized glutathione; NADPH, nicotine amide adenine dinucleotide phosphate; SOD, superoxide dismutase.
Figure 1. Bone and joint damage and the generation of reactive oxygen species in an inflamed joint affected by rheumatoid arthritis. Abbreviations: RA, rheumatoid arthritis; GSH, reduced glutathione; GSSH, oxidized glutathione; NADPH, nicotine amide adenine dinucleotide phosphate; SOD, superoxide dismutase.
Cimb 45 00197 g001
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

Kondo, N.; Kanai, T.; Okada, M. Rheumatoid Arthritis and Reactive Oxygen Species: A Review. Curr. Issues Mol. Biol. 2023, 45, 3000-3015. https://doi.org/10.3390/cimb45040197

AMA Style

Kondo N, Kanai T, Okada M. Rheumatoid Arthritis and Reactive Oxygen Species: A Review. Current Issues in Molecular Biology. 2023; 45(4):3000-3015. https://doi.org/10.3390/cimb45040197

Chicago/Turabian Style

Kondo, Naoki, Tomotake Kanai, and Masayasu Okada. 2023. "Rheumatoid Arthritis and Reactive Oxygen Species: A Review" Current Issues in Molecular Biology 45, no. 4: 3000-3015. https://doi.org/10.3390/cimb45040197

Article Metrics

Back to TopTop