Preprint
Review

The Role of Dietary Phenolic Compounds in the Prevention and Treatment of Rheumatoid Arthritis: Current Reports

This version is not peer-reviewed.

Submitted:

08 April 2024

Posted:

08 April 2024

You are already at the latest version

A peer-reviewed article of this preprint also exists.

Abstract
Rheumatoid arthritis (RA) is a complex illness with both hereditary and environmental compo-nents. Globally, in 2019, 18 million people had RA. RA is characterised by persistent inflammation of the synovial membrane that lines the joints, cartilage loss, and bone erosion. Phenolic molecules are the most prevalent secondary metabolites in plants, with a diverse spectrum of biological ac-tions that benefit functional meals and nutraceuticals. These compounds have received a lot of at-tention recently because they have antioxidant, anti-inflammatory, immunomodulatory, and anti-rheumatoid activity by modulating tumour necrosis factor, mitogen-activated protein kinase, nu-clear factor kappa-light-chain-enhancer of activated B cells, and c-Jun N-terminal kinases, as well as other preventative properties. This article discusses dietary polyphenols, their pharmacological properties, and innovative delivery technologies for the treatment of RA, with a focus on their possible biological activities. Nonetheless, commercialization of polyphenols may be achievable only after confirming their safety profile and completing successful clinical trials.
Keywords: 
Subject: 
Medicine and Pharmacology  -   Dietetics and Nutrition

1. Introduction

There exist more than 100 conditions of arthritis known, and amongst them, rheumatoid arthritis (RA) is one of the most common forms observed in elderly population [1]. RA affects 0.1–2.0% of the population worldwide, being three-times more common in the female gender. The condition can begin at any age, although around 80% of all individuals begin the disease between the 35 and 50 years old. Their etiology remains poorly understood, and despite recent therapeutic advances, there is no known cure [2].
RA is a systemic autoimmune, chronic, heterogeneous, and inflammatory disease in which your immune system mistakenly attacks healthy cells in the body, reducing quality of life and shortening its duration [3,4]. RA is a complex process involving numerous inflammatory mediators, and is characterized primarily by aggressive synovial hyperplasia, synovitis, progressive cartilage degeneration, and bone erosion with painful swelling of small joints, fatigue, prolonged stiffness, and fever produced by immune responses and particular innate inflammatory processes [4,5]. Although RA affects multiple body systems, the joints of hands, wrists, feet, ankles, knees, shoulders and elbows are most often affected [6]. The lining of the joint becomes inflamed in RA joints, causing joint tissue destruction. Long-term or chronic discomfort, unsteadiness (loss of balance), and deformity (misshapenness) can result from this tissue injury. RA can also affect other tissues and organs, including the lungs, heart, and eyes [1,7].
Although RA changes have been extensively investigated, there is a still lack of effective drugs. During RA, there are many pro-inflammatory cytokines, such as interleukin (IL)-1, IL-6, IL-17 and TNF-α, and chemokines released to the synovial space [4]. Therefore, it is urgent to find effective drugs for RA by inhibiting the production and release of pro-inflammatory cytokines. Until date, non-steroidal anti-inflammatory agents and disease-modifying anti-rheumatic drugs, like immunosuppressants and monoclonal antibodies (e.g., rituximab and tocilizumab) combined with methotrexate, are largely used to attenuate RA; however, most of them are expensive, rarely effective and tolerable and lead to several unwanted effects, including allergy and infections, gastrointestinal problems, fluid retention, renal dysfunction and systemic vasculitis, cytopenia, lymphopenia, neutropenia and the elevation of transaminase and cholesterol [7,8,9,10,11,12,13]. Among the new and most promising availability strategies, special attention has been given to bioactive molecules largely found in nature, namely polyphenolics, since their chemical structure with catechol methoxy and pyrogallol groups, confers them notable health benefits, like antioxidant, anti-inflammatory and antiproliferative activities, as well as capacity to prevent neurological and cardiovascular pathologies, without, it is believed, side-effects [14,15,16,17,18,19,20,21]. In addition, polyphenolics are also easier to obtain, water-soluble and more economical than chemical drugs, and have already shown effectiveness in inhibiting JAK proteins and JAK/STAT signalling pathways [22,23,24,25,26]. Particularly, curcumin displayed capability to relieve the rheumatoid arthritis progression by suppressing the inflammatory responses, synovial hyperplasia and protein expression levels of phosphorylated JAK2 and STAT3 in mice with collagen-induced arthritis model that were treated at a dose of 100 µM curcumin/day for almost three months [27], while, by docking studies, salvianolic acid C exhibited a great affinity binding for JAK (10.7 kcal/mol) [22]. In addition, phenolic-rich extracts of sweet cherries have already displayed a notable ability to scavenge cellular nitric oxide species and diminish inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2 expression [28].
Keeping these facts in mind, the main goal of this work is to review and discuss the dietary polyphenolics commonly found in diet and their rich sources in preventing and/ or attenuating RA, the most advanced strategies and also suggest future search.

2. Data Collection

The data collection was searched from the time of their establishment to March 24th, 2024, through the scientific databases National Center for Biotechnology Information (NCBI), Google Scholar, PubMed, ResearchGate, Science Direct, Scopus, SpringerLink, Web of Science, and trusted abstracts. The free terms, keywords, or MeSH terms used were polyphenols, phenolics, total phenolics content, phenolic-rich sources, antioxidant effects, anti-inflammatory properties, health benefits and rheumatoid arthritis combined with AND, OR, or NOT operators. During the literature review, there were no restrictions on the author(s) or type of publication. In the final, 325 papers were cited in this review.

3. Major Sources of Dietary Polyphenols

Nowadays, the search for more effective, safer and cheaper bioactive compounds than chemical drugs assume great importance. Indeed, and once most chemical drugs possess several unwanted effects and are little active, many people prefer to use natural molecules [29]. Among the most promising molecules, phenolic compounds, including simple phenols (e.g., catechol, phenol, phloroglucinol and resorcinol) and polyphenolics (e.g., coumarins, anthocyanins, lignans and phenolic acids) seem to be promising therapeutic and/or adjuvant approaches (Figure 1) [30,31,32]. Simple phenolics present a low molecular weight, being rapidly absorbed by human skin, while polyphenolics are largely widespread and ubiquitous in nature, being largely found in several algae, beans, fruits, barks, herbs and spices, legumes, leaves, nuts, whole grains, oilseeds and roots (Table 1) [33,34].
In a general way, stilbenes are predominant in grapevines, peanuts and sorghums (amounts of 0.16-0.77 mg per 100 g, 1.8-787.3 µg per 100 g and up 0.01 mg per 100 g, respectively), showing total polyphenol compounds (TPC) of 9.3-75.3, 94.4-228.4 and 100-2300 mg GAE per 100 g fresh weight (fw), respectively [35,36,37,38]. Lignans are highly present in flaxseeds (amounts of 9 to 30 mg per g, being 75 to 800 times higher than in cereals, fruits, legumes and vegetables), revealing TPC scores around 3000 mg per 100 g [39,40]; in addition, flaxseeds also present considerable quantities of phenolic acids (800-1000 mg per 100 g) [41].
Focusing on phenolic acids, namely the hydroxybenzoic ones, they are mainly found in black radish, onions, oak bark, and tea, gall nuts, sumac, grapes and wine, while hydroxycinnamic acids are largely present in fruits, vegetables and coffee [42,43,44,45].
Focusing on anthocyanins, they are widely found in red and purple fruits and vegetables, being inclusive considered the main responsible for their vibrant colours and health-promoting properties exhibited by them. Specifically, cyanidin 3-O-rutinoside is mainly found in sweet cherries, while cyanidin 3-O-glucosyl-rutinoside is the predominant anthocyanin in sour cherries [46,47]. Regarding TPC values, sour cherries present higher values than the sweet ones [275.3-652.27 against 72.9-493.6 mg gallic acid equivalents (GAE) per 100 g fw for sweet cherries] [47,48,49]. On the other hand, the TPC of blueberries fluctuates between 2.7 and 585.3 mg GAE per 100 g fw, being richer in peonidin derivatives, whereas for black elderberries, TPC is around 537.9 mg GAE per 100 g fw and cyanidin 3-O-sambubioside is the main anthocyanin [50,51,52,53]. Blackberries also present considerable amounts of TPC (292.2-446.4 mg GAE per 100 g fw), being cyanidin 3-glucoside, the major anthocyanin [54,55]. For strawberries, TPC varies from 36.5 to 116.3 mg GAE per 100 g fw, being the most abundant, pelargonidin 3-O-glucoside [56,57,58], while for grapevines (TPC 9.3-75.3 mg per 100 g fw), malvidin 3-O-glucoside is the predominant anthocyanin [38,59,60]. Additionally, mulberries and chokeberries also present considerable amounts of TPC (424-485 and 1022.4-1705.9 mg GAE per 100 g fw, respectively), being cyanidin 3-sophoroside and cyanidin 3-O-galactoside, the predominant ones, respectively [61,62,63], whereas black currants are richer in delphinidin 3-rutinoside and present TPC values fluctuating from 1930.0 to 3410.0 mg GAE per 100 g fw [64,65]. Red cabbage also presents considerable TPC scores (115.31 mg per 100 g fw), being the most prevalent anthocyanin, cyanidin 3-O-diglucoside-5-O-glucoside [66,67].
Isoflavones (e.g., genistein and daidzein) are abundant in soybeans (total isoflavones and TPC values of 80.7-213.6 mg per 100 g, and 87.2-216.3 mg per 100 g fw, respectively); within isoflavones, genistein is the most found, followed by daidzein (21.4-78.3 and 15.0-67.4 mg per 100 g, respectively) [68]. Regarding coumarins, marmelosin is highly found in Aegle quince fruits (290 g per 100 g, for TPC values varying from 905.0 to 4900.0 mg GAE per 100 g) [69,70].
Regarding flavan-3-ols, the presence of (+)-catechin was reported in peaches, apricots, apples and green tea, whereas (-)-epicatechin in apricots, sour cherries, apples, cholate, cocoa and green and black tea [71,72,73], and epigallocatechin gallate, in green tea [74]. Flavonols, specially kaempferol, are largely present in leaves, papaya shoots, pumpkins, carrots and black tea [75,76], while Mentha pulegium L. is rich in quercetin [77], whortleberries, lingonberries, chokeberries and cranberries, in myricetin, and strawberries, apples, persimmons, grapes, onions, and cucumbers, in fisetin [78]. Relatively to flavanones’ compounds, higher levels of hesperetin and naringin are found in citrus fruits [79], except eriodictyol is present in Yerba Santa [80]. Regarding flavonones, hesperidin derivatives are also predominant in citrus fruits [81], while honey and propolis present higher levels of pinocembrin [82], and Glycyrrhiza Glabra L. plant parts are rich in liquiritin [83].
Of course, quantity values are greatly influenced by genotype, origin, climate, time of harvest and agricultural and processing techniques, and so on [84].
Table 1. Main polyphenols found in nature and their phenolic-rich sources.
Table 1. Main polyphenols found in nature and their phenolic-rich sources.
Polyphenols Sources References
Non-flavonoids
  Lignans Flaxseeds [85]
  Phenolic acids
    Hydroxybenzoic acids
    Hydroxycinnamic acids

Black radish, onions, and tea
Cereals, coffee, fruits, tea, vegetables and wine
[86,87,88,89]
  Stilbenes Grapevines, peanuts and sorghums [90]
Flavonoids
  Anthocyanins
    Cyanidin
    Delphinidin
    Pelargonidin
    Peonidin
    Petunidin
    Malvidin

Sweet and sour cherries, mulberries, black elderberries, chokeberries and red cabbage
Eggplant, roselle, maqui berries and black currants
Raspberries and strawberries
Cranberries, blueberries and plums
Chokeberries
Acerola, blackberries and grapevines
[46,51,58,64,91,92,93,94,95,96,97,98,99,100,101,102]
  Coumarins
    Umbelliferone
    Esculin

Aegle marmelo
Horse-chestnut barks
[70,103,104]
  Flavan-3-ols
    (+)-Catechin
    (-)-Epicatechin
    Epigallocatechin gallate

Peaches, apricots, apples and green tea
Apricots, sour cherries, apples, cholate, cocoa and green and black teas
Green tea
[71,86,105,106,107]
  Flavonols
    Kaempferol
    Quercetin
    Myricetin
    Fisetin

Onion leaves, papaya shoots, pumpkins, carrots and black tea
Mentha pulegium L.
Whortleberries, lingonberries, chokeberries and cranberries
Strawberries, apples, persimmons, grapes, onions, and cucumbers
[75,77,78,108,109,110,111]
    Flavanones
    Hesperetin
    Naringin
    Eriodictyol

Citrus fruits, namely, grapefruits, tangerines, oranges and lemons
Citrus fruits, namely grapefruits and oranges
Yerba Santa
[112,113,114,115,116]
  Flavonones
    Hesperidin
    Liquiritin
    Pinocembrin

Citrus fruits
Glycyrrhiza Glabra L. leaves and roots
Honey and propolis
[117,118,119,120]
  Isoflavones Soybeans [68,121,122,123,124,125]

4. Pharmacological Properties of Dietary Polyphenols

Knowing the deleterious effects caused by oxidative stress and exacerbated inflammatory responses, it is not surprising that these natural molecules, which are composed of many hydroxyl groups, can easily neutralize radical species or breaking chains, by giving an electron or hydrogen atom, and interfere with inflammatory pathways, and thus, contributing to increase immunity defences and promote a healthy status. In addition, evidence also refers that they are effective in acting as anti-aging, anti-microbial and antiproliferative agents, and possess abilities to increase insulin sensitivity, lower blood sugar levels and ameliorate brain and heart functions, and digestion, too.
As expectable, their benefits are closely linked to their chemical structure and also quantities. Indeed, several studies already reported that, in most cases, an increment in phenolics enhances the biological potential [40,46,48,126,127,128,129,130,131]. Therefore, it is not surprising that their interest is increasing worldwide, specially to prevent and/or attenuate several diseases, especially those without medical cure, like AR (Figure 2). In this disease, there is verified a network of inflammatory components, degrading enzymes, angiogenic molecules and cells. Altogether, this originates immune deregulation, which, without surprise, is not only associated with exacerbated inflammatory responses but also with oxidative stress, affecting particularly with nuclear factor erythroid 2–related factor 2 (Nrf2) pathway (discussed below). Phenolics have been showed potential to counteract oxidative stress levels, and down-regulate exacerbated inflammatory responses, and thus, their use as adjuvant therapy and/or combined with chemical pharmaceutics can be very useful. Similar approaches have been applied in cancer treatment. In fact, in 2019, from the 247 anticancer drugs available, 200 were from natural products, while 38 were synthetic drugs and 9 were vaccines [29].

4.1. Antioxidant Capacity

Reactive oxygen and nitrogen species, namely superoxide and nitric oxide radicals, play a vital part in human body, being signaling molecules and interfering in immune responses and signal transduction pathways, cells growth and gene expression. However, their overproduction and accumulation, have a negative impact on mitochondria and lead to the formation of more toxic free radical species, such as peroxynitrite and peroxyl radicals, promoting deleterious effects in cells, DNA mutations, disintegration and protein damage of membranes, and alterations in phagocyte-mediated activity, playing a pathogenic role in chronic inflammatory diseases. Consequently, inflammation signalling cascades and oxidative stress components are triggered, which in turn, enhance the risk of neuropathologies and chronic diseases, as atherosclerosis, cancer, metabolic syndrome, and of course, RA [133,134].
Regarding RA, its pathophysiology has been linked to oxidative stress, principally with the Nrf-2 pathway. This one is the major pathway involved in the maintenance of homeostatic responses by rising intracellular defence mechanisms and regulating the heme oxygenase-1 axis, control macrophages activation and NF-κB pathway, reducing stress oxidative and inflammation [135]. Under quiescent conditions, Kelch-like ECH-associated protein 1 (Keap 1) holds Nrf2 in the cytoplasm, promoting its ubiquitination and subsequent proteolysis; however, under pathology conditions, Nrf2 is released from Keap1, and goes inside to the nucleus, where happens the transcription of antioxidant enzymes [136,137,138].
Among the most promising therapeutic and adjuvant approaches, polyphenolics assume a prominent interest, since they already showed notable capabilities to restore oxidative stress and inflammation near basal levels, by interact with Keap1 protein, avoiding its linkae with its binding site in Nrf-2 and subsequently, cause the dissociation of Keap1 from Nrf2, leading to the transcriptional activation of Nrf-2, and hence, stimulating intracellular antioxidant enzymes (glutathione, catalase and superoxide dismutase) activity, and, of course, neutralizing oxidative stress [136,137,138].
In a general way, and in order to establish the antioxidant properties of phenolics, three statements were recently proposed, known as Bors criteria (Figure 3). The first one is the most significant, and it refers that, due to hydrogen bonding the presence of a catechol group on B ring, increases the stability of phenolics, and consequently, their antioxidant properties. The second one is the existence of a 2,3 double bond combined with a 4-oxo group on C ring, which in turn, facilitates electron delocalization and allows a better aryloxyl radical stabilization, while the third one is related to the presence of OH groups at positions 3 and 5 on A and C rings combined with a 4-oxo group on C ring, allowing electron delocalization via hydrogen bonds [139]. Thus, these characteristics make phenolics capable of acting as electron and hydrogen donors [140,141]. Among phenolics, quercetin and myricetin complete all Bors criteria, and hence, they are well-known due to their notable capacity to reduce radicals [142]. Moreover, the existence of the catechol group in quercetin and its derivatives enhances their biological potential [139].
Regarding non-flavonoid compounds, hydroxycinnamics showed better antioxidant capacities than hydroxybenzoic acids due to the CH=CH-COOH group, the 7,8-double bond, the carboxyl group with O-alkyl esters and the presence of hydroxyl groups at the ortho and/or para-positions [141,143,144,145]. For lignans, it is well-established their capacity to eliminate radicals and free species is mainly due to the presence of catechol groups [146], while the hydroxylation at position 4 and hydroxyl groups at the ortho position are the main contributors for the antioxidant potential attributed to stilbenes [147].
Comparing with flavonoids, these last ones are more effective in eliminating free species and radicals than non-flavonoids, not only due to the presence of more hydroxyl groups, but also owing to the double bond in positions 2 and 3, the o-diphenolic group and 3’,4’-catechol hydroxyl groups on B ring and the conjugation between the double-bond and the 4-oxo group on C ring [145,148].
Even so, it is important not to forget that catechol and pyrogallol groups also make phenolics vulnerable to autoxidation, particularly when they interact with transition metals; however, the glycosylation and methylation of hydroxyl groups can decline this harmful behaviour [149,150,151,152]. Furthermore, pH also influences the antioxidant properties of phenolics; in fact, and although lower pH levels increase iron-reducing activity, they diminish iron catalytic activity and chelate activities [153].
Focusing on their potential to interact with Nrf-2 pathway, Diniyah and colleagues [154] revealed that catechin, epicatechin and gallic acid from non-oilseed legumes present high binding affinities with Keap1, a protein that activates the degradation of Nrf-2, being, therefore, promising new Nrf2 activators. Similar results were found involving hesperetin, hesperidin, naringenin, naringin, narirutin, neohesperidin, neohesperidin dihydrochalcone and nobiletin citrus-derived flavonoids [155], and sesamol isolated from Sesamum indicum L. seed oil [137]. Mice treated with 37.5, 75, and 150 mg/kg green tea every 8 h by intragastric administration that, were then, sacrificed after 4,12 and 20 h after administration, showed higher levels of antioxidative enzymes and liver phase II enzymes; in addition, docking studies revealed that the most predominant compounds on this tea, which were, caffeine, catechin, catechin gallate, epicatechin, epigallocatechin, epigallocatechin gallate, epicatechin gallate, gallic acid and gallocatechin revealed strong binding affinities with the Keap1 protein [138]. In accordance with the mentioned, the daily intake of 25 mg/kg/body weight (bw) epigallocatechin gallate by rats with lung injury and oxidative stress induced by fluoride displayed capability to increase antioxidant status and Nrf2 gene expression, and reduce inflammatory cytokine [156]. Similar results were obtained using endothelial cells pre-treated with polychlorinated biphenyls 126 [157]. Furthermore, Li et al. [136] verified that concentrations of apigenin, cyanidin 3-sambubioside, echinacoside, luteolin 5-O-glucoside, quercetin 3-O-rutinoside and α−tocopherol at 10 and 50 µM can effectively enhance Nrf-2 levels in the nucleus of hydrogen peroxide−induced oxidative-injured rat adrenal pheochromocytoma PC12 cells. 10-100 µM 5-caffeoylquinic acid also demonstrated to be a potent Nrf-2 activator in human hepatocellular carcinoma HepG2 cells [158]. Finally, Mishra and colleagues revealed that the combination of curcumin (30 mg/kg/bw) with vitamin E (200 mg/kg/bw) could be very effective in counteract oxidative stress in hypothyroid rats with via interfering with NF-kB/AKT/mTOR/KEAP1 [159].
Moreover, positive correlations were already reported between polyphenol amounts and the biological potential. Particularly, positive correlations were already reported between sweet cherry anthocyanin-rich fractions against nitric oxide, superoxide radicals (r> 0.9013) [43], ferric species (r=0.739), and between TPC, and non-colored and colored fractions from sweet cherry fruits and hydrogen peroxide (r> 0.940) [48]. Additionally, a high correlation (r=0.7581) between honey TPC and the elimination of nitric oxide radicals was also found [134,160], and between total flavonoids amounts of sweet cherries vegetal parts and the inhibition of hemoglobin oxidation (r> 0.9636) [161].
Focusing on individual compounds from honey, high correlations were found between caffeoyl hexose, quercetin 7-glucoside-3-O-rutinoside, quercetin derivative and quercetin acetyl rhamnoside and NO scavenging potential (r> 0.7581) and concerning quercetin 3-O-rutinoside and hemoglobin oxidation, hemolysis and lipid peroxidation (r > 0.7355). Additionally, positive correlations were also reported between caffeoyl hexose and quercetin acetyl rhamnoside and lipid peroxidation (r=0.7352 and r=0.755, respectively) [160], whereas a mild correlation (r=0.64) between quercetin content of pollen extracts and protective effects against oxidative injury induced by AAPH on Hepa1-6 hepatic cells was also reported [162]. On the other hand, strong correlations were found involving quercetin 3-O-glucoside (r=0.8640), and ρ-coumaric acid derivative 1 (r=0.9444), ρ-coumaroylquinic (r=0.8646), ρ-coumaric (r=0.8012) and 5-O-caffeoylquinic (r=0.9907) acids of sweet cherries and nitric oxide scavenging test, and also involving this last hydroxycinnamic and superoxide radicals (r=0.9958) [134,163](, as well as between malvidin, delphinidin 3-O-arabinoside and 5-O-caffeoylquinic acid and the antioxidant potential showed by blueberry fruits (r> 0.8689) [163].
Considering all the mentioned data, phenolics seem to help prevent and/or attenuate RA by improving oxidative stress-related chronic diseases, chiefly owing to their interaction with Keap1-Nrf2 complex, activating Nrf-2 transcription. Even so, it is important to highlight that, in many cancer types, the activation of Nrf-2 is predominantly associated with drug resistance, which in turn, diminishes chemotherapy effects and promotes metastatic invasion of cancer cells [164,165,166]. Given that, several attempts have been conducted in order to reduce Nrf-2 levels [167,168]. Considering it, chrysin, luteolin, resveratrol, clofarabine, and 3’,4’,5’,5,7-pentamethoxyflavone already showed to possess promising inhibitory activities, revealing half maximal inhibitory concentrations (IC50) of 10.20, 1.5-40, 15, 15, and 10-400 µM, as well as ability to reduce heme-oxygenase 1 [169,170,171,172]. Moreover, agrimoniin isolated from Agrimonia pilosa Aitch (IC50 values of 100, 200, 300 μM), galloyl glucoses-1,2,3,4,6-penta-O-galloyl-β-D-glucose and 1,3,6-tri-O-galloyl-β-d-glucose isolated from Excoecaria formosana, schisantherin A isolated from Fructus schisandrae (IC50 = 2.5 μM), neferine isolated from leaves of Nelumbo nucifera Gaertn. (IC50=0-20 μM) and wedelolactone isolated from Eclipta prostrate Lour (IC50=2.5–20 μM) are also effective in reducing cancer cells’ proliferation [102,166,168,173,174]. Finally, Castanea crenata Siebold & Zucc, Cinnamomi Cortex, together with procyanidins isolated from it, and rosemary extracts, Chrysanthemum zawadskii and Licorice Glycyrrhiza uralensis, Bergenia ligulata and Rhododendron luteum sweet extracts, and strawberry tree honey also showed similar effects [164,165,167,175,176,177,178,179,180], as well as pterostilbene, a natural dimethoxylated analog of resveratrol [181].

4.2. Anti-Inflammatory Abilities

During inflammation, IL-1 and IL-6, tumor necrosis factor (TNF-α), prostaglandins, heat shock proteins, and nitric oxide and superoxide radicals are generated, which, in part, help in controlling this process by activating neutrophils and macrophages [43]. Hydrogen peroxide is also released, owing to the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) kinases and oxidation, and hence, stopping reactive species accumulation [182,183]. However, and although inflammatory processes are directly linked to individuals’ development, when these responses are exaggerated, carbohydrates damage, lipid peroxidation protein and nucleic acids oxidation happen, contributing to the onset of many disorders, including AR [184,185,186]. Once again, and given phenolics can counteract oxidative stress and normalize immune responses, it is not surprising that phenolics are a target of extensive research. In fact, they already showed capabilities to diminish the activity of COXs, lipoxygenase and iNOS, by interfering with different stages of the inflammatory cascade, namely by down-regulating NF-kB and activating protein-1, and stimulating Nrf-2, mitogen-activated protein kinase (MAPK), a protein complex responsible to modulate a group of protein kinases that play an essential role in signal transduction by modulating gene transcription in the nucleus, and protein kinase-C [187,188,189,190]. In fact, the inhibition of NF-kB is crucial to attenuate inflammatory responses once this complex of proteins is the main responsible for controlling the expression of many genes involved in inflammation, including IL-1, IL-2, IL-6, and TNF-α, COX-2, vascular endothelial growth factor (VEGF), IL-8, MIP-1α, and MCP-1 chemokines, immuno-receptors and adhesion molecules. In this way, phenolics can reduce reduce the severity of inflammation [191,192,193].
Within polyphenols, apigenin, catechin, ellagic acid, epigallocatechin gallate, epigallocatechin gallate, homoplantaginin, luteoloside, quercetin aglycone, quercetin 3-O-rutinoside, allopurinol, resveratrol and tangeretin already showed notable anti-inflammatory effects [157,194,195,196,197,198,199]. Moreover, curcumin, kaempferol 3-O-sophoroside, epigallocatechin-gallate, lycopene and oleanolic acid are effective in inhibiting high mobility group box 1 protein, which is an important chromatin protein involved in the transcription of nucleosomes, transcription factors and histones related to inflammatory processes [200,201,202,203]. Resveratrol can downregulate inflammatory pathway activated by TNF-α in articular chondrocytes [204].
On the other hand, caffeic acid (10 µg/mL), hydroxybenzoic derivatives (25 µg/mL), ρ-coumaric acid (50 µg/mL) and quercetin (100 µg/mL), already showed capabilities to suppress MAPK, JNK1 phosphorylation and NF-κB, and hence, down-regulate the activity of COX-2 and inducible nitric oxide synthase, and consequently, diminish the production of prostaglandins and nitric oxide radicals, respectively [205,206]. In addition, caffeic acid phenethyl ester can also avoid the activation of toll-like receptor (TLR)-4 activation and lipossacharide-mediated NF-κB in macrophages [207].
Catechin and epicatechin (1.7-17.2 µM) can also modulate phorbol 12-myristate 13-acetate-induced NF-kB activation in Jurkat T cells [208]. Particularly, quercetin can inhibit the biosynthesis of leukotrienes in human polymorphonuclear leukocytes, and the activation of NF-kB and p38 MAPK in human mast cells, by preventing the degradation of Iκ Bα and the nuclear translocation of p65, and hence it can reduce IL-1β, IL-6, IL-8 and TNF-α expression [209], as well avoid iNOS gene expression by blocking the activation of IkK kinases, NF-kB and STAT1 in mouse BV-2 microglia [210]. This one can also modulate chromatin remodeling, by blocking the recruitment of CBP/p300 to the promoters of interferon-inducible protein 10 macrophage inflammatory protein-2 genes in primary murine small intestinal epithelial cells [211]. On the other hand, kaempferol and galangin display identic properties concerning epicatechin effects in mouse BV-2 microglia [212,213].
As well as epicatechin, epigallocatechin gallate can also avoid IkK kinases activation, avoiding the degradation of Ik-Bα in culture respiratory epithelial cells and in rat models [214], and blocking DNA binding of NF-κB, and hence, diminishing IL-12 p40 and iNOS expression in murine peritoneal macrophages and J774.1 macrophage cells [187,215]. Furthermore, 10, 25, 50 and 100 µM epigallocatechin gallate also showed capacity to attenuate COX-2 expression, without affecting COX-1, in colon cancer PC-3 cells [216], as well as ability to block NF-κB activation in human epithelial cells and reduces iNOS expression, and consequently, nitric oxide radical levels in macrophages at doses of 5 and 10 µM [217]. Furthermore, and likewise genistein [218], luteolin can also suppress the activation of NF-kB and the expression of proinflammatory genes and IKK kinases in murine macrophages RAW 264.7 and mouse alveolar macrophages, as well as TNF-α secretion in co-cultured intestinal epithelial Caco-2 and RAW 264.7 cells [219,220].
On the other hand, 25, 50, and 100 μM chebulanim, a natural polyphenol acid isolated from Terminalia chebula Retz could inhibit the nuclear translocation of p38 and p65 in liposaccharide-stimulated macrophages, in a dose-dependent manner [185]. Additionally, aqueous birch leaf extract of Betula pendula inhibits the growth and cell division of inflammatory lymphocytes in a dose-dependent manner due to apoptosis induction [221].
Furthermore, hydroxytyrosol and resveratrol, two polyphenols largely abundant in olive oil and red wine can also downregulate NF-kB and the expression of vascular cell adhesion molecule-1 in umbilical veins stimulated with liposaccharide at doses varying between 1 and 100 µM/L [222]. Moreover, anthocyanins, namely cyanidin derivatives (125 µg/mL) demonstrated more efficacy in reducing COX-2 levels than naproxen (10 µM) and ibuprofen (10 µM) (47.4%, 41.3% and 39.8%, respectively) [223]. In silico tools also revealed high energy bindings of cyanidin 3-O-rutinoside (−11.4 kcal/mol), kaempferol 3-O-rutinoside (−10.8 kcal / mol) and cyanidin 3-O-glucoside (−10.1 kcal/mol) with iNOS [148]. Furthermore, homoplantaginin, luteoloside, quercetin, quercetin 3-O-rutinoside, allopurinol and resveratrol already showed potential, both in vitro and in vivo, ability to suppress NLRP3 and/or TLR4 inflammasome activation [194,196,197,198,199].
Relatively to in vivo studies, the daily gestion of cherries (141 g) for 10 days by rats and ringdoves down-regulate IL-1β and TNF-α pro-inflammatory cytokine levels, and raise IL-4 and IL-2 anti-inflammatory cytokines [224]. In addition, gingerenone A (10 mg/kg/bw), a polyphenol largely present in ginger, also displayed positive effects in suppresses obesity and adipose tissue inflammation by avoiding macrophage infiltration and enhance adiponectin, high-fat diet-fed mice which were treated during 15 weeks [225], as well as quercetin [226]. On the other hand, apigenin can stop inflammation in human THP-1-induced macrophages and mouse J 774A, by reducing IL-1β production, via inhibiting the activation of caspase-1 through the disruption of NLRP3 inflammasome, as well as diminishing TNF-α and IL-1β thanks to its ability to inactivate NF-kB [195]. Apigenin, as well as other polyphenol compounds extracted from chamomile, meadowsweet and willow bark, including quercetin and salicylic acid (0-100 μM)also revealed ability to decrease IL-1β, IL-6 and TNF-α in THP-1 macrophages and also protect them against oxidative damage [227]. Tannic acid-based nanogel is also an efficient anti-inflammatory agent, showing a notable potential to reduce neutrophil recruitment and pro-inflammatory cytokines, indicating successful alleviation of inflammation [228].
Moreover, curcumin can easily interfere with NF-κB, as well as with STAT3, reducing the expression of TLR-2 and -4, and upregulating peroxisome proliferator-activated receptor ɣ, as observed in male rats that were treated with 0.2 µM curcumin for 3 days [229,230].
Among fruits, sweet cherry phenolic-rich fractions can reduce nitric oxide radicals and decrease iNOS and COX-2 expression in RAW macrophages stimulated with lipopolysaccharide [28]. Moreover, aqueous and hydroethanolic extracts of their vegetal parts also are effective in inhibiting nitrite levels in a dose-dependent manner in these cell lines [26]. Moreover, some Brazilian plants also showed potential to reduce TNF-𝛼 and CCL2 levels in lipopolysaccharide-stimulated human monocytic THP-1 cells [231]. Regarding individual compounds, ferulic and coumaric acids, which are largely found in Chinese propolis, Lonicera japonica Thunb) and Kalanchoe gracilis showed similar capacities [232,233,234]. In addition, 1-1000 µg/mL quince (Cydonia oblonga Miller) peel polyphenols modulate liposaccharide-induced inflammation in human THP-1-derived macrophages, reducing IL-6 and increasing IL-10 expression via inhibiting NF-κB, p38MAPK and Akt pathways [235].
Focusing on clinical trials, has already been reported that the daily consumption of cherries (280 g) by women can reduce plasma C-reactive protein and nitric oxide radicals, 3 h after intake [236].

4.2.1. Anti-Rheumatoid Effects

As expectable, polyphenols and polyphenolic-rich sources also reveal promising abilities to attenuate, or even prevent, rheumatoid arthritis, as described in Table 2.

In Vitro Studies

Considering all the mentioned until now, it is not surprising that polyphenols can be considered promising molecules in preventing and/or attenuating rheumatoid arthritis.
So far, and focusing on in vitro assays, it has been already reported that slibinin, a natural flavonoid extracted from milk thistle (Silybum marianum) showed effectiveness in inhibiting Th17 cell differentiation and induce macrophage M2 polarization in RAW 264.7 cells, and promotes apoptotic events and inhibits NF-kB, SIRT1 and autophagy in fibroblast-like synoviocytes at doses of 50, 100 and 200 μM [237]. Moreover, 50, 100 and 200 µg/mL oleuropein, the most common polyphenolic detected in olive leaves showed potential to shift CD4+ T cells from peripheral blood mononuclear cells of RA patients toward CD4+CD25+FoxP3 Tregs and induce the production of IL-10 and TGF-β [238]. In addition, 12.5-50 µg/mL polyphenolic extract from extra virgin olive oil inhibits the inflammatory response in IL-1β-activated synovial fibroblasts, as well as TNF-α, IL-6, COX-2 and microsomal PGE synthase-1 production, thanks to their capability to downregulate MAPK and NF-kB signalling pathways [239].
Moreover, 5 and 10 µM curcumin showed effectiveness in reducing survivability, and decreasing levels of MMP1 and TNF-α in synovial sarcoma SW982 cells, considered the best in vitro approach to study RA [240]. Additionally, 25-100 µM curcumin can induce apoptosis and inhibit PGE2, by down-regulating anti-apoptotic Bcl-2 and the X-linked inhibitor of the apoptosis protein and upregulating pro-apoptotic Bax expression, in a concentration-dependent manner, on synovial fibroblasts obtained from patients with RA [241]. In addition, this compound at 12.5-50 µM, also showed capacity to reduce IL-1β, PMA-induced IL-6 and VEGF-A expression, by inhibiting NF-κB and induced dephosphorylation of ERK1/2 and enhancing apoptosis in both in MH7A cells and RA-fibroblast-like synoviocytes [242].
On the other hand, 1, 5 and 25 µM quercetin diminish IL-17-stimulated RANKL production in RA-fibroblasts-like synoviocytes, IL-17-stimulated osteoclast formation and Th17 differentiation, and hence, modulate bone destructive processes in RA [243]. 12.5-100 µM Punicalagin, a natural polyphenol extracted from pomegranate juice, also showed capability to reduce IL-1β, IL-6, IL-8, IL-17A, MMP-1 and MMP-13 in fibroblast-like synoviocytes [244].
Additionally, 25, 50, 100, 250, and 500 µM syringaldehyde, a small polyphenolic compound extracted from Capparis spinosa L. showed potential to diminish CD86, CD40, MHC II and IL-23 expression, and enhance IL-10 expression and antigen phagocytosis on human acute lymphoblastic leukemia T lymphocytes, via inhibiting MAPK/NF-kB signalling pathways [245].
Table 2. Main effects attributed to polyphenolic exposition and daily treatment.
Table 2. Main effects attributed to polyphenolic exposition and daily treatment.
Polyphenolic/ Plant Model Dose Effects References
In vitro studies
Slibinin RAW 264.7 cells 50, 100 and 200 μM Th17 cell differentiation inhibition
NF-kB, SIRT1 and autophagy inhibition
Macrophage M2 polarization induction
Apoptotic events promotion
[237]
Oleuropein Peripheral blood mononuclear cells of RA patient cells 50, 100 and 200 µg/mL ↑ IL-10 and TGF-β
Shift CD4+ T cells from peripheral blood mononuclear cells of RA patients toward CD4+CD25+FoxP3 Tregs
[238]
Extra virgin olive oil Synovial fibroblasts 12.5-50 µg/mL ↓ IL-1β, TNF-α, IL-6, COX-2 and microsomal PGE synthase-1, and MAPK and NF-kB signalling pathways [239]
Curcumin Synovial sarcoma SW982 cells 5 and 10 µM ↓ MMP1 and TNF-α [240]
Quercetin RA-fibroblasts-like synoviocytes 1, 5 and 25 µM ↓ IL-17-stimulated RANKL production
IL-17-stimulated Osteoclast formation
Th17 differentiation
Modulate bone destructive processes in RA
[243]
Punicalagin Fibroblast-like synoviocytes 12.5-100 µM ↓ IL-1β, IL-6, IL-8, IL-17A, MMP-1 and MMP-13 [244]
Syringaldehyde Lymphoblastic leukemia T lymphocytes 25, 50, 100, 250, and 500 µM ↓ CD86, CD40, MHC II and IL-23
↑ IL-10 and antigen phagocytosis
Inhibition MAPK/NF-kB signaling pathways
[245]
Resveratrol Fibroblast-like synoviocytes 1, 3 and 10 µg/mL ↓ Sirt1 protein, MMP1 and MMP13 [246]
Resveratrol Fibroblast-like synoviocytes 20 µM Inhibition of phosphorylation and acetylation of p65, c-Jun, and Fos
↓ COX-2 expression
[247]
Resveratrol Fibroblast-like synoviocytes 1-40 µM ↑ Nrf2-2, heme oxygenase-1, and Bcl-2/Bax, apoptosis
↓ Keap1 expression and ROS, and MDA levels
block NF-κB p6 translocation, Inhibit cell proliferation and migration
[248,249]
Resveratrol RSC-364 cells 25 and 50 µmol/L ↓ Hypoxia-inducible factor-1α and activated phosphorylation of p38 MAPK and c-Jun N-terminal kinase
arrest cells at G0/G1 cell-cycle
↑ apoptosis
[250]
Resveratrol U251 glioma cells 1-100 µM Interference on PI3K/Akt/BAD signalling pathway Inhibition of cells growth and apoptosis [251,252]
Resveratrol Human umbilical vein endothelial cells 20 µM interference on PI3K/AKT and MEK/ERK
Induce FOXO transcriptional activity
Inhibition of cell migration and capillary tube formation
Prevent angiogenesis
[253]
Resveratrol Fibroblast-like synoviocytes 50 µM Block cells at the G2/M stage
↓ TNF-α and S phase cells ratio
Promote serine-threonine kinase-p53 axis, and autophagy
Cells apoptosis
[254]
Resveratrol Human RA synovial MH7A cells 100 and 200 µM ↓ cells viability
Stimulate H2A.X phosphorylation and apoptosis events
Mitochondrial membrane potentials disruption
Stimulate cytochrome c release from the mitochondria to the cytosol
Caspase-3 and caspase-9 activation
Upregulate the expression of the NAD-dependent deacetylase SIRT1 mRNA
Downregulate the expression of the Bcl-X(L) mRNA
Hyperplasia suppression
[255]
Resveratrol Fibroblast-like synoviocytes 200 µM/L Caspase-3 activation
Inhibition of cells proliferation
Induces cell apoptosis
[256]
Resveratrol Fibroblast-like synoviocytes 25-200 µM ↓ ROS and Bax
↑ Bcl-2 levels and apoptotic cells
Regulate the expression of mitochondrial superoxide dismutase
[257]
Resveratrol Fibroblast-like synoviocytes 100 µM ↓ MMP-1, MMP-3, MMP-9, RANKL, osteoprotegrin [258]
Resveratrol Fibroblast-like synoviocytes 100 µM ↓ TNF-α by interfering with SIRT1/cortistatin pathway [259]
Resveratrol Fibroblast-like synoviocytes 100 µM ↑ the expression of genes involved in mitosis, cell cycle, chromosome segregation and apoptosis [260]
Resveratrol Fibroblast-like synoviocytes 5, 15 and 45 mg/kg ↓ IL-1, IL-6, IL-8 and TNF-α
↑ IL-10 and apoptosis
[261]
Resveratrol Fibroblast-like synoviocytes 10 and 20 µM ↓ urban particulate matter-induced COX-2/PGE2 release
Inhibition of the activation of NADPH oxidase/ROS/NF-κB
[262]
Resveratrol Mouse preosteoblastic MC3T3-E1 cells 1, 2, 3 and 5 µM Mediate SIRT-1 interactions with p300
Modulate NF-kB signaling activation
Inhibition of osteoclastogenesis
Prevent bone loss in bone-derived cells
[263]
Resveratrol + methotrexate Synovial mononuclear cells from RA patients 25 µM resveratrol with 0.5 μg/mL methotrexate ↓ monocyte chemoattractant protein 1 levels [264]
Curcumin Fibroblast-like synoviocytes 25-100 µM Induce apoptosis
PGE2 inhibition
Downregulate anti-apoptotic Bcl-2 and the X-linked inhibitor of the apoptosis protein
Upregulate pro-apoptotic Bax expression
[241]
Curcumin Fibroblast-like synoviocytes and MH7A cells 12.5-50 µM ↓ IL-1β, PMA-induced IL-6 and VEGF-A expression, and cells viability
Inhibition of NF-κB and induced dephosphorylation of ERK1/2
↑ apoptosis
[242]
Purified grape-derived compounds 1, 10 and 100 µM Human peripheral blood mononuclear cells ↓ TNF-α, IL1, IL-6 and iNOS genes [100]
Gallotannins Human mast cells 1, 1 and 10 µg/mL Downregulate NF-kB expression [265]
Ellagic acid Fibroblast-like synoviocytes 10, 25, 50 and 100 µM ↓ IL-6, IL-1β, MDA and TNF-α
↑ Superoxide dismutase and apoptosis
[266]
Gallic acid Fibroblast-like synoviocytes 0.1 and 1 µM ↑ caspase-3 activity
Regulate Bcl-2, Bax, p53 and pAkt productions
↓ IL-1β, IL-6, CCL-2/MCP-1, CCL-7/MCP-3, COX-2, and MMP-9
[267]
Rosmarinic acid nanoparticles Macrophages Not mentioned ↓ RONS and pro-inflammatory cytokines [191]
ρ-Coumaric acid encapsulated with mannosylated liposomes Macrophages Not mentioned ↓ RONS and pro-inflammatory cytokines
Inhibition of osteoclasts differentiation
Downregulate the expression of MMP-9 and NFATc1
[268]
Ferulic acid Fibroblast-like synoviocytes 25-300 µM ↓ IL-17-levels
Inhibition of IL-17/IL-17RA/STAT-3 signalling cascade
[269]
Ferulic acid RAW 264.7 macrophages 25, 50 and 100 µM Attenuate RANKL-induced osteoclast differentiation
↓ bone resorption activity
Downregulate NFATc1, c-Fos, TRAP, Cathepsin K and MMP-9 levels
[270]
Chlorogenic acid T cells c1 10-50 µg/mL Inhibition of osteoclast differentiation and bone resorption
Downregulate RANKL
Suppress mRNA expression of NFATc1, TRAP and OSCAR
[271]
Tea polyphenol carrier-enhanced dexamethasone Umbilical vein endothelial, murine fibroblast cells L929 and murine macrophage RAW 264.7 cells Not mentioned ↓ inflammatory [272]
Tinospora cordifolia RAW 264.7 cells 100, 250 and 500 µg/mL ↓ IL-6, TNF-α, PGE2, and NO, and iNOS and COX
Modulate JAK/STAT pathway
[273]
Blueberry polyphenols HIG-82 rabbit synoviocytes 100-200 µM ↓ TNF-α, IL-1β, MMP3 and NF-kB levels [274]
Cocoa polyphenols Mouse epidermal cells 10 and 20 µg/mL ↓ TNF-α-induced vascular endothelial growth factor expression
Inhibition PI3K and MEK1
[275]
Catechin-7,4'-O-digallate from Woodfordia uniflora Mouse macrophages 5-80 µM ↓ IL-6 and IL-1β levels
Regulate NF-kB signalling pathway
[276]
Salacia reticulata leaves MTS-C H7 cells IC50 score of ~850 μg/mL Inhibition of cells proliferation [277]
In vivo studies
Slibinin Rats with induced RA 50, 100 and 150 mg/kg ↓ IL-1β, IL-6 and TNF-α levels, and joint inflammation [237]
Resveratrol Rats with induced RA 5 mg/kg, 15 mg/kg and 45 mg/kg ↓ abnormal proliferation of fibroblast-like synoviocytes, swelling degree of the paw and malondialdehyde levels
↑ superoxide dismutase activity, and glutathione peroxidase and glutathione reductase ratio
[278]
Resveratrol Rats with induced RA 10 mg/kg ↓ progression of periodontitis and rheumatoid factor amount [182]
Resveratrol Rats with induced RA 10 mg/kg ↓ Wnt5a, MAPK3, Src kinase, and STAT3 levels [279]
Resveratrol Rats with induced RA 10 mg/kg ↓ IL-6 and TNF-α levels, atrial apoptosis and fibrosis, and activate the AMPK/PGC-1α pathway [280]
Resveratrol Rats with induced RA 10 mg/kg ↓ serum rheumatoid factor, MMP-3, cartilage oligomeric matrix protein, IgG, antinuclear antibody, TNF-α, MPO, C-reactive protein and MDA
↑ IL-10 and glutathione
[281]
Resveratrol Rats with induced RA 50 mg/kg ↓ paw swelling, TNF-α, IL-1β, TBARs, and NOx
Suppress NF-κB p65 expression
[282]
Resveratrol Rabbit inflammatory RA model 10 µMol/kg ↓ inflammatory responses
Prevent the loss of matrix proteoglycan content in the cartilage in
[283]
Resveratrol Murine collagen-induced arthritis 15 and 20 mg/kg Inhibition of Th17 and B-cell function [284]
Resveratrol Rats with bovine type-II collagen-induced arthritis 400 g/kg/bw ↓ oxidative stress and inflammation, and MDA levels
↑ serum superoxide dismutase
Suppress MAPK signalling pathways, and angiogenesis
[250]
Resveratrol Adjuvant arthritis rat model 45 mg/kg ↓ store-operated Ca2+ entry
↑ apoptosis
Interference on ORAI1-STIM1 complex
[285]
Resveratrol Rats with induced RA 12.5 mg/kg Induce the noncanonical autophagy pathway
↓ p62 expression, caspase-3 expression and poly(ADP-ribose) polymerase, IL-1β, C-reactive protein, and prostaglandin E2,and NF-κB synovial tissue expression
[286]
Resveratrol Rats with induced RA 12.5 mg/kg ↓ PCNA, CD68, CD3, monocyte chemoattractant protein-1 staining, cytokine-induced neutrophil chemoattractant-1 and the level of the marker of DNA damage, 8-oxo-7,8-dihydro-2'-deoxyguanine [287]
Resveratrol Collagen-induced arthritis rat model 2.5 and 10 mg/kg Suppress MMP1 and MMP13 amounts [246]
Resveratrol Adjuvant arthritis rats 10 and 50 mg/kg ↓ the proliferation of concanavalin A-stimulated spleen cells, articular cartilage degeneration with synovial hyperplasia and inflammatory cell infiltration
Suppress the production of COX-2 and PGE2
[288]
Resveratrol Rats with induced RA 10 mg/kg Alleviates adjuvant arthritis-interstitial lung disease [289]
Resveratrol Rats with induced RA 10 mg/kg Prevent the production of pro-inflammatory via modulating JAK/STAT/RANKL signalling pathway
Ameliorate fibrosis via autophagy-lysosome pathway
[186]
Resveratrol combined with methotrexate loaded-nanoemulsion Rats with induced RA Not mentioned ↓ inflammation
Better anti-arthritic effects potentiated by resveratrol
[290]
QRu-PLGA-DS nanoparticles carried resveratrol Arthritic rats Not mentioned Improvements the water solubility and targeting the effectiveness of this compound
Ameliorate anti-inflammatory effects
M2 type macrophages transformation
the recruitment of the M1 type macrophages
[291]
Ellagic acid Arthritic rats 5, 50 and 100 mg/kg ↓ oxidative stress and inflammation
↑ serum superoxide dismutase
Suppress MAPK signalling pathways, angiogenesis and MTA1/HDAC1-mediated Nur77 deacetylation
[266]
Ellagic acid Arthritic rats 25 mg/kg ↓ articular edema, NF-kB, and neutrophil elastase, neutrophil extracellular traps
Interference on TLR-4, peptidyl arginine deiminase 4 enzyme and COX-2
[292]
Epigallocatechin gallate Rats with induced RA 10 mg/kg Ameliorate RA symptoms
↓ histological scores in arthritic mice, as well as reduce IgG2a antibodies
Suppress T cell proliferation and relative frequencies of CD4 T cells, CD8 T cells and B cell subsets
↑ the frequency of CD4+-Foxp3+ Treg cells and indoleamine-2,3-dioxygenase expression by CD11b+ dendritic cells, NF-kB, Nrf-2 and heme oxygenase-1
[293]
Epigallocatechin gallate Collagen-induced arthritis rat model 50 mg/kg ↓ TNF-α, IL-17, Nrf-2 and MDA levels
↑ heme oxygenase-1, superoxide dismutase, catalase and glutathione peroxidase levels
[188]
Epigallocatechin gallate Rats with induced RA 10 mg/kg ↓ neuroinflammation, namely by activating caspase-3 [294]
Epigallocatechin gallate Mice with collagen-induced arthritis 50 mg/kg arthritis index
Protective effects against joint destruction
Inhibition of osteoclastogenesis and TH17 cells activation
the number of Treg cells
[295]
Extracellular vesicles-encapsulated epigallocatechin gallate Rats with induced RA Not mentioned Downregulate the expression of hypoxia-inducible factor 1-α
Inhibition apoptosis of chondrocytes
Promote the recovery of type II collagen
joint swelling
[296]
Epigallocatechin Arthritic rats Not mentioned ↑ reduced elastic modulus, hardness and stiffness in cartilage [297]
Epigallocatechin Rats with induced RA 10 mg/kg Prevent cartilage destruction in at, by imbibing myeloperoxidase activity. Moreover, [298]
Green tea Rats with induced RA t 2-12 g/L ↓ RA severity and IL-17 levels
↑ IL-10 levels
Suppress the anti-Bhsp65 antibody response
[299]
Tinospora cordifolia Rats with induced RA 150 mg/kg erythema, paw edema, hyperplasia, IL-6, TNF-α, IL-17, NO and PGE2 levels phosphorylation of STAT3 and the expression of VEGF [273]
Kalpaamruthaa Rats with induced RA 150 mg/kg oxidative stress, myeloperoxidase and lipid peroxide and increase the activity of enzymic and non-enzymic antioxidants [300]
Ribes orientale Sprague Dawley rats with induced RA 50, 100, 200 mg/kg ↓ paw volume/diameter, and PGE2, COX-2, IL-1β, IL-6, NF-kB and TNF-α levels
↑ IL-4 and IL-10
[183]
Chebulanin Collagen-induced arthritis mouse model 80 mg/kg suppress the progression and development of RA
↓ arthritis severity scores, paw swelling and joint destruction, IL-6 and TNF-α amounts, excised phosphorylated (p)-p38 and p-p65, phosphorylated-c-JUN N-terminal kinase and phosphorylated NF-κB and inhibitor alpha
[185]
Punicalagin Rats with induced RA 50mg/kg/ Prevent the translocation of p-65
Avoid the phosphorylation of IkK and Ik Bα, Modulate NF-kB pathway
TNFα, IL-6, CD86, CCR7, CD40 and MHC II expression, Th1, Th17 and Th17/Th1-like
IL-10 expression
Suppress dendritic cells migration, which, in turn
Promote the generation of Tregs via regulation of dendritic cells maturation
[244]
Syringaldehyde Rats with induced RA 10, 25, 50 mg/kg Alleviate paw and joint edema
↓ TNF-α and IL-6 levels
↑ IL-10
[245]
Syringaldehyde Rats with induced RA 100 and 200 mg/kg ↓ IL-6 and TNF-α levels [301]
Clitoria ternatea flower petals and its major compound, quercetin-3ß-D-glucoside Rats with induced RA 50 mg/kg Clitoria ternatea flower petals, and 2.5 mg/kg of quercetin-3ß-D-glucoside MPO activity and pro-inflammatory cytokines, chemokines, RNOS, and TNFR1, TLR2, iNOS, COX-2 and MMP-2 expression levels [302]
Berberis orthobotrys Bien ex Aitch Rats with induced RA 150 mg/kg Protection against arthritic lesions, oxidative damage and body weight alterations*
Ameliorated altered hematological parameters, rheumatoid factor
Contributed to positively modified radiographic and histopathological changes
[303]
Diospyros malabarica (Desr.) Kostel fruits Rats with induced RA 250, 500 and 750 mg/kg ↑ anti-inflammatory enzymes
↓ anti-inflammatory enzymes
[304]
ρ-Coumaric acid Rats with induced RA 100 mg/kg Suppress paw edema and body weight loss ↓ cartel [272]
ρ-Coumaric acid Rats with induced RA 100 mg/kg ↓ age, bone erosion, TNF-α, IL-1β, IL-6, IL-17 and MCP-1, and the expression of RANKL and TRAP, iNOS and COX-2, JNK, p-JNK, and ERK1/2
Regulate the RANKL/OPG imbalance
Inhibition the RANKL-induced NFATc-1 and c-Fos expression
[192,305]
Chlorogenic acid Rats with induced RA 10 mg/kg Attenuate liposaccharide-induced bone loss of rat femurs [271]
Theaflavin-3, 3'-digallate Collagen-induced RA mouse model 10 mg/kg IL-1β, TNF-α, IL-6, as well as MMP-1, MMP-2, and MMP-3 amounts
Inhibition the activation of NF-kB and the phosphorylation of P38, JNK2, and ERK
[184]
Cinnamtannin D1 Rats with induced RA 50 mg/kg Alleviate the severity of RA
↓ clinical scores and paw swelling, inflammatory cell infiltration, cartilage damage in the joints, and IL-17, IL-6, and IL-1β levels, and the frequency of Th17 cells
↑ TGF-β and IL-10 levels and the frequence of Treg cells
Inhibition of aryl hydrocarbon receptor expression and phospho-STAT3/RORγt
[306]
Cinnamon barks Mice with induced RA 200 mg/kg ↓ paw volume, weight loss, and IL-2, IL-4 and IFNγ levels [307]
N-feruloylserotonin Rats with induced RA 3mg/kg ↓ C-reactive protein, the activity of LOX, as well as mRNA transcription of TNF-α, iNOS IL-1β and IL-1β mRNA expression [308]
Extra virgin olive oil Mice with collagen-induced RA 100 and 200 mg/kg ↓ inflammatory markers, joint edema, cell migration, cartilage degradation and bone erosion, and also reducing COX-2 and microsomal prostaglandin E synthase-1 expression
Inhibition c-Jun N-terminal kinase, p38, signal transducer and activator of transcription-3
[309]
Hydroxytyrosol acetate Mice with collagen-induced RA 0.05% ↓ IgG1 and IgG2a, COMP, MMP-3, TNF-α, IFN-γ, IL-1β, IL-6 and IL-17A, and MAPKs JAK/STAT and NF-κB pathways
↑ Nrf-2 and heme oxygenase-1
[310]
mangiferin Mice with induced RA 50, 100 and 400 mg/kg Inhibition of mRNA expression of cytokine genes in thymus and spleen, and also NF-κB and activating ERK1/2
IL-1β, IL-6, TNF-α, and RANKL
[311]
Sarcococca saligna Rats with induced RA 250 mg/kg ↓ IL-1β, IL-6, COX-2, prostaglandin E2, TNF-α and NF-κB levels, arthritic index and paw inflammation
↑ IL-4 and IL-10 levels
[190]
Curcumin Rats with induced RA 10 mg/kg ↓ TNF-α and IL-1β [312]
Dichrostachys cinerea fruits Rats with induced RA 75.48 mg ↓ IL-1β, IL-6, TNF-α and cortisol levels, lipid peroxidation and NOx production [313]
Circaea mollis Sieb. & Zucc. plant Freund's complete adjuvant-induced arthritis model in rats 170-1350 mg/kg ↓ paw and inflammatory swelling, arthritis index, TNF-α and IL-1β levels
↑ IL-10 levels
[314]
Opuntia littoralis Rats with induced RA 10 and 20 mg/100 g bw ↓ joint inflammation, paw swelling, edemas, MDA, and IL-1β, IL-6R, IL-6, IL-17, and IL-23,
Ameliorated COX-2, NF-kB, STAT-3, PTEN, and RANKL expression
Upregulate the expression of miR-28 and miR-199a
[193]
Antrocaryon micraster seeds Rats with induced RA 25 and 100 mg/kg ↓ cachexia, paw edema, infiltration of inflammatory cells, pannus formation, and synovium damage [315]
Dried plums Transgenic mice with induced-RA + 20% dried plums in the normal diet Protect articular cartilage
↓ synovitis, IL-1β, MCP1, MIP1α, MMP1 and MPP3, and RANKL expression
Repress TNF-induced formation of osteoclasts and mRNA levels of cathepsin K and MMP9
Inhibition of NFATc1 expression and NF-κB activation
[316]
Opuntia monacantha Rats with induced RA 750 mg/kg ↓ paw edema, arthritic score, rheumatoid factor, inflammation, COX-2, IL-6, TNF-α, IL-1, NF-kB, bone erosion and pannus formation
Restore hemoglobin, white blood count and platelets parameters
↑ catalase and superoxide dismutase, IL-4 and IL-10 levels
Inhibition of glutaminase 1 activity
[317]
Solanum nigrum Rats with induced RA 800 mg/kg ↓ paw edema
Restore body weight, hematologic parameters, radiographic and histopathologic alterations
[318]
Quercetin and quercetin-loaded chitosan Rats with induced RA 15 mg/kg quercetin and 10 and 20 mg/kg quercetin-loaded chitosan ↓ TNF-α and IL-6
The nanoencapsulation of quercetin enhances its efficacy
[319]
Grape polyphenols + propolis Female rats with induced RA 1.25 g/kg grape polyphenols mixed with 1.25 g/kg propolis ↓ the intensity of cachexia and alleviate RA scores [320]
Malvidin 3-O-β glucoside Chronic rat adjuvant-induced arthritis with 125 mg/kg ↓ cachexia and arthritic paw scores [100]
Phoenix dactylifera L. seeds Rats with induced RA 30 mg/kg ↓ IL-1β levels, paw edema, erythrocyte sedimentation rate and C-reactive protein [321]
Liposomal drug delivery system for morin Rats with induced RA Not mentioned ↓ TNF-α, IL-1β, IL-6, IL-17, RANKL, STAT-3, p-STAT-3, VEGF, iNOS and NF-kB-p65
↑ osteoprotegerin and murin uptake by rats synovial and spleen macrophages
[189]
Clinical trials
Low-calorie cranberry juice 500 mL/day Women with RA ↓ anti-cyclic citrullinated peptide antibodies levels, pain intensity and swollen joints [322]
Low-calorie cranberry juice + fish oil ω-3 fatty acids 500 mL/day of low-calorie cranberry juice with 3 g of fish oil ω-3 fatty acids People with rheumatoid arthritis ↓ C-reactive protein, erythrocyte sedimentation rate and related-pain [323]
Pomegranate extract 250 mg RA patients ↓ swollen, pain intensity and tender joints, erythrocyte sedimentation rate and morning stiffness
↑ glutathione peroxidase
[324]
Resveratrol 1 g RA patients ↓ joint swelling, tenderness, TNF-α, IL-6, protein C-reactive, MMP-3, erythrocyte sedimentation rate and undercarboxylated osteocalcin [325]
↑: enhance; ↓ diminish; IL: interleukin; ROS: reactive oxygen species; RONS: reactive oxygen and nitrogen species; NO: nitric oxide; TNF: Tumor necrosis factor; MMP: matrix metalloproteinase; SIRT: sirtuin; MDA: malondialdehyde; iNOS: Inducible nitric oxide synthase; VEGF: Vascular endothelial growth factor; STAT: signal transducers and activators of transcription; RANKL: eceptor activator of nuclear factor kappa beta; RA: rheumatoid arthritis; NF-kB: nuclear factor kappa B; MPO: myeloperoxidase; COX: cyclooxygenase; JNK: c-Jun N-terminal kinase; ERK: ; BAX: Bcl-2 associated X protein; Bcl-2: B-cell lymphoma 2; MAPK : mitogen-activated protein kinase; ERK: extracellular signal-regulated kinase.
1, 3 and 10 µg/mL Resveratrol also showed ability to reduce Sirt1 protein, MMP1 and MMP13 expression [246], and, at 20 µM, inhibits the phosphorylation and acetylation of p65, c-Jun, and Fos and reduced the binding to the COX-2 promoter, thereby attenuated the COX-2 expression in fibroblast-like synoviocytes [247]. In addition, this compound at 1-40 µM also showed ability to activate Nrf2-2, heme oxygenase-1, and Bcl-2/Bax, induce apoptosis, reduce Keap1 expression and reative oxigen species and malondialdehyde levels, block NF-κB p6 translocation, inhibit cell proliferation and migration on fibroblast-like synoviocytes, in a dose-dependent manner [248,249]. Moreover, Yang and co-workers [250] demonstrated that 25 and 50 µmol/L resveratrol can reduce hypoxia-inducible factor-1α and activated phosphorylation of p38 MAPK and c-Jun N-terminal kinase in IL-1β-stimulated RSC-364 cells, as well as arrest these cells at G0/G1 cell-cycle and enhance their apoptosis. Similar data was reported by Tian et al. [251,252], who also verified that resveratrol can also interfere PI3K/Akt/BAD signalling pathway, which consequently, also promotes the inhibition of cells growth and apoptosis. In addition to that, 20 µM resveratrol can also interfere not only with PI3K/AKT but also with MEK/ERK pathway, and thus, inducing FOXO transcriptional activity and inhibiting cell migration and capillary tube formation in human umbilical vein endothelial cells, preventing angiogenesis [253]. On the other hand, Li et al. [254] reported that 50 μM resveratrol can block fibroblast-like synoviocytes in RA cells at the G2/M stage and reduce TNF-α levels and S phase cells ratio via promoting the serine-threonine kinase-p53 axis, and autophagy, which, subsequently, consequently lead to cells apoptosis. Moreover, 100 and 200 µM resveratrol, also showed ability to reduce the viability of human RA synovial MH7A cells, by stimulating H2A.X phosphorylation and consequent apoptosis events, disrupte mitochondrial membrane potentials and stimulated cytochrome c release from the mitochondria to the cytosol, activate caspase-3 and caspase-9 but not caspase-8, upregulated the expression of the NAD-dependent deacetylase sirtuin (SIRT) 1 mRNA and downregulated the expression of the Bcl-X(L) mRNA, and hence, suppressing synovial cells hyperplasia [255]. The capacity of resveratrol to activate caspase-3, and consequent inhibits the proliferation of synoviocytes and induces cell apoptosis in synoviocytes in RA has already been reported by Tiang et al. [256]. Furthermore, Wang and colleagues revealed that 25-200 µM of this compound can also reduce mitochondrial reactive oxygen species, Bcl-2 associated X protein (Bax) and increase B-cell-lymphoma-2 (Bcl-2 levels) and apoptotic cells, namely by regulating the expression of mitochondrial superoxide dismutase [257]. On the other hand, a dose of 100 µM resveratrol reduces the expression of MMP-1, MMP-3, MMP-9, RANKL, osteoprotegrin [258], and TNF-α by interfering with sirtuin1/cortistatin pathway [259], as well as increase the expression of genes involved in mitosis, cell cycle, chromosome segregation and apoptosis in RA fibroblast-like synoviocytes [260]. Similar data was obtained by Lu and co-workers [261] who also verified that resveratrol can diminish the expression of IL-1, IL-6, IL-8 and TNF-α, raise the expression of IL-10, as well as the administration of resveratrol together with 5 µM hydrogen peroxide induce fibroblast-like synoviocytes apoptosis probably via mitochondrial dysfunction and endoplasmic reticulum stress. Resveratrol can also diminish urban particulate matter-induced COX-2/PGE2 release in human fibroblast-like synoviocytes by inhibiting the activation of NADPH oxidase/ROS/NF-κB [262]. Finally, 1, 2, 3 and 5 µM resveratrol can mediated SIRT-1 interactions with p300, modulating RANKL activation of NF-kB signaling, inhibiting osteoclastogenesis, and thus, preventing bone loss in bone-derived cells [263].
In addition, it was also already reported that the combination of 25 µM resveratrol with 0.5 μg/mL methotrexate significantly reduced monocyte chemoattractant protein 1 levels in synovial mononuclear cells from RA patients [264].
1, 10 and 100 µM purified grape-derived compounds, whose main compound is malvidin 3-O-β glucoside, showed capacity to inhibit the expression of TNF-α, IL1, IL-6 and iNOS genes from secretion-activated macrophages of human peripheral blood mononuclear cells [100].
1, 1 and 10 µg/mL 1,2,3,4,6-penta-O-galloyl-beta-D-glucose, 1,2,6-tri-O-galloyl-beta-D-allopyanose, and 1,2,3,6-tetra-O-galloyl-beta-D-allopyranose gallotannins also showed potential to downregulate NF-kB expression, in a dose dependent in human mast cells [265].
Focusing on phenolic acids, 10, 25, 50 and 100 µM ellagic acid already showed potential to reduce IL-6, IL-1β, malondialdehyde and TNF-α and raise superoxide dismutase and apoptosis in fibroblast-like synoviocyte MH7A cells pre-treated with TNF-α to induce inflammation by inhibiting metastasis-associated gene 1, which is a component of NF-kB signalling, and a upstream modulator of inflammation and immunologic responses [266]. On the other hand, 0.1 and 1 µM gallic acid increase caspase-3 activity, regulate Bcl-2, Bax, p53 and pAkt productions and reduce IL-1β, IL-6, CCL-2/MCP-1, CCL-7/MCP-3, COX-2, and MMP-9 from on fibroblast-like synoviocytes from patients with RA [267]. More recently, it was reported rosmarinic acid nanoparticles showed a favourable capability in scavenging reactive oxygen and nitrogen species and pro-inflammatory cytokines produced by macrophages [191]. ρ-Coumaric acid encapsulated with mannosylated liposomes showed similar effects, as well as ability to inhibit the osteoclasts differentiation and downregulate the expression of MMP-9 and NFATc1 [268]. Moreover, 25-300 µM ferulic acid showed ability to reduce IL-17-levels in RA fibroblast-like synoviocytes, thanks to its ability to inhibit IL-17/IL-17RA/STAT-3 signalling cascade [269]. In addition, this acid at doses of 25, 50 and 100 µM can attenuate RANKL-induced osteoclast differentiation, and consequently decrease bone resorption activity by downregulating NFATc1, c-Fos, TRAP, Cathepsin K and MMP-9 levels in RAW 264.7 macrophages [270]. On the other hand, 10-50 µg/mL chlorogenic acid can inhibit osteoclast differentiation and bone resorption by down-regulating RANKL on activated T cells c1, as well as by suppressing mRNA expression of NFATc1, TRAP and OSCAR [271].
Tea polyphenol carrier-enhanced dexamethasone showed ability to reduce inflammatory mediators in the LPS/INF-γ-induced inflammatory cell models, including umbilical vein endothelial, murine fibroblast cells L929 and murine macrophage RAW 264.7 cells [272]. Tinospora cordifolia also showed anti-inflammatory effects on lypossacharide-stimulated RAW 264.7 cells, by reducing IL-6, TNF-α, PGE2, and nitric oxide levels and iNOS and COX expression via modulation of JAK/STAT pathway [273]. On the other hand, 100-200 µM blueberry polyphenols can reduce TNF-α, IL-1β, MMP3 and NF-kB levels on HIG-82 rabbit synoviocytes previous stimulated with TNF-α [274]. Moreover, 10 and 20 µg/mL cocoa polyphenols can suppress TNF-α-induced vascular endothelial growth factor expression by inhibiting phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase kinase-1 (MEK1) activities in mouse epidermal cells [275]. 5-80 µM Catechin-7,4'-O-digallate from Woodfordia uniflora can also regulate NF-kB signalling pathway in mouse macrophages and reduce IL-6 and IL-1β levels [276]. On the other hand, Salacia reticulata leaves showed capacity to inhibit MTS-C H7 cells proliferation (IC50 score of ~850 μg/mL) [277].

In Vivo Studies

On the other hand, in RA-mice models, 50, 100 and 150 mg/kg/day slibinin showed ability to diminish IL-1β, IL-6 and TNF-α levels, and joint inflammation [237].
Resveratrol revealed to be a promising strategy for attenuating RA in mice model. In fact, a 12-day treatment with 5 mg/kg, 15 mg/kg and 45 mg/kg resveratrol can reduce the abnormal proliferation of fibroblast-like synoviocytes, swelling degree of the paw and malondialdehyde levels, and increase superoxide dismutase activity, and glutathione peroxidase and glutathione reductase ratio [278]. Furthermore, this compound at 10 mg/kg, also showed ability to reduce the progression of periodontitis and rheumatoid factor amount [182], Wnt5a, MAPK3, Src kinase, and STAT3 levels [279], diminish IL-6 and TNF-α levels, atrial apoptosis and fibrosis, and activate the AMPK/PGC-1α pathway [280], as well as decreased to about 37, 59, 44, 70, 5, 30, 23, 33 and 28% serum rheumatoid factor, MMP-3, cartilage oligomeric matrix protein, IgG, antinuclear antibody, TNF-α, myeloperoxidase, C-reactive protein and malondialdehyde, respectively, and enhance to about 225 and 273% IL-10 and glutathione, respectively in RA-rats [281]. Furthermore, 50 mg/kg resveratrol can diminish paw swelling, TNF-α, IL-1β, TBARs, and NOx, namely by suppressing NF-κB p65 expression [282], while 10 µMol/kg can reduce inflammatory responses and prevent the loss of matrix proteoglycan content in the cartilage in a rabbit inflammatory arthritis model [283]. Moreover, a 10-day treatment with 15 and 20 mg/kg resveratrol modulates murine collagen-induced arthritis by inhibiting Th17 and B-cell function [284]. In accordance with other studies, Yang et al. [250] also revealed 400 g/kg/bw resveratrol diminishes oxidative stress and inflammation, by raising serum superoxide dismutase and reducing malonaldehyde, and suppress MAPK signalling pathways, and angiogenesis in rats with bovine type-II collagen-induced arthritis. Similar results were observed in arthritic rats treated with 5, 50 and 100 mg/kg ellagic acid for 21 days, where ellagic acid also showed capacity to suppress MTA1/HDAC1-mediated Nur77 deacetylation [266]. Furthermore, 25 mg/kg of this compound can also decrease articular edema, NF-kB, and neutrophil elastase, and hence, diminishing neutrophil extracellular traps, on RA rats namely by interfering with TLR-4, peptidyl arginine deiminase 4 enzyme and COX-2 [292]. 45 mg/kg Resveratrol can also reduce store-operated Ca2+ entry and enhance the apoptosis in adjuvant arthritis rat model via targeting ORAI1-STIM1 complex [285]. Additionally, Fernández-Rodríguez et al. [286] reported that 12.5 mg/kg/day resveratrol can also induce the noncanonical autophagy pathway, reduces p62 expression, caspase-3 expression and poly(ADP-ribose) polymerase, IL-1β, C-reactive protein, and prostaglandin E2, as well as NF-κB synovial tissue expression, which is correlated with p62 expression [286], as well as diminish PCNA, CD68, CD3, monocyte chemoattractant protein-1 staining, cytokine-induced neutrophil chemoattractant-1 and the level of the marker of DNA damage, 8-oxo-7,8-dihydro-2'-deoxyguanine in RA rats [287]. 2.5 and 10 mg/kg/day resveratrol can also display ability to suppress MMP1 and MMP13 amounts in a collagen-induced arthritis rat model [246], while 10 and 50 mg/kg resveratrol can reduce the proliferation of concanavalin A-stimulated spleen cells, articular cartilage degeneration with synovial hyperplasia and inflammatory cell infiltration, namely by suppressing the production of COX-2 and PGE2 in adjuvant arthritis rats [288]. 10 mg/kg/day Resveratrol alleviates adjuvant arthritis-interstitial lung disease in rats with induced RA, namely by preventing the production of pro-inflammatory via modulating JAK/STAT/RANKL signalling pathway [289], as well as ameliorate fibrosis in rats with induced-RA via autophagy-lysosome pathway [186].
More recently, and to improve resveratrol’s bioavailability and effectiveness, it was shown that its combination with methotrexate loaded-nanoemulsion in a transdermal delivery system resulted in a reduction of inflammation by 78.76% and better anti-arthritic effects [290]. Moreover, the nanocarrier QRu-PLGA-DS nanoparticles effectively improved the water solubility and targeting the ability of this compound to reverse the M1 to the M2 type macrophages and ameliorate its anti-inflammatory effects in arthritic rats; in addition, the accumulation of nanoparticles in the lesion area with an exogenous stimulus, significantly raise the transformation of the M2 type macrophages and decrease the recruitment of the M1 type macrophages [291].
Regarding epigallocatechin gallate, 10 mg/kg of this one can ameliorate clinical symptoms and reduce histological scores in arthritic mice, as well as reduce IgG2a antibodies, suppress T cell proliferation and relative frequencies of CD4 T cells, CD8 T cells and B cell subsets, including marginal zone B cells, T1 and T2 transitional B cells, and increase the frequency of CD4+-Foxp3+ Treg cells and indoleamine-2,3-dioxygenase expression by CD11b+ dendritic cells, NF-kB, Nrf-2 and heme oxygenase-1 [293]. On the other hand, in collagen-induced arthritis rat model, 50 mg/kg/day epigallocatechin gallate also revealed capacity to diminish TNF-α, IL-17, Nrf-2 and malondialdehyde levels and improve heme oxygenase-1, superoxide dismutase, catalase and glutathione peroxidase levels [188]. Moreover, this one can also reduce neuroinflammation in RA rats after 2 months of treatment, namely by activating caspase-3 [294]. Moreover, 50 mg/kg epigallocatechin gallate already showed ability to decrease arthritis index, show protective effects against joint destruction in mice with collagen-induced arthritis, inhibit osteoclastogenesis and TH17 cells activation, and increase the number of Treg cells [295]. On the other hand, the administration of extracellular vesicles-encapsulated epigallocatechin gallate for cartilage repair in rat arthritis reduced down-regulate the expression of hypoxia-inducible factor 1-α, inhibit apoptosis of chondrocytes, promote the recovery of type II collagen and reduce joint swelling in rheumatoid rats by approximately 39.5% [296]. In addition, the nanoindentation of epigallocatechin significantly increases reduced elastic modulus (57.5%), hardness (83.2%), and stiffness (17.6%) in cartilage of arthritic rats [297]. This one also showed capability of preventing cartilage destruction in rats with induced-RA at 10 mg/kg, by imbibing myeloperoxidase activity [298].
Regarding green tea, it was also reported that rats with induced-RA that ingest 2-12 g/L in drinking water for 1-3 weeks showed significant reductions in RA severity, lower levels of IL-17 and higher levels of IL-10, and this is probably due to the capacity of green tea polyphenols to suppress the anti-Bhsp65 antibody response [299].
150 mg/kg Tinospora cordifolia reduced erythema, paw edema, hyperplasia, IL-6, TNF-α, IL-17, nitric oxide and PGE2 levels, as well as phosphorylation of STAT3 and the expression of VEGF [273].
150 mg/kg/bw Kalpaamruthaa, a modified indigenous Siddha preparation constituting Semecarpus anacardium nut milk extract, Emblica officinalis and honey also showed potential to reduce oxidative stress, myeloperoxidase and lipid peroxide and increase the activity of enzymic and non-enzymic antioxidants in arthritic rats [300].
In addition, 50, 100, 200 mg/kg doses of Ribes orientale aqueous ethanolic extract (30:70) showed potential to reduce paw volume/diameter, and PGE2, COX-2, IL-1β, IL-6, NF-kB and TNF-α levels, and enhance IL-4 and IL-10 contents in Sprague Dawley rats with induced RA [183], Additionally, 80 mg/kg daily of chebulanin for 3 weeks significantly suppressed the progression and development of RA in collagen-induced arthritis mouse model, by decreasing the arthritis severity scores, attenuating paw swelling and joint destruction, and reducing IL-6 and TNF-α amounts, excised phosphorylated (p)-p38 and p-p65, phosphorylated-c-JUN N-terminal kinase, and phosphorylated NF-κB inhibitor alpha, but without effects on extracellular-signal-regulated kinase levels [185].
50mg/kg/d Punicalagin also showed capacity to prevent the translocation of p-65 and avoid the phosphorylation of IkK and Ik Bα, by modulating NF-kB pathway in arthritic rats, as well as to reduce TNFα, IL-6, CD86, CCR7, CD40 and MHC II expression, raise IL-10 expression and suppress dendritic cells migration, which, in turn, causes diminish the differentiation of Th1, Th17 and Th17/Th1-like and promoting the generation of Tregs via regulation of dendritic cells maturation [244]. 10, 25, 50 mg/kg Syringaldehyde also showed ability to alleviate paw and joint edema, reduce TNF-α and IL-6 levels and increase the level of IL-10 in arthritic rats’ serum [245]. Lower levels of IL-6 and TNF-α were also obtained by Toy and colleagues at doses of 100 mg/kg and 200 mg/kg/bw [301]. Furthermore, 50 mg/kg Clitoria ternatea flower petals and 2.5mg/kg of its major compound, which is quercetin-3ß-D-glucoside showed ability to reduce MPO activity and low pro-inflammatory cytokines, chemokines, reactive oxygen and nitrogen species, and TNFR1, TLR2, iNOS, COX-2 and MMP-2 expression levels in rats with RA [302]. Similar effects were observed by 150 mg/kg Berberis orthobotrys Bien ex Aitch, which also offered protection against arthritic lesions, oxidative damage and body weight alterations, ameliorated altered hematological parameters, rheumatoid factor and contributed to positively modified radiographic and histopathological changes [303]. 250, 500 and 750 mg/kg Diospyros malabarica (Desr.) Kostel fruits also showed capacity to raise anti-inflammatory enzymes and diminish the anti-inflammatory ones in RA rat model [304].
On the other hand, the daily administration of 100 mg/kg daily ρ-coumaric acid for 2 weeks showed remarkable capacity to suppress paw edema, body weight loss, curtails cartel [272], age, bone erosion, TNF-α, IL-1β, IL-6, IL-17 and MCP-1 quantities in serum and ankle joint of arthritic rats, as well as the expression of RANKL and TRAP, iNOS and COX-2, JNK, p-JNK, and ERK1/2., namely by regulating the RANKL/OPG imbalance and inhibit the RANKL-induced NFATc-1 and c-Fos expression [192]. Similar data was reported by Pragasam and colleagues [305]. Focusing on chlorogenic acid, this one at 10 mg/kg can attenuate liposaccharide-induced bone loss based on micro-computed tomography and histologic analysis of femurs from arthritic rats [271].
10 mg/kg Theaflavin-3, 3'-digallate administrated three times per week 9 continuous weeks also showed capability of reducing the expression of IL-1β, TNF-α, IL-6, as well as MMP-1, MMP-2, and MMP-3 amounts in the synovium of collagen-induced arthritis mouse model, chiefly by inhibiting the activation of NF-kB and the phosphorylation of P38, JNK2, and ERK [184]. Moreover, 50 mg/kg cinnamtannin D1, a polyphenolic compound isolated from Cinnamomum tamala, alleviates the severity of RA, affording reduced clinical scores and paw swelling, and reduced inflammatory cell infiltration, cartilage damage in the joints, IL-17, IL-6, and IL-1β, and enhanced TGF-β and IL-10 levels, as well as reduce the frequency of Th17 cells and enhance the frequency of Treg cells, namely by its potential to inhibit aryl hydrocarbon receptor expression and phospho-STAT3/RORγt [306]. On the other hand, 200 mg/kg cinnamon barks reduce paw volume, weight loss, and, IL-2, IL-4 and IFNγ in RA mice model [307].
3mg/kg/day orally N-feruloylserotonin, a natural polyphenol that belongs to indole hydroxycinnamic acid amides, reduced C-reactive protein in plasma and the activity of LOX in the liver rats, as well as mRNA transcription of TNF-α and iNOS in the liver and IL-1β in plasma and IL-1β mRNA expression in the liver and spleen of arthritic rats [308].
Moreover, 100 and 200 mg/kg extra virgin olive oil showed capability to decrease inflammatory markers, joint edema, cell migration, cartilage degradation and bone erosion in collagen-induced arthritis model mice, namely by inhibiting c-Jun N-terminal kinase, p38, signal transducer and activator of transcription-3, as well as reducing COX-2 and microsomal prostaglandin E synthase-1 expression [309]. Furthermore, 0.05% hydroxytyrosol acetate, a polyphenol present in extra virgin olive oil showed ability to diminish IgG1 and IgG2a, COMP, MMP-3, TNF-α, IFN-γ, IL-1β, IL-6 and IL-17A, and MAPKs JAK/STAT and NF-κB pathways and enhance Nrf-2 and heme oxygenase-1 in mice with collagen-induced arthritis [310].
In addition, 50, 100 and 400 mg/kg mangiferin inhibits mRNA expression of cytokine genes in thymus and spleen of mice with induced-arthritis and lead to decreased serum levels of IL-1β, IL-6, TNF-α, and RANKL by downregulating NF-κB and activating ERK1/2 [311].
250 mg/kg Sarcococca saligna plant also showed ability to diminish IL-1β, IL-6, COX-2, prostaglandin E2, TNF-α and NF-κB levels, arthritic index and paw inflammation, and enhance the expression of IL-4 and IL-10 [190]. On the other hand, 10 mg/kg curcumin showed similar effects as positive control methotrexate (0.5 mg/kg) in reducing TNF-α and IL-1β in both synovial fluid and blood serum of arthritic rats [312]. Aqueous extracts of Dichrostachys cinerea fruits at doses of 150.96 mg GAE/g and 75.48 mg GAE/g reduced IL-1β, IL-6, TNF-α and cortisol levels, lipid peroxidation and NOx production [313]. In addition, 170-1350 mg/kg Circaea mollis Sieb. & Zucc. plant can also reduce paw and inflammatory swelling, arthritis index, TNF-α and IL-1β, and increase the production of serum IL-10 in Freund's complete adjuvant-induced arthritis model in rats [314]. 10 and 20 mg/100 g bw Opuntia littoralis also revealed potential to reduce joint inflammation, paw swelling, edemas, MDA, and IL-1β, IL-6R, IL-6, IL-17, and IL-23, and ameliorated COX-2, NF-kB, STAT-3, PTEN, and RANKL expression, namely by up-regulating the expression of miR-28 and miR-199a [193]. Furthermore, 25 and 100 mg/kg Antrocaryon micraster seed extract also showed capacity to diminish cachexia, paw edema, infiltration of inflammatory cells, pannus formation, and synovium damage in rats with induced RA [315]. Moreover, the addition of 20% dried plums in the normal diet of transgenic mice with induced-RA showed notable protective effects in protecting articular cartilage, reducing synovitis, IL-1β, MCP1, MIP1α, MMP1 and MPP3, and RANKL expression and repressing TNF-induced formation of osteoclasts and mRNA levels of cathepsin K and MMP9 by inhibiting nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) expression and NF-κB activation [316]. On the other hand, 750 mg/kg Opuntia monacantha showed effectiveness in reducing paw edema, arthritic score, rheumatoid factor, inflammation, COX-2, IL-6, TNF-α, IL-1, NF-kB, bone erosion and pannus formation, restore hemoglobin, white blood count and platelets parameteres and increase catalase and superoxide dismutase levels, and enhance IL-4 and IL-10, namely by its potential to inhibit glutaminase 1 activity in a RA rat model [317]. In addition, 800 mg/kg Solanum nigrum also showed effectiveness in reducing paw edema, and restoring body weight, hematologic parameters, radiographic and histopathologic alterations towards normal in different RA rat models [318].
In addition, 15 mg/kg quercetin and 10 and 20 mg/kg quercetin-loaded chitosan showed capacity to reduce TNF-α and IL-6, being observed that the nanoencapsulation of quercetin is an added value to improve its efficacy in rats with induced RA [319]. 1.25 g/kg grape polyphenols mixed with 1.25 g/kg propolis showed potential to diminish the intensity of cachexia and alleviate RA scores in RA female rats [320]. Reduced cachexia and arthritic paw scores were also reported by Decendit [100] who treated chronic rat adjuvant-induced arthritis with 125 mg/kg malvidin 3-O-β glucoside.
Phoenix dactylifera L. seeds can diminish IL-1β levels, paw edema, erythrocyte sedimentation rate and C-reactive protein in rats [321].
Finally, the liposomal drug delivery system for morin, a dietary polyphenol, showed capacity to ameliorate RA in rats, mainly by increasing its uptake by synovial and spleen macrophages, and reducing the mRNA expression and consequent production of TNF-α, IL-1β, IL-6, IL-17, RANKL, STAT-3, p-STAT-3, VEGF, iNOS and NF-kB-p65 and increase the expression of osteoprotegerin [189].

Clinical Trials

Regarding clinical trials, there were already related that 500 mL/day of low-calorie cranberry juice is able to attenuate RA symptoms in women with this disease after a 90-day treatment, by decreasing anti-cyclic citrullinated peptide antibodies levels, pain intensity and swollen joints; however, no inflammatory biomarkers were verified [322]. Additionally, the daily ingestion of 500 mL/day of low-calorie cranberry juice with 3 g of fish oil ω-3 fatty acids showed effectiveness in C-reactive protein, erythrocyte sedimentation rate and related-pain [323]. On the other hand, pomegranate extract (250 mg for 8 weeks), in addition to alleviating swollen, pain intensity and tender joints in RA patients, this one can also diminish erythrocyte sedimentation rate and morning stiffness and enhance glutathione peroxidase. No alterations were found in CRP, MDA and MMP3 levels [324]. Finally, a 1 g daily of resveratrol for 3 months [325] also showed effectiveness in reducing joint swelling and tenderness, as well as TNF-α, IL-6, protein C-reactive, MMP-3, erythrocyte sedimentation rate and undercarboxylated osteocalcin in RA patients of both genders [325].

5. Conclusions

In a general way, the present review is another evidence that supports the therapeutic effect of polyphenolics. Indeed, they seem to be a promising approach to boost immune system and contribute to a healthy status, mainly due to their capacity of counteract oxidative stress and ineract with inflammatory pathways. Given that, it is not surprising that their use as an adjuvant to conventional antirheumatic drugs is possible.
However, and although phenolics are easy to obtain and economical, most of them present poor bioavailability, and therefore, their encapsulation is an added value to increase their efficacy. In addition, their safety profile and successful clinical trials need to be more explored.

Author Contributions

Conceptualization, A.C.G., S.R. and R.F.; methodology, A.C.G., L.R.S.; software, A.C.G., S.R. and R.F..; validation, L.R.S.; formal analysis, A.C.G., S.R., R.F., L.R.S.; investigation, A.C.G., S.R., R.F. L.R.S.; resources, L.R.S.; data curation, A.C.G., S.R., R.F., L.R.S.; writing—original draft preparation, A.C.G.; writing—review and editing, L.R.S.; visualization, A.C.G., S.R., R.F. and L.R.S.; supervision, L.R.S.; project administration, L.R.S.; funding acquisition, L.R.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by CICS-UBI (UIDP/00709/2020) and financed by the National Funds from Fundação para a Ciência e a Tecnologia (FCT) and CENTRO-04-3559-FSE-000162. This work was also financed by project PRR-C05-i03-I-000143 (RedFruit4Health), and by Fundação La Caixa and Fundação para a Ciência e Tecnologia (FCT) under the Programa Promove Project PD21-00023 (PharmaStar). The authors are also grateful to FCT, the Ministry of Science, Technology and Higher Education (MCTES), the European Social Fund (EFS), and the European Union (EU) for the PhD fellowship of Ana C. Gonçalves (2020.04947.BD).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Maity, S.; Wairkar, S. Dietary polyphenols for management of rheumatoid arthritis: Pharmacotherapy and novel delivery systems. Phytother Res 2022, 36, 2324–2341. [Google Scholar] [CrossRef] [PubMed]
  2. Almutairi, K.; Nossent, J.; Preen, D.; Keen, H.; Inderjeeth, C. The global prevalence of rheumatoid arthritis: a meta-analysis based on a systematic review. Rheumatology International 2021, 41, 863–877. [Google Scholar] [CrossRef] [PubMed]
  3. Smolen, J.S.; Aletaha, D.; Barton, A.; Burmester, G.R.; Emery, P.; Firestein, G.S.; Kavanaugh, A.; McInnes, I.B.; Solomon, D.H.; Strand, V.; et al. Rheumatoid arthritis. Nat Rev Dis Primers 2018, 4, 18001. [Google Scholar] [CrossRef] [PubMed]
  4. Giannini, D.; Antonucci, M.; Petrelli, F.; Bilia, S.; Alunno, A.; Puxeddu, I. One year in review 2020: pathogenesis of rheumatoid arthritis. Clin Exp Rheumatol 2020, 38, 387–397. [Google Scholar] [CrossRef] [PubMed]
  5. Silman, A.J.; Pearson, J.E. Epidemiology and genetics of rheumatoid arthritis. Arthritis Research & Therapy 2002, 4, S265. [Google Scholar] [CrossRef] [PubMed]
  6. Long, H.; Liu, Q.; Yin, H.; Wang, K.; Diao, N.; Zhang, Y.; Lin, J.; Guo, A. Prevalence Trends of Site-Specific Osteoarthritis From 1990 to 2019: Findings From the Global Burden of Disease Study 2019. Arthritis & Rheumatology 2022, 74, 1172–1183. [Google Scholar] [CrossRef] [PubMed]
  7. Guo, Q.; Wang, Y.; Xu, D.; Nossent, J.; Pavlos, N.J.; Xu, J. Rheumatoid arthritis: pathological mechanisms and modern pharmacologic therapies. Bone Research 2018, 6, 15. [Google Scholar] [CrossRef] [PubMed]
  8. Schnitzer, T.J.; Burmester, G.R.; Mysler, E.; Hochberg, M.C.; Doherty, M.; Ehrsam, E.; Gitton, X.; Krammer, G.; Mellein, B.; Matchaba, P.; et al. Comparison of lumiracoxib with naproxen and ibuprofen in the Therapeutic Arthritis Research and Gastrointestinal Event Trial (TARGET), reduction in ulcer complications: randomised controlled trial. Lancet (London, England) 2004, 364, 665–674. [Google Scholar] [CrossRef]
  9. Prieto, M.; Niño, A.; Acosta-Guzmán, P.; Guevara-Pulido, J. Design and synthesis of a potential selective JAK-3 inhibitor for the treatment of rheumatoid arthritis using predictive QSAR models. Informatics in Medicine Unlocked 2024, 45, 101464. [Google Scholar] [CrossRef]
  10. Kumar, P.; Banik, S. Pharmacotherapy Options in Rheumatoid Arthritis. Clinical Medicine Insights: Arthritis and Musculoskeletal Disorders 2013, 6, CMAMD–S5558. [Google Scholar] [CrossRef] [PubMed]
  11. 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. Journal of cellular physiology 2019, 234, 10018–10031. [Google Scholar] [CrossRef] [PubMed]
  12. Systematic Review: Comparative Effectiveness and Harms of Disease-Modifying Medications for Rheumatoid Arthritis. Annals of Internal Medicine 2008, 148, 124–134. [CrossRef]
  13. Wang, W.; Zhou, H.; Liu, L. Side effects of methotrexate therapy for rheumatoid arthritis: A systematic review. European Journal of Medicinal Chemistry 2018, 158, 502–516. [Google Scholar] [CrossRef] [PubMed]
  14. Brickman, A.M.; Yeung, L.K.; Alschuler, D.M.; Ottaviani, J.I.; Kuhnle, G.G.C.; Sloan, R.P.; Luttmann-Gibson, H.; Copeland, T.; Schroeter, H.; Sesso, H.D.; et al. Dietary flavanols restore hippocampal-dependent memory in older adults with lower diet quality and lower habitual flavanol consumption. Proceedings of the National Academy of Sciences of the United States of America 2023, 120, e2216932120. [Google Scholar] [CrossRef] [PubMed]
  15. Gamel, T.H.; Abdel-Aal, E.M.; Tucker, A.J.; Pare, S.M.; Faughnan, K.; O'Brien, C.D.; Dykun, A.; Rabalski, I.; Pickard, M.; Wright, A.J. Consumption of whole purple and regular wheat modestly improves metabolic markers in adults with elevated high-sensitivity C-reactive protein: a randomised, single-blind parallel-arm study. The British journal of nutrition 2020, 124, 1179–1189. [Google Scholar] [CrossRef] [PubMed]
  16. Shoji, T.; Masumoto, S.; Moriichi, N.; Ohtake, Y.; Kanda, T. Administration of Apple Polyphenol Supplements for Skin Conditions in Healthy Women: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2020, 12. [Google Scholar] [CrossRef] [PubMed]
  17. Arcusa, R.; Carrillo, J.; Xandri-Martínez, R.; Cerdá, B.; Villaño, D.; Marhuenda, J.; Zafrilla, M.P. Effects of a Fruit and Vegetable-Based Nutraceutical on Biomarkers of Inflammation and Oxidative Status in the Plasma of a Healthy Population: A Placebo-Controlled, Double-Blind, and Randomized Clinical Trial. Molecules (Basel, Switzerland) 2021, 26. [Google Scholar] [CrossRef] [PubMed]
  18. Woolf, E.K.; Terwoord, J.D.; Litwin, N.S.; Vazquez, A.R.; Lee, S.Y.; Ghanem, N.; Michell, K.A.; Smith, B.T.; Grabos, L.E.; Ketelhut, N.B.; et al. Daily blueberry consumption for 12 weeks improves endothelial function in postmenopausal women with above-normal blood pressure through reductions in oxidative stress: a randomized controlled trial. Food & function 2023, 14, 2621–2641. [Google Scholar] [CrossRef] [PubMed]
  19. Ruiz-García, I.; Ortíz-Flores, R.; Badía, R.; García-Borrego, A.; García-Fernández, M.; Lara, E.; Martín-Montañez, E.; García-Serrano, S.; Valdés, S.; Gonzalo, M.; et al. Rich oleocanthal and oleacein extra virgin olive oil and inflammatory and antioxidant status in people with obesity and prediabetes. The APRIL study: A randomised, controlled crossover study. Clinical nutrition (Edinburgh, Scotland) 2023, 42, 1389–1398. [Google Scholar] [CrossRef] [PubMed]
  20. Lackner, S.; Mahnert, A.; Moissl-Eichinger, C.; Madl, T.; Habisch, H.; Meier-Allard, N.; Kumpitsch, C.; Lahousen, T.; Kohlhammer-Dohr, A.; Mörkl, S.; et al. Interindividual differences in aronia juice tolerability linked to gut microbiome and metabolome changes-secondary analysis of a randomized placebo-controlled parallel intervention trial. Microbiome 2024, 12, 49. [Google Scholar] [CrossRef] [PubMed]
  21. Chiu, H.F.; Liao, Y.R.; Shen, Y.C.; Han, Y.C.; Golovinskaia, O.; Venkatakrishnan, K.; Hung, C.C.; Wang, C.K. Improvement on blood pressure and skin using roselle drink: A clinical trial. Journal of food biochemistry 2022, 46, e14287. [Google Scholar] [CrossRef]
  22. Liao, H.-J.; Tzen, J.T.C. The Potential Role of Phenolic Acids from Salvia miltiorrhiza and Cynara scolymus and Their Derivatives as JAK Inhibitors: An In Silico Study. International journal of molecular sciences 2022, 23, 4033. [Google Scholar] [CrossRef]
  23. Moon, S.Y.; Kim, K.D.; Yoo, J.; Lee, J.-H.; Hwangbo, C. Phytochemicals Targeting JAK–STAT Pathways in Inflammatory Bowel Disease: Insights from Animal Models. Molecules (Basel, Switzerland) 2021, 26, 2824. [Google Scholar] [CrossRef] [PubMed]
  24. Nunes, C.; Almeida, L.; Barbosa, R.M.; Laranjinha, J. Luteolin suppresses the JAK/STAT pathway in a cellular model of intestinal inflammation. Food & function 2017, 8, 387–396. [Google Scholar] [CrossRef] [PubMed]
  25. Kour, G.; Choudhary, R.; Anjum, S.; Bhagat, A.; Bajaj, B.K.; Ahmed, Z. Phytochemicals targeting JAK/STAT pathway in the treatment of rheumatoid arthritis: Is there a future? Biochemical Pharmacology 2022, 197, 114929. [Google Scholar] [CrossRef] [PubMed]
  26. Nunes, A.R.; Flores-Félix, J.D.; Gonçalves, A.C.; Falcão, A.; Alves, G.; Silva, L.R. Anti-Inflammatory and Antimicrobial Activities of Portuguese Prunus avium L. (Sweet Cherry) By-Products Extracts. Nutrients 2022, 14, 4576. [Google Scholar] [CrossRef]
  27. Zhao, L.; Wu, Q.; Long, Y.; Qu, Q.; Qi, F.; Liu, L.; Zhang, L.; Ai, K. microRNAs: critical targets for treating rheumatoid arthritis angiogenesis. Journal of Drug Targeting 2024, 32, 1–20. [Google Scholar] [CrossRef]
  28. Gonçalves, A.C.; Costa, A.R.; Flores-Félix, J.D.; Falcão, A.; Alves, G.; Silva, L.R. Anti-Inflammatory and Antiproliferative Properties of Sweet Cherry Phenolic-Rich Extracts. Molecules 2022, 27. [Google Scholar] [CrossRef] [PubMed]
  29. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. Journal of Natural Products 2020, 83, 770–803. [Google Scholar] [CrossRef]
  30. Hamad, H.A.M. Phenolic Compounds: Classification, Chemistry, and Updated Techniques of Analysis and Synthesis. In Phenolic Compounds; Farid, A.B., Ed.; IntechOpen: Rijeka, 2021; p. Ch. 4. [Google Scholar]
  31. Gonçalves, A.C.; Gaspar, D.; Flores-Félix, J.D.; Falcão, A.; Alves, G.; Silva, L.R. Effects of Functional Phenolics Dietary Supplementation on Athletes' Performance and Recovery: A Review. International journal of molecular sciences 2022, 23. [Google Scholar] [CrossRef] [PubMed]
  32. Matsumura, Y.; Kitabatake, M.; Kayano, S.-i.; Ito, T. Dietary Phenolic Compounds: Their Health Benefits and Association with the Gut Microbiota. Antioxidants 2023, 12, 880. [Google Scholar] [CrossRef] [PubMed]
  33. Carregosa, D.; Carecho, R.; Figueira, I.; N Santos, C. Low-Molecular Weight Metabolites from Polyphenols as Effectors for Attenuating Neuroinflammation. Journal of Agricultural and Food Chemistry 2020, 68, 1790–1807. [Google Scholar] [CrossRef] [PubMed]
  34. Capanoglu, E.; Chen, F. The interaction between food components and gut microbiota. Food chemistry 2024, 442, 138117. [Google Scholar] [CrossRef] [PubMed]
  35. Chukwumah, Y.; Walker, L.T.; Verghese, M. Peanut skin color: a biomarker for total polyphenolic content and antioxidative capacities of peanut cultivars. International journal of molecular sciences 2009, 10, 4941–4952. [Google Scholar] [CrossRef] [PubMed]
  36. Favre, G.; Piccardo, D.; Sergio, G.-A.; Pérez-Navarro, J.; García-Romero, E.; Mena-Morales, A.; González-Neves, G. Stilbenes in grapes and wines of Tannat, Marselan and Syrah from Uruguay: This article is published in cooperation with the 11th OenoIVAS International Symposium, June 25–28 2019, Bordeaux, France. OENO One 2020, 54, 27–36. [Google Scholar] [CrossRef]
  37. Sobolev, V.S.; Cole, R.J. trans-Resveratrol Content in Commercial Peanuts and Peanut Products. Journal of Agricultural and Food Chemistry 1999, 47, 1435–1439. [Google Scholar] [CrossRef] [PubMed]
  38. Yilmaz, Y.; Göksel, Z.; Erdoğan, S.S.; Öztürk, A.; Atak, A.; Özer, C. Antioxidant Activity and Phenolic Content of Seed, Skin and Pulp Parts of 22 Grape (Vitis vinifera L.) Cultivars (4 Common and 18 Registered or Candidate for Registration). Journal of Food Processing and Preservation 2015, 39, 1682–1691. [Google Scholar] [CrossRef]
  39. De Silva, S.F.; Alcorn, J. Flaxseed Lignans as Important Dietary Polyphenols for Cancer Prevention and Treatment: Chemistry, Pharmacokinetics, and Molecular Targets. Pharmaceuticals (Basel, Switzerland) 2019, 12. [Google Scholar] [CrossRef] [PubMed]
  40. Gai, F.; Janiak, M.A.; Sulewska, K.; Peiretti, P.G.; Karamać, M. Phenolic Compound Profile and Antioxidant Capacity of Flax (Linum usitatissimum L.) Harvested at Different Growth Stages. Molecules (Basel, Switzerland) 2023, 28. [Google Scholar] [CrossRef] [PubMed]
  41. Oomah, B.D.; Kenaschuk, E.O.; Mazza, G. Phenolic Acids in Flaxseed. Journal of Agricultural and Food Chemistry 1995, 43, 2016–2019. [Google Scholar] [CrossRef]
  42. Yoon, C.H.; Chung, S.J.; Lee, S.W.; Park, Y.B.; Lee, S.K.; Park, M.C. Gallic acid, a natural polyphenolic acid, induces apoptosis and inhibits proinflammatory gene expressions in rheumatoid arthritis fibroblast-like synoviocytes. Joint bone spine 2013, 80, 274–279. [Google Scholar] [CrossRef] [PubMed]
  43. Gonçalves, A.C.; Rodrigues, M.; Santos, A.O.; Alves, G.; Silva, L.R. Antioxidant Status, Antidiabetic Properties and Effects on Caco-2 Cells of Colored and Non-Colored Enriched Extracts of Sweet Cherry Fruits. Nutrients 2018, 10, 1688. [Google Scholar] [CrossRef] [PubMed]
  44. Kalinowska, M.; Gołębiewska, E.; Świderski, G.; Męczyńska-Wielgosz, S.; Lewandowska, H.; Pietryczuk, A.; Cudowski, A.; Astel, A.; Świsłocka, R.; Samsonowicz, M.; et al. Plant-Derived and Dietary Hydroxybenzoic Acids—A Comprehensive Study of Structural, Anti-/Pro-Oxidant, Lipophilic, Antimicrobial, and Cytotoxic Activity in MDA-MB-231 and MCF-7 Cell Lines. Nutrients 2021, 13, 3107. [Google Scholar] [CrossRef] [PubMed]
  45. Khawula, S.; Gokul, A.; Niekerk, L.-A.; Basson, G.; Keyster, M.; Badiwe, M.; Klein, A.; Nkomo, M. Insights into the Effects of Hydroxycinnamic Acid and Its Secondary Metabolites as Antioxidants for Oxidative Stress and Plant Growth under Environmental Stresses. Current Issues in Molecular Biology 2024, 46, 81–95. [Google Scholar] [CrossRef] [PubMed]
  46. Gonçalves, A.C.; Campos, G.; Alves, G.; Garcia-Viguera, C.; Moreno, D.A.; Silva, L.R. Physical and phytochemical composition of 23 Portuguese sweet cherries as conditioned by variety (or genotype). Food Chem 2021, 335, 127637. [Google Scholar] [CrossRef] [PubMed]
  47. Oksuz, T.; Tacer-Caba, Z.; Nilufer-Erdil, D.; Boyacioglu, D. Changes in bioavailability of sour cherry (Prunus cerasus L.) phenolics and anthocyanins when consumed with dairy food matrices. Journal of food science and technology 2019, 56, 4177–4188. [Google Scholar] [CrossRef] [PubMed]
  48. Gonçalves, A.C.; Rodrigues, M.; Flores-Félix, J.D.; Campos, G.; Nunes, A.R.; Ribeiro, A.B.; Silva, L.R.; Alves, G. Sweet cherry phenolics revealed to be promising agents in inhibiting P-glycoprotein activity and increasing cellular viability under oxidative stress conditions: in vitro and in silico study. Journal of food science 2022, 87, 450–465. [Google Scholar] [CrossRef] [PubMed]
  49. Nowicka, P.; Wojdyło, A.; Samoticha, J. Evaluation of phytochemicals, antioxidant capacity, and antidiabetic activity of novel smoothies from selected Prunus fruits. Journal of Functional Foods 2016, 25, 397–407. [Google Scholar] [CrossRef]
  50. Rojas-Ocampo, E.; Torrejón-Valqui, L.; Muñóz-Astecker, L.D.; Medina-Mendoza, M.; Mori-Mestanza, D.; Castro-Alayo, E.M. Antioxidant capacity, total phenolic content and phenolic compounds of pulp and bagasse of four Peruvian berries. Heliyon 2021, 7, e07787. [Google Scholar] [CrossRef] [PubMed]
  51. Flores-Félix, J.D.; Gonçalves, A.C.; Meirinho, S.; Nunes, A.R.; Alves, G.; Garcia-Viguera, C.; Moreno, D.A.; Silva, L.R. Differential response of blueberry to the application of bacterial inoculants to improve yield, organoleptic qualities and concentration of bioactive compounds. Microbiological Research 2024, 278, 127544. [Google Scholar] [CrossRef] [PubMed]
  52. Sellappan, S.; Akoh, C.C.; Krewer, G. Phenolic compounds and antioxidant capacity of Georgia-grown blueberries and blackberries. J Agric Food Chem 2002, 50, 2432–2438. [Google Scholar] [CrossRef] [PubMed]
  53. Młynarczyk, K.; Walkowiak-Tomczak, D.; Łysiak, G.P. Bioactive properties of Sambucus nigra L. as a functional ingredient for food and pharmaceutical industry. Journal of Functional Foods 2018, 40, 377–390. [Google Scholar] [CrossRef] [PubMed]
  54. Li, J.; Shi, C.; Shen, D.; Han, T.; Wu, W.; Lyu, L.; Li, W. Composition and Antioxidant Activity of Anthocyanins and Non-Anthocyanin Flavonoids in Blackberry from Different Growth Stages. Foods 2022, 11. [Google Scholar] [CrossRef] [PubMed]
  55. Albert, C.; Codină, G.G.; Héjja, M.; András, C.D.; Chetrariu, A.; Dabija, A. Study of Antioxidant Activity of Garden Blackberries (Rubus fruticosus L.) Extracts Obtained with Different Extraction Solvents. Applied Sciences 2022, 12, 4004. [Google Scholar] [CrossRef]
  56. Yu, C.; Ranieri, M.; Lv, D.; Zhang, M.; Charles, M.T.; Tsao, R.; Rekika, D.; Khanizadeh, S. Phenolic Composition and Antioxidant Capacity of Newly Developed Strawberry Lines from British Columbia and Quebec. International Journal of Food Properties 2011, 14, 59–67. [Google Scholar] [CrossRef]
  57. Sondheimer, E.; Karash, C.B. The Major Anthocyanin Pigments of the Wild Strawberry (Fragaria vesca). Nature 1956, 178, 648–649. [Google Scholar] [CrossRef]
  58. Taghavi, T.; Patel, H.; Akande, O.E.; Galam, D.C.A. Total Anthocyanin Content of Strawberry and the Profile Changes by Extraction Methods and Sample Processing. Foods 2022, 11. [Google Scholar] [CrossRef] [PubMed]
  59. Orak, H.H. Total antioxidant activities, phenolics, anthocyanins, polyphenoloxidase activities of selected red grape cultivars and their correlations. Scientia Horticulturae 2007, 111, 235–241. [Google Scholar] [CrossRef]
  60. Kőrösi, L.; Molnár, S.; Teszlák, P.; Dörnyei, Á.; Maul, E.; Töpfer, R.; Marosvölgyi, T.; Szabó, É.; Röckel, F. Comparative Study on Grape Berry Anthocyanins of Various Teinturier Varieties. Foods 2022, 11, 3668. [Google Scholar] [CrossRef] [PubMed]
  61. Negro, C.; Aprile, A.; De Bellis, L.; Miceli, A. Nutraceutical Properties of Mulberries Grown in Southern Italy (Apulia). Antioxidants (Basel, Switzerland) 2019, 8. [Google Scholar] [CrossRef] [PubMed]
  62. Denev, P.; Kratchanova, M.; Petrova, I.; Klisurova, D.; Georgiev, Y.; Ognyanov, M.; Yanakieva, I. Black Chokeberry (<i>Aronia melanocarpa</i> (Michx.) Elliot) Fruits and Functional Drinks Differ Significantly in Their Chemical Composition and Antioxidant Activity. Journal of Chemistry 2018, 2018, 9574587. [Google Scholar] [CrossRef]
  63. Kasprzak-Drozd, K.; Oniszczuk, T.; Soja, J.; Gancarz, M.; Wojtunik-Kulesza, K.; Markut-Miotła, E.; Oniszczuk, A. The Efficacy of Black Chokeberry Fruits against Cardiovascular Diseases. International journal of molecular sciences 2021, 22, 6541. [Google Scholar] [CrossRef] [PubMed]
  64. Šimerdová, B.; Bobríková, M.; Lhotská, I.; Kaplan, J.; Křenová, A.; Šatínský, D. Evaluation of Anthocyanin Profiles in Various Blackcurrant Cultivars over a Three-Year Period Using a Fast HPLC-DAD Method. Foods 2021, 10. [Google Scholar] [CrossRef] [PubMed]
  65. Milić, A.; Daničić, T.; Tepić Horecki, A.; Šumić, Z.; Teslić, N.; Bursać Kovačević, D.; Putnik, P.; Pavlić, B. Sustainable Extractions for Maximizing Content of Antioxidant Phytochemicals from Black and Red Currants. Foods 2022, 11, 325. [Google Scholar] [CrossRef] [PubMed]
  66. Wiczkowski, W.; Szawara-Nowak, D.; Topolska, J. Red cabbage anthocyanins: Profile, isolation, identification, and antioxidant activity. Food Research International 2013, 51, 303–309. [Google Scholar] [CrossRef]
  67. Fabek Uher, S.; Radman, S.; Opačić, N.; Dujmović, M.; Benko, B.; Lagundžija, D.; Mijić, V.; Prša, L.; Babac, S.; Šic Žlabur, J. Alfalfa, Cabbage, Beet and Fennel Microgreens in Floating Hydroponics—Perspective Nutritious Food? Plants 2023, 12, 2098. [Google Scholar] [CrossRef] [PubMed]
  68. Mujić, I.; Šertović, E.; Jokić, S.; Sarić, Z.; Alibabić, V.; Vidovic, S.S.; Zivkovic, J.V. Isoflavone content and antioxidant properties of soybean seeds. Croatian journal of food science and technology 2011, 3, 16–20. [Google Scholar]
  69. Dhanda, T.; Madan, V.K.; Beniwal, R. Quantitative analysis of phenols, flavonoids in different parts of Aegle marmelos (Bael) along with the evaluation of Antioxidant potential using different extracts. Journal of Pharmacognosy and Phytochemistry 2020, 9, 1192–1198. [Google Scholar]
  70. Shinde, P.B.; Katekhaye, S.D.; Mulik, M.B.; Laddha, K.S. Rapid simultaneous determination of marmelosin, umbelliferone and scopoletin from Aegle marmelos fruit by RP-HPLC. Journal of food science and technology 2014, 51, 2251–2255. [Google Scholar] [CrossRef] [PubMed]
  71. Bento, C.; Gonçalves, A.C.; Silva, B.; Silva, L.R. Assessing the phenolic profile, antioxidant, antidiabetic and protective effects against oxidative damage in human erythrocytes of peaches from Fundão. Journal of Functional Foods 2018, 43, 224–233. [Google Scholar] [CrossRef]
  72. Arts, I.C.W.; van de Putte, B.; Hollman, P.C.H. Catechin Contents of Foods Commonly Consumed in The Netherlands. 2. Tea, Wine, Fruit Juices, and Chocolate Milk. Journal of Agricultural and Food Chemistry 2000, 48, 1752–1757. [Google Scholar] [CrossRef] [PubMed]
  73. Almanza-Aguilera, E.; Ceballos-Sánchez, D.; Achaintre, D.; Rothwell, J.A.; Laouali, N.; Severi, G.; Katzke, V.; Johnson, T.; Schulze, M.B.; Palli, D.; et al. Urinary Concentrations of (+)-Catechin and (-)-Epicatechin as Biomarkers of Dietary Intake of Flavan-3-ols in the European Prospective Investigation into Cancer and Nutrition (EPIC) Study. Nutrients 2021, 13, 4157. [Google Scholar] [CrossRef] [PubMed]
  74. Wangkarn, S.; Grudpan, K.; Khanongnuch, C.; Pattananandecha, T.; Apichai, S.; Saenjum, C. Development of HPLC Method for Catechins and Related Compounds Determination and Standardization in Miang (Traditional Lanna Fermented Tea Leaf in Northern Thailand). Molecules (Basel, Switzerland) 2021, 26. [Google Scholar] [CrossRef] [PubMed]
  75. Miean, K.H.; Mohamed, S. Flavonoid (myricetin, quercetin, kaempferol, luteolin, and apigenin) content of edible tropical plants. J Agric Food Chem 2001, 49, 3106–3112. [Google Scholar] [CrossRef] [PubMed]
  76. Jin, S.; Zhang, L.; Wang, L. Kaempferol, a potential neuroprotective agent in neurodegenerative diseases: From chemistry to medicine. Biomedicine & Pharmacotherapy 2023, 165, 115215. [Google Scholar] [CrossRef] [PubMed]
  77. Abbou, F.; Azzi, R.; Ouffai, K.; El Haci, I.A.; Belyagoubi-Benhammou, N.; Bensouici, C.; Benamar, H. Phenolic profile, antioxidant and enzyme inhibitory properties of phenolic-rich fractions from the aerial parts of Mentha pulegium L. South African Journal of Botany 2022, 146, 196–204. [Google Scholar] [CrossRef]
  78. Häkkinen, S.H.; Kärenlampi, S.O.; Heinonen, I.M.; Mykkänen, H.M.; Törrönen, A.R. Content of the flavonols quercetin, myricetin, and kaempferol in 25 edible berries. J Agric Food Chem 1999, 47, 2274–2279. [Google Scholar] [CrossRef] [PubMed]
  79. Demonty, I.; Lin, Y.; Zebregs, Y.E.M.P.; Vermeer, M.A.; van der Knaap, H.C.M.; Jäkel, M.; Trautwein, E.A. The Citrus Flavonoids Hesperidin and Naringin Do Not Affect Serum Cholesterol in Moderately Hypercholesterolemic Men and Women. The Journal of Nutrition 2010, 140, 1615–1620. [Google Scholar] [CrossRef] [PubMed]
  80. Wang, M.; Zhao, J.; Avula, B.; Lee, J.; Upton, R.; Khan, I.A. Chemical characterization and quantitative determination of flavonoids and phenolic acids in yerba santa (Eriodictyon spp.) using UHPLC/DAD/Q-ToF. Journal of Pharmaceutical and Biomedical Analysis 2023, 234, 115570. [Google Scholar] [CrossRef] [PubMed]
  81. García-Nicolás, M.; Ledesma-Escobar, C.A.; Priego-Capote, F. Spatial Distribution and Antioxidant Activity of Extracts from Citrus Fruits. Antioxidants (Basel, Switzerland) 2023, 12. [Google Scholar] [CrossRef] [PubMed]
  82. Elezovic, A.; Uzunovic, A.; Hadzidedic, S.; Pilipovic, S.; Sapcanin, A. New and fast HPLC method for analysis of flavonoids in honey and propolis samples. Planta Med 2010, 76, P526. [Google Scholar] [CrossRef]
  83. Yang, F.; Chu, T.; Zhang, Y.; Liu, X.; Sun, G.; Chen, Z. Quality assessment of licorice (Glycyrrhiza glabra L.) from different sources by multiple fingerprint profiles combined with quantitative analysis, antioxidant activity and chemometric methods. Food chemistry 2020, 324, 126854. [Google Scholar] [CrossRef] [PubMed]
  84. Guofang, X.; Xiaoyan, X.; Xiaoli, Z.; Yongling, L.; Zhibing, Z. Changes in phenolic profiles and antioxidant activity in rabbiteye blueberries during ripening. International Journal of Food Properties 2019, 22, 320–329. [Google Scholar] [CrossRef]
  85. Popova, I.E.; Hall, C.; Kubátová, A. Determination of lignans in flaxseed using liquid chromatography with time-of-flight mass spectrometry. Journal of Chromatography A 2009, 1216, 217–229. [Google Scholar] [CrossRef] [PubMed]
  86. Herrmann, K.; Nagel, C.W. Occurrence and content of hydroxycinnamic and hydroxybenzoic acid compounds in foods. Critical Reviews in Food Science and Nutrition 1989, 28, 315–347. [Google Scholar] [CrossRef] [PubMed]
  87. Ramli; , N.; Yatim; , A.M.; Said; , M.; Hok, H.C. HPLC determination of methylxanthines and polyphenols levels In cocoa and chocolate products Malaysian Journal of Analytical Sciences 2001, 7, 377–386.
  88. Sova, M.; Saso, L. Natural Sources, Pharmacokinetics, Biological Activities and Health Benefits of Hydroxycinnamic Acids and Their Metabolites. Nutrients 2020, 12. [Google Scholar] [CrossRef] [PubMed]
  89. Inglett;, G.E.; Chen;, D.; Liu, S.X. Antioxidant Activities of Selective Gluten Free Ancient Grains. Food and Nutrition Sciences 2015, 6. [CrossRef]
  90. Reinisalo, M.; Kårlund, A.; Koskela, A.; Kaarniranta, K.; Karjalainen, R.O. Polyphenol Stilbenes: Molecular Mechanisms of Defence against Oxidative Stress and Aging-Related Diseases. Oxidative medicine and cellular longevity 2015, 2015, 340520. [Google Scholar] [CrossRef]
  91. Wang, X.; Luo, S.; Li, Q.; Song, L.; Zhang, W.; Yu, P.; Xuan, S.; Wang, Y.; Zhao, J.; Chen, X.; et al. Delphinidins and Naringenin Chalcone Underlying the Fruit Color Changes during Maturity Stages in Eggplant. Agronomy 2022, 12, 1036. [Google Scholar] [CrossRef]
  92. Usenik, V.; Štampar, F.; Veberič, R. Anthocyanins and fruit colour in plums (Prunus domestica L.) during ripening. Food chemistry 2009, 114, 529–534. [Google Scholar] [CrossRef]
  93. Hariram Nile, S.; Hwan Kim, D.; Keum, Y.-S. Determination of Anthocyanin Content and Antioxidant Capacity of Different Grape Varieties. Ciência Téc. Vitiv. 2015, 30, 60–68. [Google Scholar] [CrossRef]
  94. Rajan, V.K.; Hasna, C.K.; Muraleedharan, K. The natural food colorant Peonidin from cranberries as a potential radical scavenger – A DFT based mechanistic analysis. Food chemistry 2018, 262, 184–190. [Google Scholar] [CrossRef] [PubMed]
  95. Sokół-Łętowska, A.; Kucharska, A.Z.; Hodun, G.; Gołba, M. Chemical Composition of 21 Cultivars of Sour Cherry (Prunus cerasus) Fruit Cultivated in Poland. Molecules (Basel, Switzerland) 2020, 25, 4587. [Google Scholar] [CrossRef] [PubMed]
  96. Santoni;, A.; Amanda;, H.; Darwis, D. Characterization of pelargonidin compound from raspberry fruit (Rubus rosifolius Sm) with mass spectroscopy method. Journal of Chemical and Pharmaceutical Research 2015, 7, 804–808.
  97. Alvarado, J.L.; Leschot, A.; Olivera-Nappa, Á.; Salgado, A.M.; Rioseco, H.; Lyon, C.; Vigil, P. Delphinidin-Rich Maqui Berry Extract (Delphinol®) Lowers Fasting and Postprandial Glycemia and Insulinemia in Prediabetic Individuals during Oral Glucose Tolerance Tests. BioMed research international 2016, 2016, 9070537. [Google Scholar] [CrossRef] [PubMed]
  98. Sun, B.; Li, F.; Zhang, X.; Wang, W.; Shao, J.; Zheng, Y. Delphinidin-3-O-glucoside, an active compound of Hibiscus sabdariffa calyces, inhibits oxidative stress and inflammation in rabbits with atherosclerosis. Pharmaceutical biology 2022, 60, 247–254. [Google Scholar] [CrossRef] [PubMed]
  99. Vera de Rosso, V.; Hillebrand, S.; Cuevas Montilla, E.; Bobbio, F.O.; Winterhalter, P.; Mercadante, A.Z. Determination of anthocyanins from acerola (Malpighia emarginata DC.) and açai (Euterpe oleracea Mart.) by HPLC–PDA–MS/MS. Journal of Food Composition and Analysis 2008, 21, 291–299. [Google Scholar] [CrossRef]
  100. Decendit, A.; Mamani-Matsuda, M.; Aumont, V.; Waffo-Teguo, P.; Moynet, D.; Boniface, K.; Richard, E.; Krisa, S.; Rambert, J.; Mérillon, J.M.; et al. Malvidin-3-O-β glucoside, major grape anthocyanin, inhibits human macrophage-derived inflammatory mediators and decreases clinical scores in arthritic rats. Biochem Pharmacol 2013, 86, 1461–1467. [Google Scholar] [CrossRef] [PubMed]
  101. Silva, L.R.; Queiroz, M. Bioactive compounds of red grapes from Dão region (Portugal): Evaluation of phenolic and organic profile. Asian Pacific Journal of Tropical Biomedicine 2016, 6, 315–321. [Google Scholar] [CrossRef]
  102. Kang, H.-J.; Ko, M.-J.; Chung, M.-S. Anthocyanin Structure and pH Dependent Extraction Characteristics from Blueberries (Vaccinium corymbosum) and Chokeberries (Aronia melanocarpa) in Subcritical Water State. Foods 2021, 10, 527. [Google Scholar] [CrossRef] [PubMed]
  103. Lončar, M.; Jakovljević, M.; Šubarić, D.; Pavlić, M.; Buzjak Služek, V.; Cindrić, I.; Molnar, M. Coumarins in Food and Methods of Their Determination. Foods 2020, 9. [Google Scholar] [CrossRef] [PubMed]
  104. Stanic, G.; Jurisic, B.; Brkic, D. HPLC Analysis of Esculin and Fraxin in Horse-Chestnut Bark (Aesculus hippocastanum L.). Croatica Chemica Acta 1999, 72, 827–834. [Google Scholar]
  105. Wang, H.; Provan, G.J.; Helliwell, K. HPLC determination of catechins in tea leaves and tea extracts using relative response factors. Food chemistry 2003, 81, 307–312. [Google Scholar] [CrossRef]
  106. Ramesh, E.; Jayakumar, T.; Elanchezhian, R.; Sakthivel, M.; Geraldine, P.; Thomas, P.A. Green tea catechins, alleviate hepatic lipidemic-oxidative injury in Wistar rats fed an atherogenic diet. Chemico-Biological Interactions 2009, 180, 10–19. [Google Scholar] [CrossRef] [PubMed]
  107. Becerra, L.D.; Quintanilla-Carvajal, M.X.; Escobar, S.; Ruiz Pardo, R.Y. From controlled transformed cocoa beans to chocolate: Bioactive properties, metabolomic profile, and in vitro bioaccessibility. Food chemistry 2024, 433, 137321. [Google Scholar] [CrossRef] [PubMed]
  108. Alam, W.; Khan, H.; Shah, M.A.; Cauli, O.; Saso, L. Kaempferol as a Dietary Anti-Inflammatory Agent: Current Therapeutic Standing. Molecules (Basel, Switzerland) 2020, 25. [Google Scholar] [CrossRef] [PubMed]
  109. FIESCHI, M.; CODIGNOLA, A.; MOSCA, A.M.L. Mutagenic Flavonol Aglycones in Infusions and in Fresh and Pickled Vegetables. Journal of food science 1989, 54, 1492–1495. [Google Scholar] [CrossRef]
  110. Khan, N.; Syed, D.N.; Ahmad, N.; Mukhtar, H. Fisetin: a dietary antioxidant for health promotion. Antioxidants & redox signaling 2013, 19, 151–162. [Google Scholar] [CrossRef] [PubMed]
  111. Imran, M.; Saeed, F.; Hussain, G.; Imran, A.; Mehmood, Z.; Gondal, T.A.; El-Ghorab, A.; Ahmad, I.; Pezzani, R.; Arshad, M.U.; et al. Myricetin: A comprehensive review on its biological potentials. Food science & nutrition 2021, 9, 5854–5868. [Google Scholar] [CrossRef] [PubMed]
  112. Kanaze, F.I.; Gabrieli, C.; Kokkalou, E.; Georgarakis, M.; Niopas, I. Simultaneous reversed-phase high-performance liquid chromatographic method for the determination of diosmin, hesperidin and naringin in different citrus fruit juices and pharmaceutical formulations. Journal of Pharmaceutical and Biomedical Analysis 2003, 33, 243–249. [Google Scholar] [CrossRef] [PubMed]
  113. Waheed Janabi, A.H.; Kamboh, A.A.; Saeed, M.; Xiaoyu, L.; BiBi, J.; Majeed, F.; Naveed, M.; Mughal, M.J.; Korejo, N.A.; Kamboh, R.; et al. Flavonoid-rich foods (FRF): A promising nutraceutical approach against lifespan-shortening diseases. Iranian journal of basic medical sciences 2020, 23, 140–153. [Google Scholar] [CrossRef] [PubMed]
  114. Cannataro, R.; Fazio, A.; La Torre, C.; Caroleo, M.C.; Cione, E. Polyphenols in the Mediterranean Diet: From Dietary Sources to microRNA Modulation. Antioxidants (Basel, Switzerland) 2021, 10. [Google Scholar] [CrossRef]
  115. A Food Supplement with Antioxidative Santa Herba Extract Modulates Energy Metabolism and Contributes to Weight Management. Journal of Medicinal Food 2021, 24, 1235–1242. [CrossRef] [PubMed]
  116. Chun, O.K.; Chung, S.J.; Song, W.O. Estimated Dietary Flavonoid Intake and Major Food Sources of U.S. Adults1,2. The Journal of Nutrition 2007, 137, 1244–1252. [Google Scholar] [CrossRef]
  117. Bogdanov, S. Determination of Pinocembrin in Honey Using HPLC. Journal of Apicultural Research 1989, 28, 55–57. [Google Scholar] [CrossRef]
  118. Dong, Y.; Zhao, M.; Zhao, T.; Feng, M.; Chen, H.; Zhuang, M.; Lin, L. Bioactive profiles, antioxidant activities, nitrite scavenging capacities and protective effects on H2O2-injured PC12 cells of Glycyrrhiza glabra L. leaf and root extracts. Molecules (Basel, Switzerland) 2014, 19, 9101–9113. [Google Scholar] [CrossRef] [PubMed]
  119. Adham, A.N. Qualitative and quantitative estimation of hesperidin in peel and juice of citrus fruits by RP-HPLC method growing in Kurdistan region/Iraq. International Journal of Pharmaceutical Sciences Review and Research 2015, 33, 220–224. [Google Scholar]
  120. Volpi, N.; Bergonzini, G. Analysis of flavonoids from propolis by on-line HPLC–electrospray mass spectrometry. Journal of Pharmaceutical and Biomedical Analysis 2006, 42, 354–361. [Google Scholar] [CrossRef] [PubMed]
  121. Fukutake, M.; Takahashi, M.; Ishida, K.; Kawamura, H.; Sugimura, T.; Wakabayashi, K. Quantification of genistein and genistin in soybeans and soybean products. Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association 1996, 34, 457–461. [Google Scholar] [CrossRef] [PubMed]
  122. Mitani, K.; Narimatsu, S.; Kataoka, H. Determination of daidzein and genistein in soybean foods by automated on-line in-tube solid-phase microextraction coupled to high-performance liquid chromatography. Journal of Chromatography A 2003, 986, 169–177. [Google Scholar] [CrossRef] [PubMed]
  123. Oshima, A.; Mine, W.; Nakada, M.; Yanase, E. Analysis of isoflavones and coumestrol in soybean sprouts. Bioscience, Biotechnology, and Biochemistry 2016, 80, 2077–2079. [Google Scholar] [CrossRef] [PubMed]
  124. Franke, A.A.; Custer, L.J.; Wang, W.; Shi, C.Y. HPLC analysis of isoflavonoids and other phenolic agents from foods and from human fluids. Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine (New York, N.Y.) 1998, 217, 263–273. [Google Scholar] [CrossRef]
  125. Isoflavone Composition, Total Polyphenolic Content, and Antioxidant Activity in Soybeans of Different Origin. Journal of Medicinal Food 2010, 13, 657–664. [CrossRef]
  126. De Leo, M.; Iannuzzi, A.M.; Germanò, M.P.; D'Angelo, V.; Camangi, F.; Sevi, F.; Diretto, G.; De Tommasi, N.; Braca, A. Comparative chemical analysis of six ancient italian sweet cherry (Prunus avium L.) varieties showing antiangiogenic activity. Food chemistry 2021, 360, 129999. [Google Scholar] [CrossRef] [PubMed]
  127. Kumar, N.; Goel, N. Phenolic acids: Natural versatile molecules with promising therapeutic applications. Biotechnology Reports 2019, 24, e00370. [Google Scholar] [CrossRef] [PubMed]
  128. Debelo, H.; Li, M.; Ferruzzi, M.G. Processing influences on food polyphenol profiles and biological activity. Current Opinion in Food Science 2020, 32, 90–102. [Google Scholar] [CrossRef]
  129. Guneidy, R.A.; Zaki, E.R.; Gad, A.A.M.; Saleh, N.S.E.; Shokeer, A. Evaluation of Phenolic Content Diversity along with Antioxidant/Pro-Oxidant, Glutathione Transferase Inhibition, and Cytotoxic Potential of Selected Commonly Used Plants. Preventive nutrition and food science 2022, 27, 282–298. [Google Scholar] [CrossRef]
  130. Nurzyńska-Wierdak, R. Phenolic Compounds from New Natural Sources-Plant Genotype and Ontogenetic Variation. Molecules (Basel, Switzerland) 2023, 28. [Google Scholar] [CrossRef]
  131. Gebregziabher, B.S.; Zhang, S.; Ghosh, S.; Shaibu, A.S.; Azam, M.; Abdelghany, A.M.; Qi, J.; Agyenim-Boateng, K.G.; Htway, H.T.P.; Feng, Y.; et al. Origin, Maturity Group and Seed Coat Color Influence Carotenoid and Chlorophyll Concentrations in Soybean Seeds. Plants 2022, 11, 848. [Google Scholar] [CrossRef] [PubMed]
  132. Weyand, C.M.; Goronzy, J.J. The immunology of rheumatoid arthritis. Nature Immunology 2021, 22, 10–18. [Google Scholar] [CrossRef] [PubMed]
  133. Christoph Jan, W.; Athanassios, F.; Agata, G.; Lars-Ove, B.; Yuet Wai, K.; Kaimin, C.; Joachim, H.; Sandra, F.-W.; Deike, V.; Sebastian, L.; et al. Role of oxidative stress in rheumatoid arthritis: insights from the Nrf2-knockout mice. Annals of the Rheumatic Diseases 2011, 70, 844. [Google Scholar] [CrossRef] [PubMed]
  134. Gonçalves, A.C.; Bento, C.; Silva, B.M.; Silva, L.R. Sweet cherries from Fundão possess antidiabetic potential and protect human erythrocytes against oxidative damage. Food research international (Ottawa, Ont.) 2017, 95, 91–100. [Google Scholar] [CrossRef] [PubMed]
  135. Brogi, S.; Guarino, I.; Flori, L.; Sirous, H.; Calderone, V. In Silico Identification of Natural Products and World-Approved Drugs Targeting the KEAP1/NRF2 Pathway Endowed with Potential Antioxidant Profile. Computation 2023, 11, 255. [Google Scholar] [CrossRef]
  136. Li, M.; Huang, W.; Jie, F.; Wang, M.; Zhong, Y.; Chen, Q.; Lu, B. Discovery of Keap1-Nrf2 small-molecule inhibitors from phytochemicals based on molecular docking. Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association 2019, 133, 110758. [Google Scholar] [CrossRef] [PubMed]
  137. Mili, A.; Birangal, S.; Nandakumar, K.; Lobo, R. A computational study to identify Sesamol derivatives as NRF2 activator for protection against drug-induced liver injury (DILI). Molecular Diversity 2023. [Google Scholar] [CrossRef] [PubMed]
  138. Lv, L.; Shu, H.; Mo, X.; Tian, Y.; Guo, H.; Sun, H.-Y. Activation of the Nrf2 Antioxidant Pathway by Longjing Green Tea Polyphenols in Mice Livers. Natural Product Communications 2022, 17, 1934578X221139409. [Google Scholar] [CrossRef]
  139. Platzer, M.; Kiese, S.; Herfellner, T.; Schweiggert-Weisz, U.; Eisner, P. How Does the Phenol Structure Influence the Results of the Folin-Ciocalteu Assay? Antioxidants (Basel, Switzerland) 2021, 10. [Google Scholar] [CrossRef] [PubMed]
  140. Rice-Evans, C.A.; Miller, N.J.; Paganga, G. Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radical Biology and Medicine 1996, 20, 933–956. [Google Scholar] [CrossRef] [PubMed]
  141. Soobrattee, M.A.; Neergheen, V.S.; Luximon-Ramma, A.; Aruoma, O.I.; Bahorun, T. Phenolics as potential antioxidant therapeutic agents: mechanism and actions. Mutation research 2005, 579, 200–213. [Google Scholar] [CrossRef]
  142. Platzer, M.; Kiese, S.; Herfellner, T.; Schweiggert-Weisz, U.; Miesbauer, O.; Eisner, P. Common Trends and Differences in Antioxidant Activity Analysis of Phenolic Substances Using Single Electron Transfer Based Assays. Molecules (Basel, Switzerland) 2021, 26, 1244. [Google Scholar] [CrossRef] [PubMed]
  143. Zhu, F.; Yuan, Z.; Xueqing Zhao; Yin, Y.; Feng, L. COMPOSITION AND CONTENTS OF ANTHOCYANINS IN DIFFERENT POMEGRANATE CULTIVARS. Acta Horticulturae 2015, 1089, 35–41. [CrossRef]
  144. Passafiume, R.; Perrone, A.; Sortino, G.; Gianguzzi, G.; Saletta, F.; Gentile, C.; Farina, V. Chemical–physical characteristics, polyphenolic content and total antioxidant activity of three Italian-grown pomegranate cultivars. NFS Journal 2019, 16, 9–14. [Google Scholar] [CrossRef]
  145. Heim, K.E.; Tagliaferro, A.R.; Bobilya, D.J. Flavonoid antioxidants: chemistry, metabolism and structure-activity relationships. The Journal of Nutritional Biochemistry 2002, 13, 572–584. [Google Scholar] [CrossRef] [PubMed]
  146. Eklund, P.C.; Långvik, O.K.; Wärnå, J.P.; Salmi, T.O.; Willför, S.M.; Sjöholm, R.E. Chemical studies on antioxidant mechanisms and free radical scavenging properties of lignans. Organic & Biomolecular Chemistry 2005, 3, 3336–3347. [Google Scholar] [CrossRef] [PubMed]
  147. Rühlmann, A.; Antovic, D.; Müller, T.J.J.; Urlacher, V.B. Regioselective Hydroxylation of Stilbenes by Engineered Cytochrome P450 from Thermobifida fusca YX. Advanced Synthesis & Catalysis 2017, 359, 984–994. [Google Scholar] [CrossRef]
  148. Gonçalves, A.C.; Flores-Félix, J.D.; Costa, A.R.; Falcão, A.; Alves, G.; Silva, L.R. Hepatoprotective Effects of Sweet Cherry Extracts (cv. Saco). Foods 2021, 10, 2623. [Google Scholar] [CrossRef] [PubMed]
  149. Simić, A.; Manojlović, D.; Segan, D.; Todorović, M. Electrochemical behavior and antioxidant and prooxidant activity of natural phenolics. Molecules (Basel, Switzerland) 2007, 12, 2327–2340. [Google Scholar] [CrossRef] [PubMed]
  150. Alov, P.; Tsakovska, I.; Pajeva, I. Computational studies of free radical-scavenging properties of phenolic compounds. Current topics in medicinal chemistry 2015, 15, 85–104. [Google Scholar] [CrossRef] [PubMed]
  151. Bayliak, M.M.; Burdyliuk, N.I.; Lushchak, V.I. Effects of pH on antioxidant and prooxidant properties of common medicinal herbs. Open Life Sciences 2016, 11, 298–307. [Google Scholar] [CrossRef]
  152. Cao, G.; Sofic, E.; Prior, R.L. Antioxidant and Prooxidant Behavior of Flavonoids: Structure-Activity Relationships. Free Radical Biology and Medicine 1997, 22, 749–760. [Google Scholar] [CrossRef] [PubMed]
  153. Moran, J.F.; Klucas, R.V.; Grayer, R.J.; Abian, J.; Becana, M. Complexes of Iron with Phenolic Compounds from Soybean Nodules and Other Legume Tissues: Prooxidant and Antioxidant Properties. Free Radical Biology and Medicine 1997, 22, 861–870. [Google Scholar] [CrossRef] [PubMed]
  154. Diniyah, N.; Alam, M.B.; Javed, A.; Alshammari, F.H.; Choi, H.-J.; Lee, S.-H. In silico and docking studies on the binding activities of Keap1 of antioxidant compounds in non-oilseed legumes. Arabian Journal of Chemistry 2023, 16, 104414. [Google Scholar] [CrossRef]
  155. Guan, T.; Bian, C.; Ma, Z. In vitro and in silico perspectives on the activation of antioxidant responsive element by citrus-derived flavonoids. Frontiers in Nutrition 2023, 10. [Google Scholar] [CrossRef] [PubMed]
  156. Shanmugam, T.; Selvaraj, M.; Poomalai, S. Epigallocatechin gallate potentially abrogates fluoride induced lung oxidative stress, inflammation via Nrf2/Keap1 signaling pathway in rats: An in-vivo and in-silico study. International Immunopharmacology 2016, 39, 128–139. [Google Scholar] [CrossRef]
  157. Han, S.G.; Han, S.S.; Toborek, M.; Hennig, B. EGCG protects endothelial cells against PCB 126-induced inflammation through inhibition of AhR and induction of Nrf2-regulated genes. Toxicology and applied pharmacology 2012, 261, 181–188. [Google Scholar] [CrossRef] [PubMed]
  158. Chen, X.; Yang, J.H.; Cho, S.S.; Kim, J.H.; Xu, J.; Seo, K.; Ki, S.H. 5-Caffeoylquinic acid ameliorates oxidative stress-mediated cell death via Nrf2 activation in hepatocytes. Pharmaceutical biology 2020, 58, 999–1005. [Google Scholar] [CrossRef] [PubMed]
  159. Mishra, P.; Paital, B.; Jena, S.; Swain, S.S.; Kumar, S.; Yadav, M.K.; Chainy, G.B.N.; Samanta, L. Possible activation of NRF2 by Vitamin E/Curcumin against altered thyroid hormone induced oxidative stress via NFĸB/AKT/mTOR/KEAP1 signalling in rat heart. Scientific reports 2019, 9, 7408. [Google Scholar] [CrossRef]
  160. Gonçalves, A.C.; Lahlou, R.A.; Alves, G.; Garcia-Viguera, C.; Moreno, D.A.; Silva, L.R. Potential Activity of Abrantes Pollen Extract: Biochemical and Cellular Model Studies. Foods 2021, 10, 2804. [Google Scholar] [CrossRef]
  161. Jesus, F.; Gonçalves, A.C.; Alves, G.; Silva, L.R. Exploring the phenolic profile, antioxidant, antidiabetic and anti-hemolytic potential of Prunus avium vegetal parts. Food Res Int 2019, 116, 600–610. [Google Scholar] [CrossRef] [PubMed]
  162. Oyarzún, J.E.; Andia, M.E.; Uribe, S.; Núñez Pizarro, P.; Núñez, G.; Montenegro, G.; Bridi, R. Honeybee Pollen Extracts Reduce Oxidative Stress and Steatosis in Hepatic Cells. Molecules (Basel, Switzerland) 2020, 26. [Google Scholar] [CrossRef] [PubMed]
  163. Gonçalves, A.C.; Nunes, A.R.; Meirinho, S.; Ayuso-Calles, M.; Roca-Couso, R.; Rivas, R.; Falcão, A.; Alves, G.; Silva, L.R.; Flores-Félix, J.D. Exploring the Antioxidant, Antidiabetic, and Antimicrobial Capacity of Phenolics from Blueberries and Sweet Cherries. Applied Sciences 2023, 13, 6348. [Google Scholar] [CrossRef]
  164. Ohnuma, T.; Sakamoto, K.; Shinoda, A.; Takagi, C.; Ohno, S.; Nishiyama, T.; Ogura, K.; Hiratsuka, A. Procyanidins from Cinnamomi Cortex promote proteasome-independent degradation of nuclear Nrf2 through phosphorylation of insulin-like growth factor-1 receptor in A549 cells. Archives of Biochemistry and Biophysics 2017, 635, 66–73. [Google Scholar] [CrossRef]
  165. Woo, Y.; Oh, J.; Kim, J.-S. Suppression of Nrf2 Activity by Chestnut Leaf Extract Increases Chemosensitivity of Breast Cancer Stem Cells to Paclitaxel. Nutrients 2017, 9, 760. [Google Scholar] [CrossRef] [PubMed]
  166. Zhu, X.; Zhang, Y.; Wang, Y.; Zhang, H.; Wang, X.; Tang, H.; Huang, H.; Zhou, Z.; Chen, B.; Sun, L. Agrimoniin sensitizes pancreatic cancer to apoptosis through ROS-mediated energy metabolism dysfunction. Phytomedicine : international journal of phytotherapy and phytopharmacology 2022, 96, 153807. [Google Scholar] [CrossRef] [PubMed]
  167. Pérez-Sánchez, A.; Barrajón-Catalán, E.; Ruiz-Torres, V.; Agulló-Chazarra, L.; Herranz-López, M.; Valdés, A.; Cifuentes, A.; Micol, V. Rosemary (Rosmarinus officinalis) extract causes ROS-induced necrotic cell death and inhibits tumor growth in vivo. Scientific reports 2019, 9, 808. [Google Scholar] [CrossRef]
  168. Wu, H.-C.; Cheng, M.-J.; Yen, C.-H.; Chen, Y.-M.A.; Chen, Y.-S.; Chen, I.-S.; Chang, H.-S. Chemical Constituents with GNMT-Promoter-Enhancing and NRF2-Reduction Activities from Taiwan Agarwood Excoecaria formosana. Molecules (Basel, Switzerland) 2020, 25, 1746. [Google Scholar] [CrossRef]
  169. Gao, A.-M.; Ke, Z.-P.; Shi, F.; Sun, G.-C.; Chen, H. Chrysin enhances sensitivity of BEL-7402/ADM cells to doxorubicin by suppressing PI3K/Akt/Nrf2 and ERK/Nrf2 pathway. Chemico-Biological Interactions 2013, 206, 100–108. [Google Scholar] [CrossRef] [PubMed]
  170. Hou, X.; Bai, X.; Gou, X.; Zeng, H.; Xia, C.; Zhuang, W.; Chen, X.; Zhao, Z.; Huang, M.; Jin, J. 3′,4′,5′,5,7-Pentamethoxyflavone Sensitizes Cisplatin-Resistant A549 Cells to Cisplatin by Inhibition of Nrf2 Pathway. Molecules and Cells 2015, 38, 396–401. [Google Scholar] [CrossRef] [PubMed]
  171. Y-J, L.; J-H, I.; DM, L.; J-S, P.; SY, W.; M-K, C.; al., e. Synergistic inhibition of mesothelioma cell growth by the combination of clofarabine and resveratrol involves Nrf2 downregulation. BMB Reports 2012, 45, 647–652. [CrossRef] [PubMed]
  172. Tan, X.; Jin, P.; Feng, L.; Song, J.; Sun, E.; Liu, W.; Shu, L.; Jia, X. Protective effect of luteolin on cigarette smoke extract-induced cellular toxicity and apoptosis in normal human bronchial epithelial cells via the Nrf2 pathway. Oncol Rep 2014, 31, 1855–1862. [Google Scholar] [CrossRef] [PubMed]
  173. Wang, Z.; Yu, K.; Hu, Y.; Su, F.; Gao, Z.; Hu, T.; Yang, Y.; Cao, X.; Qian, F. Schisantherin A induces cell apoptosis through ROS/JNK signaling pathway in human gastric cancer cells. Biochemical Pharmacology 2020, 173, 113673. [Google Scholar] [CrossRef] [PubMed]
  174. Ding, S.; Hou, X.; Yuan, J.; Tan, X.; Chen, J.; Yang, N.; Luo, Y.; Jiang, Z.; Jin, P.; Dong, Z.; et al. Wedelolactone protects human bronchial epithelial cell injury against cigarette smoke extract-induced oxidant stress and inflammation responses through Nrf2 pathway. International Immunopharmacology 2015, 29, 648–655. [Google Scholar] [CrossRef] [PubMed]
  175. Ohnuma, T.; Matsumoto, T.; Itoi, A.; Kawana, A.; Nishiyama, T.; Ogura, K.; Hiratsuka, A. Enhanced sensitivity of A549 cells to the cytotoxic action of anticancer drugs via suppression of Nrf2 by procyanidins from Cinnamomi Cortex extract. Biochemical and Biophysical Research Communications 2011, 413, 623–629. [Google Scholar] [CrossRef] [PubMed]
  176. Ohnuma, T.; Anzai, E.; Suzuki, Y.; Shimoda, M.; Saito, S.; Nishiyama, T.; Ogura, K.; Hiratsuka, A. Selective antagonization of activated Nrf2 and inhibition of cancer cell proliferation by procyanidins from Cinnamomi Cortex extract. Archives of Biochemistry and Biophysics 2015, 585, 17–24. [Google Scholar] [CrossRef] [PubMed]
  177. Wu, T.-Y.; Khor, T.O.; Saw, C.L.L.; Loh, S.C.; Chen, A.I.; Lim, S.S.; Park, J.H.Y.; Cai, L.; Kong, A.-N.T. Anti-inflammatory/Anti-oxidative Stress Activities and Differential Regulation of Nrf2-Mediated Genes by Non-Polar Fractions of Tea Chrysanthemum zawadskii and Licorice Glycyrrhiza uralensis. The AAPS Journal 2011, 13, 1–13. [Google Scholar] [CrossRef] [PubMed]
  178. Afrin, S.; Forbes-Hernández, T.Y.; Cianciosi, D.; Pistollato, F.; Zhang, J.; Pacetti, M.; Amici, A.; Reboredo-Rodríguez, P.; Simal-Gandara, J.; Bompadre, S.; et al. Strawberry tree honey as a new potential functional food. Part 2: Strawberry tree honey increases ROS generation by suppressing Nrf2-ARE and NF-кB signaling pathways and decreases metabolic phenotypes and metastatic activity in colon cancer cells. Journal of Functional Foods 2019, 57, 477–487. [Google Scholar] [CrossRef]
  179. Ghosh, S.; Dutta, N.; Banerjee, P.; Gajbhiye, R.L.; Sareng, H.R.; Kapse, P.; Pal, S.; Burdelya, L.; Mandal, N.C.; Ravichandiran, V.; et al. Induction of monoamine oxidase A-mediated oxidative stress and impairment of NRF2-antioxidant defence response by polyphenol-rich fraction of Bergenia ligulata sensitizes prostate cancer cells in vitro and in vivo. Free Radical Biology and Medicine 2021, 172, 136–151. [Google Scholar] [CrossRef] [PubMed]
  180. Turan, I.; Demir, S.; Yaman, S.O.; Canbolat, D.; Mentese, A.; Aliyazicioglu, Y. An Investigation of the Antiproliferative Effect of Rhododendron luteum Extract on Cervical Cancer (HeLa) Cells via Nrf2 Signaling Pathway. Nutrition and Cancer 2022, 74, 1882–1893. [Google Scholar] [CrossRef]
  181. Pterostilbene Decreases the Antioxidant Defenses of Aggressive Cancer Cells In Vivo: A Physiological Glucocorticoids- and Nrf2-Dependent Mechanism. Antioxidants & redox signaling 2016, 24, 974–990. [CrossRef] [PubMed]
  182. Corrêa, M.G.; Pires, P.R.; Ribeiro, F.V.; Pimentel, S.P.; Cirano, F.R.; Napimoga, M.H.; Casati, M.Z.; Casarin, R.C.V. Systemic treatment with resveratrol reduces the progression of experimental periodontitis and arthritis in rats. PloS one 2018, 13, e0204414. [Google Scholar] [CrossRef] [PubMed]
  183. Uttra, A.M.; Alamgeer; Shahzad, M.; Shabbir, A.; Jahan, S.; Bukhari, I.A.; Assiri, A.M. Ribes orientale: A novel therapeutic approach targeting rheumatoid arthritis with reference to pro-inflammatory cytokines, inflammatory enzymes and anti-inflammatory cytokines. Journal of ethnopharmacology 2019, 237, 92–107. [CrossRef] [PubMed]
  184. Liu, W.; Li, J. Theaflavin-3, 3'-Digallate Attenuates Rheumatoid Inflammation in Mice Through the Nuclear Factor-κB and MAPK Pathways. Archivum immunologiae et therapiae experimentalis 2019, 67, 153–160. [Google Scholar] [CrossRef] [PubMed]
  185. Liu, F.; Liu, Y.; Zhan, S.; Lv, J.; Sun, F.; Weng, B.; Liu, S.; Xia, P. Chebulanin exerts its anti-inflammatory and anti-arthritic effects via inhibiting NF-κB and MAPK activation in collagen-induced arthritis mice. International Immunopharmacology 2020, 88, 106823. [Google Scholar] [CrossRef] [PubMed]
  186. Bao, L.; Ye, J.; Liu, N.; Shao, Y.; Li, W.; Fan, X.; Zhao, D.; Wang, H.; Chen, X. Resveratrol Ameliorates Fibrosis in Rheumatoid Arthritis-Associated Interstitial Lung Disease via the Autophagy-Lysosome Pathway. Molecules (Basel, Switzerland) 2022, 27. [Google Scholar] [CrossRef] [PubMed]
  187. Lin, Y.-L.; Lin, J.-K. (−)-Epigallocatechin-3-gallate Blocks the Induction of Nitric Oxide Synthase by Down-Regulating Lipopolysaccharide-Induced Activity of Transcription Factor Nuclear Factor-κB. Molecular Pharmacology 1997, 52, 465–472. [Google Scholar] [CrossRef] [PubMed]
  188. Karatas, A.; Dagli, A.F.; Orhan, C.; Gencoglu, H.; Ozgen, M.; Sahin, N.; Sahin, K.; Koca, S.S. Epigallocatechin 3-gallate attenuates arthritis by regulating Nrf2, HO-1, and cytokine levels in an experimental arthritis model. Biotechnology and applied biochemistry 2020, 67, 317–322. [Google Scholar] [CrossRef] [PubMed]
  189. Sultana, F.; Neog, M.K.; Rasool, M. Targeted delivery of morin, a dietary bioflavanol encapsulated mannosylated liposomes to the macrophages of adjuvant-induced arthritis rats inhibits inflammatory immune response and osteoclastogenesis. European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V 2017, 115, 229–242. [Google Scholar] [CrossRef] [PubMed]
  190. Farrukh, M.; Saleem, U.; Qasim, M.; Manan, M.; Shah, M.A. Sarcococca saligna extract attenuates formaldehyde-induced arthritis in Wistar rats via modulation of pro-inflammatory and inflammatory biomarkers. Inflammopharmacology 2022, 30, 579–597. [Google Scholar] [CrossRef] [PubMed]
  191. Lu, B.; Li, C.; Jing, L.; Zhuang, F.; Xiang, H.; Chen, Y.; Huang, B. Rosmarinic acid nanomedicine for rheumatoid arthritis therapy: Targeted RONS scavenging and macrophage repolarization. Journal of controlled release : official journal of the Controlled Release Society 2023, 362, 631–646. [Google Scholar] [CrossRef] [PubMed]
  192. Neog, M.K.; Joshua Pragasam, S.; Krishnan, M.; Rasool, M. p-Coumaric acid, a dietary polyphenol ameliorates inflammation and curtails cartilage and bone erosion in the rheumatoid arthritis rat model. BioFactors (Oxford, England) 2017, 43, 698–717. [Google Scholar] [CrossRef] [PubMed]
  193. Almansour, Z.H.; Ibrahim, H.M.; Hamad, R.S.; Abd El-Moaty, H.I. Phenolic-Compound-Rich Opuntia littoralis Ethyl Acetate Extract Relaxes Arthritic Symptoms in Collagen-Induced Mice Model via Bone Morphogenic Markers. Nutrients 2022, 14. [Google Scholar] [CrossRef]
  194. He, B.; Zhang, B.; Wu, F.; Wang, L.; Shi, X.; Qin, W.; Lin, Y.; Ma, S.; Liang, J. Homoplantaginin Inhibits Palmitic Acid-induced Endothelial Cells Inflammation by Suppressing TLR4 and NLRP3 Inflammasome. Journal of cardiovascular pharmacology 2016, 67, 93–101. [Google Scholar] [CrossRef] [PubMed]
  195. Zhang, X.; Wang, G.; Gurley, E.C.; Zhou, H. Flavonoid apigenin inhibits lipopolysaccharide-induced inflammatory response through multiple mechanisms in macrophages. PloS one 2014, 9, e107072. [Google Scholar] [CrossRef] [PubMed]
  196. Fan, S.H.; Wang, Y.Y.; Lu, J.; Zheng, Y.L.; Wu, D.M.; Li, M.Q.; Hu, B.; Zhang, Z.F.; Cheng, W.; Shan, Q. Luteoloside suppresses proliferation and metastasis of hepatocellular carcinoma cells by inhibition of NLRP3 inflammasome. PloS one 2014, 9, e89961. [Google Scholar] [CrossRef] [PubMed]
  197. Wang, C.; Pan, Y.; Zhang, Q.Y.; Wang, F.M.; Kong, L.D. Quercetin and allopurinol ameliorate kidney injury in STZ-treated rats with regulation of renal NLRP3 inflammasome activation and lipid accumulation. PloS one 2012, 7, e38285. [Google Scholar] [CrossRef] [PubMed]
  198. Yang, S.J.; Lim, Y. Resveratrol ameliorates hepatic metaflammation and inhibits NLRP3 inflammasome activation. Metabolism: clinical and experimental 2014, 63, 693–701. [Google Scholar] [CrossRef] [PubMed]
  199. Aruna, R.; Geetha, A.; Suguna, P. Rutin modulates ASC expression in NLRP3 inflammasome: a study in alcohol and cerulein-induced rat model of pancreatitis. Molecular and cellular biochemistry 2014, 396, 269–280. [Google Scholar] [CrossRef] [PubMed]
  200. Kim, T.H.; Ku, S.K.; Bae, J.S. Inhibitory effects of kaempferol-3-O-sophoroside on HMGB1-mediated proinflammatory responses. Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association 2012, 50, 1118–1123. [Google Scholar] [CrossRef] [PubMed]
  201. Yang, E.J.; Lee, W.; Ku, S.K.; Song, K.S.; Bae, J.S. Anti-inflammatory activities of oleanolic acid on HMGB1 activated HUVECs. Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association 2012, 50, 1288–1294. [Google Scholar] [CrossRef] [PubMed]
  202. Yang, B.; Gao, P.; Wu, X.; Yu, J.; Li, Y.; Meng, R.; Li, Y.; Yan, J.; Jin, X. Epigallocatechin-3-gallate attenuates neointimal hyperplasia in a rat model of carotid artery injury by inhibition of high mobility group box 1 expression. Experimental and therapeutic medicine 2017, 14, 1975–1982. [Google Scholar] [CrossRef] [PubMed]
  203. He, W.; Yuan, K.; Ji, B.; Han, Y.; Li, J. Protective effects of curcumin against neuroinflammation induced by Aβ25-35 in primary rat microglia: modulation of high-mobility group box 1, toll-like receptor 4 and receptor for advanced glycation end products expression. Annals of translational medicine 2020, 8, 88. [Google Scholar] [CrossRef] [PubMed]
  204. Buhrmann, C.; Popper, B.; Aggarwal, B.B.; Shakibaei, M. Resveratrol downregulates inflammatory pathway activated by lymphotoxin α (TNF-β) in articular chondrocytes: Comparison with TNF-α. PloS one 2017, 12, e0186993. [Google Scholar] [CrossRef] [PubMed]
  205. Karatas, A.; Orhan, C.; Tuzcu, M.; Şahin, N.; Özercan, İ.H.; Koca, S.S.; Juturu, V.; Şahin, K. Mango ginger (curcuma amada) inhibits collagen-induced arthritis by modulatinginflammatory cytokine levels in rats,. Turkich Journal of Medical Sciences 2020, 50. [Google Scholar] [CrossRef] [PubMed]
  206. Xiong, H.; Cheng, Y.; Zhang, X.; Zhang, X. Effects of taraxasterol on iNOS and COX-2 expression in LPS-induced RAW 264.7 macrophages. Journal of ethnopharmacology 2014, 155, 753–757. [Google Scholar] [CrossRef] [PubMed]
  207. Akyol, S.; Ozturk, G.; Ginis, Z.; Armutcu, F.; Yigitoglu, M.R.; Akyol, O. In vivo and in vitro antıneoplastic actions of caffeic acid phenethyl ester (CAPE): therapeutic perspectives. Nutr Cancer 2013, 65, 515–526. [Google Scholar] [CrossRef] [PubMed]
  208. Mackenzie, G.G.; Carrasquedo, F.; Delfino, J.M.; Keen, C.L.; Fraga, C.G.; Oteiza, P.I. Epicatechin, catechin, and dimeric procyanidins inhibit PMA-induced NF-kappaB activation at multiple steps in Jurkat T cells. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 2004, 18, 167–169. [Google Scholar] [CrossRef] [PubMed]
  209. Min, Y.D.; Choi, C.H.; Bark, H.; Son, H.Y.; Park, H.H.; Lee, S.; Park, J.W.; Park, E.K.; Shin, H.I.; Kim, S.H. Quercetin inhibits expression of inflammatory cytokines through attenuation of NF-kappaB and p38 MAPK in HMC-1 human mast cell line. Inflammation research : official journal of the European Histamine Research Society... [et al.] 2007, 56, 210–215. [Google Scholar] [CrossRef] [PubMed]
  210. Chen, J.-C.; Ho, F.-M.; Pei-Dawn Lee, C.; Chen, C.-P.; Jeng, K.-C.G.; Hsu, H.-B.; Lee, S.-T.; Wen Tung, W.; Lin, W.-W. Inhibition of iNOS gene expression by quercetin is mediated by the inhibition of IκB kinase, nuclear factor-kappa B and STAT1, and depends on heme oxygenase-1 induction in mouse BV-2 microglia. European journal of pharmacology 2005, 521, 9–20. [Google Scholar] [CrossRef]
  211. Ruiz, P.A.; Braune, A.; Hölzlwimmer, G.; Quintanilla-Fend, L.; Haller, D. Quercetin inhibits TNF-induced NF-kappaB transcription factor recruitment to proinflammatory gene promoters in murine intestinal epithelial cells. J Nutr 2007, 137, 1208–1215. [Google Scholar] [CrossRef]
  212. Park, S.E.; Sapkota, K.; Kim, S.; Kim, H.; Kim, S.J. Kaempferol acts through mitogen-activated protein kinases and protein kinase B/AKT to elicit protection in a model of neuroinflammation in BV2 microglial cells. British journal of pharmacology 2011, 164, 1008–1025. [Google Scholar] [CrossRef] [PubMed]
  213. Kim, H.H.; Bae, Y.; Kim, S.H. Galangin attenuates mast cell-mediated allergic inflammation. Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association 2013, 57, 209–216. [Google Scholar] [CrossRef] [PubMed]
  214. Wheeler, D.S.; Catravas, J.D.; Odoms, K.; Denenberg, A.; Malhotra, V.; Wong, H.R. Epigallocatechin-3-gallate, a green tea-derived polyphenol, inhibits IL-1 beta-dependent proinflammatory signal transduction in cultured respiratory epithelial cells. J Nutr 2004, 134, 1039–1044. [Google Scholar] [CrossRef] [PubMed]
  215. Ichikawa, D.; Matsui, A.; Imai, M.; Sonoda, Y.; Kasahara, T. Effect of various catechins on the IL-12p40 production by murine peritoneal macrophages and a macrophage cell line, J774.1. Biological & pharmaceutical bulletin 2004, 27, 1353–1358. [Google Scholar] [CrossRef] [PubMed]
  216. Hussain, T.; Gupta, S.; Adhami, V.M.; Mukhtar, H. Green tea constituent epigallocatechin-3-gallate selectively inhibits COX-2 without affecting COX-1 expression in human prostate carcinoma cells. International journal of cancer 2005, 113, 660–669. [Google Scholar] [CrossRef] [PubMed]
  217. Lin, Y.L.; Lin, J.K. (-)-Epigallocatechin-3-gallate blocks the induction of nitric oxide synthase by down-regulating lipopolysaccharide-induced activity of transcription factor nuclear factor-kappaB. Mol Pharmacol 1997, 52, 465–472. [Google Scholar] [CrossRef] [PubMed]
  218. Ji, G.; Zhang, Y.; Yang, Q.; Cheng, S.; Hao, J.; Zhao, X.; Jiang, Z. Genistein suppresses LPS-induced inflammatory response through inhibiting NF-κB following AMP kinase activation in RAW 264.7 macrophages. PloS one 2012, 7, e53101. [Google Scholar] [CrossRef] [PubMed]
  219. Nishitani, Y.; Yamamoto, K.; Yoshida, M.; Azuma, T.; Kanazawa, K.; Hashimoto, T.; Mizuno, M. Intestinal anti-inflammatory activity of luteolin: role of the aglycone in NF-κB inactivation in macrophages co-cultured with intestinal epithelial cells. BioFactors (Oxford, England) 2013, 39, 522–533. [Google Scholar] [CrossRef] [PubMed]
  220. Chen, C.Y.; Peng, W.H.; Tsai, K.D.; Hsu, S.L. Luteolin suppresses inflammation-associated gene expression by blocking NF-kappaB and AP-1 activation pathway in mouse alveolar macrophages. Life sciences 2007, 81, 1602–1614. [Google Scholar] [CrossRef] [PubMed]
  221. Gründemann, C.; Gruber, C.W.; Hertrampf, A.; Zehl, M.; Kopp, B.; Huber, R. An aqueous birch leaf extract of Betula pendula inhibits the growth and cell division of inflammatory lymphocytes. Journal of ethnopharmacology 2011, 136, 444–451. [Google Scholar] [CrossRef] [PubMed]
  222. Carluccio, M.A.; Siculella, L.; Ancora, M.A.; Massaro, M.; Scoditti, E.; Storelli, C.; Visioli, F.; Distante, A.; De Caterina, R. Olive oil and red wine antioxidant polyphenols inhibit endothelial activation: antiatherogenic properties of Mediterranean diet phytochemicals. Arteriosclerosis, thrombosis, and vascular biology 2003, 23, 622–629. [Google Scholar] [CrossRef] [PubMed]
  223. Seeram, N.P.; Momin, R.A.; Nair, M.G.; Bourquin, L.D. Cyclooxygenase inhibitory and antioxidant cyanidin glycosides in cherries and berries. Phytomedicine : international journal of phytotherapy and phytopharmacology 2001, 8, 362–369. [Google Scholar] [CrossRef] [PubMed]
  224. Delgado, J.; del Pilar Terrón, M.; Garrido, M.; Barriga, C.; Espino, J.; Paredes, D.S.; Rodríguez, B.A. Jerte Valley cherry-based product modulates serum inflammatory markers in rats and ringdoves. Journal of Applied Biomedicine 2012, 10, 41–50. [Google Scholar] [CrossRef]
  225. Suk, S.; Kwon, G.T.; Lee, E.; Jang, W.J.; Yang, H.; Kim, J.H.; Thimmegowda, N.R.; Chung, M.-Y.; Kwon, J.Y.; Yang, S.; et al. Gingerenone A, a polyphenol present in ginger, suppresses obesity and adipose tissue inflammation in high-fat diet-fed mice. Molecular nutrition & food research 2017, 61, 1700139. [Google Scholar] [CrossRef] [PubMed]
  226. Pei, Y.; Parks, J.S.; Kang, H.W. Quercetin alleviates high-fat diet-induced inflammation in brown adipose tissue. Journal of Functional Foods 2021, 85, 104614. [Google Scholar] [CrossRef]
  227. Drummond, E.M.; Harbourne, N.; Marete, E.; Martyn, D.; Jacquier, J.; O'Riordan, D.; Gibney, E.R. Inhibition of proinflammatory biomarkers in THP1 macrophages by polyphenols derived from chamomile, meadowsweet and willow bark. Phytotherapy research : PTR 2013, 27, 588–594. [Google Scholar] [CrossRef] [PubMed]
  228. Yeo, J.; Lee, J.; Yoon, S.; Kim, W.J. Tannic acid-based nanogel as an efficient anti-inflammatory agent. Biomaterials Science 2020, 8, 1148–1159. [Google Scholar] [CrossRef]
  229. Siddiqui, A.M.; Cui, X.; Wu, R.; Dong, W.; Zhou, M.; Hu, M.; Simms, H.H.; Wang, P. The anti-inflammatory effect of curcumin in an experimental model of sepsis is mediated by up-regulation of peroxisome proliferator-activated receptor-gamma. Critical care medicine 2006, 34, 1874–1882. [Google Scholar] [CrossRef]
  230. Lee, T.-P.; Matteliano, M.L.; Middleton, E. Effect of quercetin on human polymorphonuclear leukocyte lysosomal enzyme release and phospholipid metabolism. Life sciences 1982, 31, 2765–2774. [Google Scholar] [CrossRef]
  231. Gusman, G.S.; Campana, P.R.V.; Castro, L.C.; Castilho, R.O.; Teixeira, M.M.; Braga, F.C. Evaluation of the Effects of Some Brazilian Medicinal Plants on the Production of TNF-<b><i>α</i></b> and CCL2 by THP-1 Cells. Evidence-Based Complementary and Alternative Medicine 2015, 2015, 497123. [Google Scholar] [CrossRef]
  232. Wang, K.; Ping, S.; Huang, S.; Hu, L.; Xuan, H.; Zhang, C.; Hu, F. Molecular mechanisms underlying the in vitro anti-inflammatory effects of a flavonoid-rich ethanol extract from chinese propolis (poplar type). Evidence-based complementary and alternative medicine : eCAM 2013, 2013, 127672. [Google Scholar] [CrossRef] [PubMed]
  233. Lai, Z.R.; Ho, Y.L.; Huang, S.C.; Huang, T.H.; Lai, S.C.; Tsai, J.C.; Wang, C.Y.; Huang, G.J.; Chang, Y.S. Antioxidant, anti-inflammatory and antiproliferative activities of Kalanchoe gracilis (L.) DC stem. The American journal of Chinese medicine 2011, 39, 1275–1290. [Google Scholar] [CrossRef] [PubMed]
  234. Park, K.I.; Kang, S.R.; Park, H.S.; Lee, D.H.; Nagappan, A.; Kim, J.A.; Shin, S.C.; Kim, E.H.; Lee, W.S.; Chung, H.J.; et al. Regulation of Proinflammatory Mediators via NF-κB and p38 MAPK-Dependent Mechanisms in RAW 264.7 Macrophages by Polyphenol Components Isolated from Korea Lonicera japonica THUNB. Evidence-based complementary and alternative medicine : eCAM 2012, 2012, 828521. [Google Scholar] [CrossRef] [PubMed]
  235. Essafi-Benkhadir, K.; Refai, A.; Riahi, I.; Fattouch, S.; Karoui, H.; Essafi, M. Quince (Cydonia oblonga Miller) peel polyphenols modulate LPS-induced inflammation in human THP-1-derived macrophages through NF-κB, p38MAPK and Akt inhibition. Biochem Biophys Res Commun 2012, 418, 180–185. [Google Scholar] [CrossRef] [PubMed]
  236. Jacob, R.A.; Spinozzi, G.M.; Simon, V.A.; Kelley, D.S.; Prior, R.L.; Hess-Pierce, B.; Kader, A.A. Consumption of Cherries Lowers Plasma Urate in Healthy Women. The Journal of Nutrition 2003, 133, 1826–1829. [Google Scholar] [CrossRef] [PubMed]
  237. Tong, W.W.; Zhang, C.; Hong, T.; Liu, D.H.; Wang, C.; Li, J.; He, X.K.; Xu, W.D. Silibinin alleviates inflammation and induces apoptosis in human rheumatoid arthritis fibroblast-like synoviocytes and has a therapeutic effect on arthritis in rats. Scientific reports 2018, 8, 3241. [Google Scholar] [CrossRef] [PubMed]
  238. Yousefi, Z.; Mirsanei, Z.; Bitaraf, F.S.; Mahdavi, S.; Mirzaii, M.; Jafari, R. Dose-dependent effects of oleuropein administration on regulatory T-cells in patients with rheumatoid arthritis: An in vitro approach. International journal of immunopathology and pharmacology 2022, 36, 3946320221086084. [Google Scholar] [CrossRef] [PubMed]
  239. Rosillo, M.Á.; 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. British Journal of Nutrition 2018, 121, 55–62. [Google Scholar] [CrossRef] [PubMed]
  240. Więcek, K.; Kupczyk, P.; Chodaczek, G.; Woźniak, M. The Impact of Curcumin on the Inflammatory Profile of SW982 Cells in a Rheumatoid Arthritis Model. Journal of immunology research 2022, 2022, 1208970. [Google Scholar] [CrossRef] [PubMed]
  241. Park, C.; Moon, D.O.; Choi, I.W.; Choi, B.T.; Nam, T.J.; Rhu, C.H.; Kwon, T.K.; Lee, W.H.; Kim, G.Y.; Choi, Y.H. Curcumin induces apoptosis and inhibits prostaglandin E(2) production in synovial fibroblasts of patients with rheumatoid arthritis. International journal of molecular medicine 2007, 20, 365–372. [Google Scholar] [CrossRef] [PubMed]
  242. Kloesch, B.; Becker, T.; Dietersdorfer, E.; Kiener, H.; Steiner, G. Anti-inflammatory and apoptotic effects of the polyphenol curcumin on human fibroblast-like synoviocytes. Int Immunopharmacol 2013, 15, 400–405. [Google Scholar] [CrossRef] [PubMed]
  243. Kim, H.R.; Kim, B.M.; Won, J.Y.; Lee, K.A.; Ko, H.M.; Kang, Y.S.; Lee, S.H.; Kim, K.W. Quercetin, a Plant Polyphenol, Has Potential for the Prevention of Bone Destruction in Rheumatoid Arthritis. J Med Food 2019, 22, 152–161. [Google Scholar] [CrossRef] [PubMed]
  244. Huang, M.; Wu, K.; Zeng, S.; Liu, W.; Cui, T.; Chen, Z.; Lin, L.; Chen, D.; Ouyang, H. Punicalagin Inhibited Inflammation and Migration of Fibroblast-Like Synoviocytes Through NF-κB Pathway in the Experimental Study of Rheumatoid Arthritis. Journal of inflammation research 2021, 14, 1901–1913. [Google Scholar] [CrossRef] [PubMed]
  245. Li, T.; Liu, X.; Han, P.; Aimaier, A.; Zhang, Y.; Li, J. Syringaldehyde ameliorates mouse arthritis by inhibiting dendritic cell maturation and proinflammatory cytokine secretion. Int Immunopharmacol 2023, 121, 110490. [Google Scholar] [CrossRef] [PubMed]
  246. Hao, L.; Wan, Y.; Xiao, J.; Tang, Q.; Deng, H.; Chen, L. A study of Sirt1 regulation and the effect of resveratrol on synoviocyte invasion and associated joint destruction in rheumatoid arthritis. Molecular medicine reports 2017, 16, 5099–5106. [Google Scholar] [CrossRef] [PubMed]
  247. Yang, C.M.; Chen, Y.W.; Chi, P.L.; Lin, C.C.; Hsiao, L.D. Resveratrol inhibits BK-induced COX-2 transcription by suppressing acetylation of AP-1 and NF-κB in human rheumatoid arthritis synovial fibroblasts. Biochem Pharmacol 2017, 132, 77–91. [Google Scholar] [CrossRef] [PubMed]
  248. 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. Annals of the New York Academy of Sciences 2019, 1457, 166–178. [Google Scholar] [CrossRef] [PubMed]
  249. 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 (Oxford, England) 2020, 46, 441–453. [Google Scholar] [CrossRef] [PubMed]
  250. Yang, G.; Chang, C.C.; Yang, Y.; Yuan, L.; Xu, L.; Ho, C.T.; Li, S. Resveratrol Alleviates Rheumatoid Arthritis via Reducing ROS and Inflammation, Inhibiting MAPK Signaling Pathways, and Suppressing Angiogenesis. J Agric Food Chem 2018, 66, 12953–12960. [Google Scholar] [CrossRef] [PubMed]
  251. Tian, J.; Gao, J.; Chen, J.; Li, F.; Xie, X.; Du, J.; Wang, J.; Mao, N. [Effects of resveratrol on proliferation and apoptosis of TNF-alpha induced rheumatoid arthritis fibroblast-like synoviocytes]. Zhongguo Zhong yao za zhi = Zhongguo zhongyao zazhi = China journal of Chinese materia medica 2010, 35, 1878–1882. [Google Scholar] [CrossRef] [PubMed]
  252. Tian, J.; Chen, J.W.; Gao, J.S.; Li, L.; Xie, X. Resveratrol inhibits TNF-α-induced IL-1β, MMP-3 production in human rheumatoid arthritis fibroblast-like synoviocytes via modulation of PI3kinase/Akt pathway. Rheumatol Int 2013, 33, 1829–1835. [Google Scholar] [CrossRef] [PubMed]
  253. Srivastava, R.K.; Unterman, T.G.; Shankar, S. FOXO transcription factors and VEGF neutralizing antibody enhance antiangiogenic effects of resveratrol. Molecular and cellular biochemistry 2010, 337, 201–212. [Google Scholar] [CrossRef] [PubMed]
  254. Li, S.; Du, J.; Gan, H.; Chen, J.; Zhou, Y.; Tian, J.; Ling, G.; Li, F. Resveratrol promotes apoptosis and G2/M cell cycle arrest of fibroblast-like synoviocytes in rheumatoid arthritis through regulation of autophagy and the serine-threonine kinase-p53 axis. Archives of medical science : AMS 2024, 20, 280–288. [Google Scholar] [CrossRef] [PubMed]
  255. Nakayama, H.; Yaguchi, T.; Yoshiya, S.; Nishizaki, T. Resveratrol induces apoptosis MH7A human rheumatoid arthritis synovial cells in a sirtuin 1-dependent manner. Rheumatol Int 2012, 32, 151–157. [Google Scholar] [CrossRef]
  256. Tang, L.L.; Gao, J.S.; Chen, X.R.; Xie, X. [Inhibitory effect of resveratrol on the proliferation of synoviocytes in rheumatoid arthritis and its mechanism in vitro]. Zhong nan da xue xue bao. Yi xue ban = Journal of Central South University. Medical sciences 2006, 31, 528–533. [Google Scholar] [PubMed]
  257. Wang, T.; Wang, G.; Zhang, Y.; Zhang, J.; Cao, W.; Chen, X. Effect of lentivirus-mediated overexpression or silencing of MnSOD on apoptosis of resveratrol-treated fibroblast-like synoviocytes in rheumatoid arthritis. European journal of pharmacology 2019, 844, 65–72. [Google Scholar] [CrossRef] [PubMed]
  258. Glehr, M.; Breisach, M.; Walzer, S.; Lohberger, B.; Fürst, F.; Friesenbichler, J.; Rinner, B.; Avian, A.; Windhager, R.; Leithner, A. The influence of resveratrol on the synovial expression of matrix metalloproteinases and receptor activator of NF-kappaB ligand in rheumatoid arthritis fibroblast-like synoviocytes. Zeitschrift fur Naturforschung. C, Journal of biosciences 2013, 68, 336–342. [Google Scholar] [CrossRef] [PubMed]
  259. Chen, X.M.; Guo, Y.J.; Ling, H.W.; Zeng, R. The Effect of Resveratrol in Sirt1/CST Pathway to Inhibit TNF-α Induced Inflammatory Response in Rat Primary Fibroblast-Like Synoviocytes. Biological & pharmaceutical bulletin 2023, 46, 1592–1600. [Google Scholar] [CrossRef] [PubMed]
  260. Glehr, M.; Fritsch-Breisach, M.; Lohberger, B.; Walzer, S.M.; Moazedi-Fuerst, F.; Rinner, B.; Gruber, G.; Graninger, W.; Leithner, A.; Windhager, R. Influence of resveratrol on rheumatoid fibroblast-like synoviocytes analysed with gene chip transcription. Phytomedicine : international journal of phytotherapy and phytopharmacology 2013, 20, 310–318. [Google Scholar] [CrossRef] [PubMed]
  261. Lu, J.; Zheng, Y.; Yang, J.; Zhang, J.; Cao, W.; Chen, X.; Fang, S. Resveratrol alleviates inflammatory injury and enhances the apoptosis of fibroblast-like synoviocytes via mitochondrial dysfunction and ER stress in rats with adjuvant arthritis. Molecular medicine reports 2019, 20, 463–472. [Google Scholar] [CrossRef] [PubMed]
  262. Tsai, M.-H.; Hsu, L.-F.; Lee, C.-W.; Chiang, Y.-C.; Lee, M.-H.; How, J.-M.; Wu, C.-M.; Huang, C.-L.; Lee, I.T. Resveratrol inhibits urban particulate matter-induced COX-2/PGE2 release in human fibroblast-like synoviocytes via the inhibition of activation of NADPH oxidase/ROS/NF-κB. The International Journal of Biochemistry & Cell Biology 2017, 88, 113–123. [Google Scholar] [CrossRef] [PubMed]
  263. Shakibaei, M.; Buhrmann, C.; Mobasheri, A. Resveratrol-mediated SIRT-1 interactions with p300 modulate receptor activator of NF-kappaB ligand (RANKL) activation of NF-kappaB signaling and inhibit osteoclastogenesis in bone-derived cells. The Journal of biological chemistry 2011, 286, 11492–11505. [Google Scholar] [CrossRef] [PubMed]
  264. Lomholt, S.; Mellemkjaer, A.; Iversen, M.B.; Pedersen, S.B.; Kragstrup, T.W. Resveratrol displays anti-inflammatory properties in an ex vivo model of immune mediated inflammatory arthritis. BMC rheumatology 2018, 2, 27. [Google Scholar] [CrossRef] [PubMed]
  265. Lee, S.H.; Park, H.H.; Kim, J.E.; Kim, J.A.; Kim, Y.H.; Jun, C.D.; Kim, S.H. Allose gallates suppress expression of pro-inflammatory cytokines through attenuation of NF-kappaB in human mast cells. Planta Med 2007, 73, 769–773. [Google Scholar] [CrossRef] [PubMed]
  266. Song, H.; Wu, H.; Dong, J.; Huang, S.; Ye, J.; Liu, R. Ellagic Acid Alleviates Rheumatoid Arthritis in Rats through Inhibiting MTA1/HDAC1-Mediated Nur77 Deacetylation. Mediators of inflammation 2021, 2021, 6359652. [Google Scholar] [CrossRef]
  267. Al-Kafaween, M.A.; Alwahsh, M.; Mohd Hilmi, A.B.; Abulebdah, D.H. Physicochemical Characteristics and Bioactive Compounds of Different Types of Honey and Their Biological and Therapeutic Properties: A Comprehensive Review. Antibiotics (Basel, Switzerland) 2023, 12. [Google Scholar] [CrossRef] [PubMed]
  268. Neog, M.K.; Rasool, M. Targeted delivery of p-coumaric acid encapsulated mannosylated liposomes to the synovial macrophages inhibits osteoclast formation and bone resorption in the rheumatoid arthritis animal model. European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V 2018, 133, 162–175. [Google Scholar] [CrossRef] [PubMed]
  269. Ganesan, R.; Rasool, M. Ferulic acid inhibits interleukin 17-dependent expression of nodal pathogenic mediators in fibroblast-like synoviocytes of rheumatoid arthritis. Journal of cellular biochemistry 2019, 120, 1878–1893. [Google Scholar] [CrossRef] [PubMed]
  270. Doss, H.M.; Samarpita, S.; Ganesan, R.; Rasool, M. Ferulic acid, a dietary polyphenol suppresses osteoclast differentiation and bone erosion via the inhibition of RANKL dependent NF-κB signalling pathway. Life sciences 2018, 207, 284–295. [Google Scholar] [CrossRef]
  271. Kwak, S.C.; Lee, C.; Kim, J.Y.; Oh, H.M.; So, H.S.; Lee, M.S.; Rho, M.C.; Oh, J. Chlorogenic acid inhibits osteoclast differentiation and bone resorption by down-regulation of receptor activator of nuclear factor kappa-B ligand-induced nuclear factor of activated T cells c1 expression. Biological & pharmaceutical bulletin 2013, 36, 1779–1786. [Google Scholar] [CrossRef] [PubMed]
  272. Yi, Z.; Ran, Y.; Chen, X.; Tong, Q.; Ma, L.; Tan, Y.; Ma, X.; Li, X. Tea polyphenol carrier-enhanced dexamethasone nanomedicines for inflammation-targeted treatment of rheumatoid arthritis. Journal of materials chemistry. B 2023, 11, 11505–11518. [Google Scholar] [CrossRef] [PubMed]
  273. George, G.; Shyni, G.L.; Mohan, S.; Abraham, B.; Nisha, P.; Ranjith, S.; Rajankutty, K.; Raghu, K.G. In vitro and in vivo anti-inflammatory and anti-arthritic effect of Tinospora cordifolia via modulation of JAK/STAT pathway. Inflammopharmacology 2023, 31, 1009–1025. [Google Scholar] [CrossRef] [PubMed]
  274. South, S.; Lucero, J.; Vijayagopal, P.; Juma, S. Anti-Inflammatory Action of Blueberry Polyphenols in HIG-82 Rabbit Synoviocytes. J Med Food 2019, 22, 1032–1040. [Google Scholar] [CrossRef] [PubMed]
  275. Kim, J.E.; Son, J.E.; Jung, S.K.; Kang, N.J.; Lee, C.Y.; Lee, K.W.; Lee, H.J. Cocoa polyphenols suppress TNF-α-induced vascular endothelial growth factor expression by inhibiting phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase kinase-1 (MEK1) activities in mouse epidermal cells. The British journal of nutrition 2010, 104, 957–964. [Google Scholar] [CrossRef] [PubMed]
  276. Kim, E.J.; Seo, J.B.; Yu, J.S.; Lee, S.; Lim, J.S.; Choi, J.U.; Lee, C.M.; Rashan, L.; Kim, K.H.; Cho, Y.C. Anti-Inflammatory Effects of a Polyphenol, Catechin-7,4'-O-Digallate, from Woodfordia uniflora by Regulating NF-κB Signaling Pathway in Mouse Macrophages. Pharmaceutics 2021, 13. [Google Scholar] [CrossRef]
  277. Sekiguchi, Y.; Mano, H.; Nakatani, S.; Shimizu, J.; Kobata, K.; Wada, M. Anti-proliferative effects of Salacia reticulata leaves hot-water extract on interleukin-1β-activated cells derived from the synovium of rheumatoid arthritis model mice. BMC research notes 2012, 5, 198. [Google Scholar] [CrossRef] [PubMed]
  278. Zhang, J.; Song, X.; Cao, W.; Lu, J.; Wang, X.; Wang, G.; Wang, Z.; Chen, X. Autophagy and mitochondrial dysfunction in adjuvant-arthritis rats treatment with resveratrol. Scientific reports 2016, 6, 32928. [Google Scholar] [CrossRef] [PubMed]
  279. Oz, B.; Yildirim, A.; Yolbas, S.; Celik, Z.B.; Etem, E.O.; Deniz, G.; Akin, M.; Akar, Z.A.; Karatas, A.; Koca, S.S. Resveratrol inhibits Src tyrosine kinase, STAT3, and Wnt signaling pathway in collagen induced arthritis model. BioFactors (Oxford, England) 2019, 45, 69–74. [Google Scholar] [CrossRef]
  280. Zhang, Y.; Zhang, S.; Liu, Z.; Zhao, X.; Yuan, Y.; Sheng, L.; Li, Y. Resveratrol prevents atrial fibrillation by inhibiting atrial structural and metabolic remodeling in collagen-induced arthritis rats. Naunyn-Schmiedeberg's archives of pharmacology 2018, 391, 1179–1190. [Google Scholar] [CrossRef] [PubMed]
  281. Wahba, M.G.; Messiha, B.A.; Abo-Saif, A.A. Protective effects of fenofibrate and resveratrol in an aggressive model of rheumatoid arthritis in rats. Pharmaceutical biology 2016, 54, 1705–1715. [Google Scholar] [CrossRef] [PubMed]
  282. El-Ghazaly, M.A.; Fadel, N.A.; Abdel-Naby, D.H.; Abd El-Rehim, H.A.; Zaki, H.F.; Kenawy, S.A. Amelioration of adjuvant-induced arthritis by exposure to low dose gamma radiation and resveratrol administration in rats. International journal of radiation biology 2020, 96, 857–867. [Google Scholar] [CrossRef] [PubMed]
  283. Elmali, N.; Baysal, O.; Harma, A.; Esenkaya, I.; Mizrak, B. Effects of resveratrol in inflammatory arthritis. Inflammation 2007, 30, 1–6. [Google Scholar] [CrossRef] [PubMed]
  284. Xuzhu, G.; Komai-Koma, M.; Leung, B.P.; Howe, H.S.; McSharry, C.; McInnes, I.B.; Xu, D. Resveratrol modulates murine collagen-induced arthritis by inhibiting Th17 and B-cell function. Ann Rheum Dis 2012, 71, 129–135. [Google Scholar] [CrossRef] [PubMed]
  285. Lu, J.; Yang, J.; Zheng, Y.; Fang, S.; Chen, X. Resveratrol reduces store-operated Ca(2+) entry and enhances the apoptosis of fibroblast-like synoviocytes in adjuvant arthritis rats model via targeting ORAI1-STIM1 complex. Biological research 2019, 52, 45. [Google Scholar] [CrossRef]
  286. Fernández-Rodríguez, J.A.; Almonte-Becerril, M.; Ramil-Gómez, O.; Hermida-Carballo, L.; Viñas-Diz, S.; Vela-Anero, Á.; Concha, Á.; Camacho-Encina, M.; Blanco, F.J.; López-Armada, M.J. Autophagy Activation by Resveratrol Reduces Severity of Experimental Rheumatoid Arthritis. Molecular nutrition & food research 2021, 65, e2000377. [Google Scholar] [CrossRef] [PubMed]
  287. Riveiro-Naveira, R.R.; Valcárcel-Ares, M.N.; Almonte-Becerril, M.; Vaamonde-García, C.; Loureiro, J.; Hermida-Carballo, L.; López-Peláez, E.; Blanco, F.J.; López-Armada, M.J. Resveratrol lowers synovial hyperplasia, inflammatory markers and oxidative damage in an acute antigen-induced arthritis model. Rheumatology (Oxford, England) 2016, 55, 1889–1900. [Google Scholar] [CrossRef]
  288. Chen, X.; Lu, J.; An, M.; Ma, Z.; Zong, H.; Yang, J. Anti-inflammatory effect of resveratrol on adjuvant arthritis rats with abnormal immunological function via the reduction of cyclooxygenase-2 and prostaglandin E2. Molecular medicine reports 2014, 9, 2592–2598. [Google Scholar] [CrossRef] [PubMed]
  289. Yang, G.; Lyu, L.; Wang, X.; Bao, L.; Lyu, B.; Lin, Z. Systemic treatment with resveratrol alleviates adjuvant arthritis-interstitial lung disease in rats via modulation of JAK/STAT/RANKL signaling pathway. Pulmonary pharmacology & therapeutics 2019, 56, 69–74. [Google Scholar] [CrossRef]
  290. Poonia, N.; Lather, V.; Kaur, B.; Kirthanashri, S.V.; Pandita, D. Optimization and Development of Methotrexate- and Resveratrol-Loaded Nanoemulsion Formulation Using Box-Behnken Design for Rheumatoid Arthritis. Assay and drug development technologies 2020, 18, 356–368. [Google Scholar] [CrossRef] [PubMed]
  291. Chen, X.; Zhu, X.; Ma, L.; Lin, A.; Gong, Y.; Yuan, G.; Liu, J. A core-shell structure QRu-PLGA-RES-DS NP nanocomposite with photothermal response-induced M2 macrophage polarization for rheumatoid arthritis therapy. Nanoscale 2019, 11, 18209–18223. [Google Scholar] [CrossRef] [PubMed]
  292. Chen, Z.; Xiao, G.; Ao, J. Resveratrol Attenuates Rheumatoid Arthritis Induce Neutrophil Extracellular Traps via TLR-4 Mediated Inflammation in C57BL/6 Mice. Physiological research 2024, 73, 91–104. [Google Scholar] [CrossRef] [PubMed]
  293. Min, S.Y.; Yan, M.; Kim, S.B.; Ravikumar, S.; Kwon, S.R.; Vanarsa, K.; Kim, H.Y.; Davis, L.S.; Mohan, C. Green Tea Epigallocatechin-3-Gallate Suppresses Autoimmune Arthritis Through Indoleamine-2,3-Dioxygenase Expressing Dendritic Cells and the Nuclear Factor, Erythroid 2-Like 2 Antioxidant Pathway. Journal of inflammation (London, England) 2015, 12, 53. [Google Scholar] [CrossRef] [PubMed]
  294. Chen, M.S.; Tsai, B.C.; Sitorus, M.A.; Kuo, C.H.; Kuo, W.W.; Chen, T.S.; Fu, C.Y.; Ho, T.J.; Huang, C.Y.; Ju, D.T. Epigallocatechin-3-Gallate Pretreatment Improves Autologous Adipose-derived Stem Cells Against Rheumatoid Arthritis-induced Neuroinflammation in the Brain of Collagen-induced Rats. Neurotoxicity research 2022, 40, 1223–1234. [Google Scholar] [CrossRef] [PubMed]
  295. Lee, S.-Y.; Jung, Y.O.; Ryu, J.-G.; Oh, H.-J.; Son, H.-J.; Lee, S.H.; Kwon, J.-E.; Kim, E.-K.; Park, M.-K.; Park, S.-H.; et al. Epigallocatechin-3-gallate ameliorates autoimmune arthritis by reciprocal regulation of T helper-17 regulatory T cells and inhibition of osteoclastogenesis by inhibiting STAT3 signaling. Journal of Leukocyte Biology 2016, 100, 559–568. [Google Scholar] [CrossRef] [PubMed]
  296. Song, C.; Xu, S.; Chang, L.; Zhao, X.; Mei, X.; Ren, X.; Chen, Z. Preparation of EGCG decorated, injectable extracellular vesicles for cartilage repair in rat arthritis. Regenerative biomaterials 2021, 8, rbab067. [Google Scholar] [CrossRef] [PubMed]
  297. Reiter, M.P.; Ward, S.H.; Perry, B.; Mann, A.; Freeman, J.W.; Tiku, M.L. Intra-articular injection of epigallocatechin (EGCG) crosslinks and alters biomechanical properties of articular cartilage, a study via nanoindentation. PloS one 2022, 17, e0276626. [Google Scholar] [CrossRef] [PubMed]
  298. Leichsenring, A.; Bäcker, I.; Furtmüller, P.G.; Obinger, C.; Lange, F.; Flemmig, J. Long-Term Effects of (-)-Epigallocatechin Gallate (EGCG) on Pristane-Induced Arthritis (PIA) in Female Dark Agouti Rats. PloS one 2016, 11, e0152518. [Google Scholar] [CrossRef] [PubMed]
  299. Kim, H.R.; Rajaiah, R.; Wu, Q.L.; Satpute, S.R.; Tan, M.T.; Simon, J.E.; Berman, B.M.; Moudgil, K.D. Green tea protects rats against autoimmune arthritis by modulating disease-related immune events. J Nutr 2008, 138, 2111–2116. [Google Scholar] [CrossRef] [PubMed]
  300. 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] [PubMed]
  301. Roy, S.; Sannigrahi, S.; Majumdar, S.; Ghosh, B.; Sarkar, B. Resveratrol regulates antioxidant status, inhibits cytokine expression and restricts apoptosis in carbon tetrachloride induced rat hepatic injury. Oxidative medicine and cellular longevity 2011, 2011, 703676. [Google Scholar] [CrossRef] [PubMed]
  302. Adhikary, R.; Sultana, S.; Bishayi, B. Clitoria ternatea flower petals: Effect on TNFR1 neutralization via downregulation of synovial matrix metalloproteases. Journal of ethnopharmacology 2018, 210, 209–222. [Google Scholar] [CrossRef] [PubMed]
  303. Alamgeer; Uttra, A.M.; Hasan, U.H. Anti-arthritic activity of aqueous-methanolic extract and various fractions of Berberis orthobotrys Bien ex Aitch. BMC complementary and alternative medicine 2017, 17, 371. [CrossRef] [PubMed]
  304. Zia, S.; Saleem, M.; Asif, M.; Hussain, K.; Butt, B.Z. Diospyros malabarica (Desr.) Kostel fruits extract attenuated acute and chronic inflammation through modulation of the expression of pro- and anti-inflammatory biomarkers in rat models. Inflammopharmacology 2022, 30, 2211–2227. [Google Scholar] [CrossRef] [PubMed]
  305. Pragasam, S.J.; Venkatesan, V.; Rasool, M. Immunomodulatory and anti-inflammatory effect of p-coumaric acid, a common dietary polyphenol on experimental inflammation in rats. Inflammation 2013, 36, 169–176. [Google Scholar] [CrossRef] [PubMed]
  306. Shi, C.; Zhang, H.; Wang, X.; Jin, B.; Jia, Q.; Li, Y.; Yang, Y. Cinnamtannin D1 attenuates autoimmune arthritis by regulating the balance of Th17 and treg cells through inhibition of aryl hydrocarbon receptor expression. Pharmacological research 2020, 151, 104513. [Google Scholar] [CrossRef] [PubMed]
  307. Rathi, B.; Bodhankar, S.; Mohan, V.; Thakurdesai, P. Ameliorative Effects of a Polyphenolic Fraction of Cinnamomum zeylanicum L. Bark in Animal Models of Inflammation and Arthritis. Scientia pharmaceutica 2013, 81, 567–589. [Google Scholar] [CrossRef]
  308. Pašková, Ľ.; Kuncírová, V.; Poništ, S.; Mihálová, D.; Nosáľ, R.; Harmatha, J.; Hrádková, I.; Čavojský, T.; Bilka, F.; Šišková, K.; et al. Effect of N-Feruloylserotonin and Methotrexate on Severity of Experimental Arthritis and on Messenger RNA Expression of Key Proinflammatory Markers in Liver. Journal of immunology research 2016, 2016, 7509653. [Google Scholar] [CrossRef]
  309. Rosillo, M.; Alcaraz, M.J.; Sánchez-Hidalgo, M.; Fernández-Bolaños, J.G.; Alarcón-de-la-Lastra, C.; Ferrándiz, M.L. Anti-inflammatory and joint protective effects of extra-virgin olive-oil polyphenol extract in experimental arthritis. J Nutr Biochem 2014, 25, 1275–1281. [Google Scholar] [CrossRef]
  310. Rosillo, M.A.; Sánchez-Hidalgo, M.; González-Benjumea, A.; Fernández-Bolaños, J.G.; Lubberts, E.; Alarcón-de-la-Lastra, C. Preventive effects of dietary hydroxytyrosol acetate, an extra virgin olive oil polyphenol in murine collagen-induced arthritis. Molecular nutrition & food research 2015, 59, 2537–2546. [Google Scholar] [CrossRef] [PubMed]
  311. Tsubaki, M.; Takeda, T.; Kino, T.; Itoh, T.; Imano, M.; Tanabe, G.; Muraoka, O.; Satou, T.; Nishida, S. Mangiferin suppresses CIA by suppressing the expression of TNF-α, IL-6, IL-1β, and RANKL through inhibiting the activation of NF-κB and ERK1/2. American journal of translational research 2015, 7, 1371–1381. [Google Scholar]
  312. Zheng, Z.; Sun, Y.; Liu, Z.; Zhang, M.; Li, C.; Cai, H. The effect of curcumin and its nanoformulation on adjuvant-induced arthritis in rats. Drug design, development and therapy 2015, 9, 4931–4942. [Google Scholar] [CrossRef] [PubMed]
  313. Kiki, G.; Pop, R.M.; Sabin, O.; Bocsan, I.C.; Chedea, V.S.; Socaci, S.A.; Pârvu, A.E.; Finsia, E.; Francis, T.; Mathieu, Z.; et al. Polyphenols from Dichrostachys cinerea Fruits Anti-Inflammatory, Analgesic, and Antioxidant Capacity in Freund's Adjuvant-Induced Arthritic Rat Model. Molecules (Basel, Switzerland) 2022, 27. [Google Scholar] [CrossRef] [PubMed]
  314. Zhang, Q.; Yu, Y.; Li, J.; Guan, Y.; Huang, J.; Wang, Z.; Zhang, Z.; Zhang, W.; Guo, J.; Li, J.; et al. Anti-arthritic activities of ethanol extracts of Circaea mollis Sieb. & Zucc. (whole plant) in rodents. Journal of ethnopharmacology 2018, 225, 359–366. [Google Scholar] [CrossRef] [PubMed]
  315. Kumatia, E.K.; Baffour, P.K.; Bolah, P. Antiarthritic and Antioxidant Activities of Antrocaryon micraster Seed Extract and Its Fractions. BioMed research international 2024, 2024, 8838626. [Google Scholar] [CrossRef] [PubMed]
  316. Mirza, F.; Lorenzo, J.; Drissi, H.; Lee, F.Y.; Soung, D.Y. Dried plum alleviates symptoms of inflammatory arthritis in TNF transgenic mice. The Journal of nutritional biochemistry 2018, 52, 54–61. [Google Scholar] [CrossRef] [PubMed]
  317. Abid, F.; Saleem, M.; Jamshaid, T.; Jamshaid, U.; Youssef, F.S.; Diri, R.M.; Elhady, S.S.; Ashour, M.L. Opuntia monacantha: Validation of the anti-inflammatory and anti-arthritic activity of its polyphenolic rich extract in silico and in vivo via assessment of pro- and anti-inflammatory cytokines. Journal of ethnopharmacology 2024, 326, 117884. [Google Scholar] [CrossRef] [PubMed]
  318. Alamgeer; Shanila, A.; Ambreen Malik, U.; Umme Habiba, H. Alkaloids, flavonoids, polyphenols might be responsible for potent antiarthritic effect of Solanum nigrum. Journal of traditional Chinese medicine = Chung i tsa chih ying wen pan 2019, 39, 632–641.
  319. Hannan, A.; Akhtar, B.; Sharif, A.; Anjum, F.; Pasha, I.; Khan, A.; Akhtar, M.F.; Saleem, A. Quercetin-loaded chitosan nanoparticles ameliorate adjuvant-induced arthritis in rats by regulating anti-oxidant enzymes and downregulating pro- and inflammatory cytokines. Inflammopharmacology 2023, 31, 287–300. [Google Scholar] [CrossRef] [PubMed]
  320. Mossalayi, M.D.; Rambert, J.; Renouf, E.; Micouleau, M.; Mérillon, J.M. Grape polyphenols and propolis mixture inhibits inflammatory mediator release from human leukocytes and reduces clinical scores in experimental arthritis. Phytomedicine : international journal of phytotherapy and phytopharmacology 2014, 21, 290–297. [Google Scholar] [CrossRef]
  321. Bouhlali, E.d.T.; Hmidani, A.; Bourkhis, B.; Khouya, T.; Ramchoun, M.; Filali-Zegzouti, Y.; Alem, C. Phenolic profile and anti-inflammatory activity of four Moroccan date (Phoenix dactylifera L.) seed varieties. Heliyon 2020, 6, e03436. [Google Scholar] [CrossRef] [PubMed]
  322. Thimóteo, N.S.B.; Iryioda, T.M.V.; Alfieri, D.F.; Rego, B.E.F.; Scavuzzi, B.M.; Fatel, E.; Lozovoy, M.A.B.; Simão, A.N.C.; Dichi, I. Cranberry juice decreases disease activity in women with rheumatoid arthritis. Nutrition (Burbank, Los Angeles County, Calif.) 2019, 60, 112–117. [Google Scholar] [CrossRef] [PubMed]
  323. Fatel, E.C.S.; Rosa, F.T.; Alfieri, D.F.; Flauzino, T.; Scavuzzi, B.M.; Lozovoy, M.A.B.; Iriyoda, T.M.V.; Simão, A.N.C.; Dichi, I. Beneficial effects of fish oil and cranberry juice on disease activity and inflammatory biomarkers in people with rheumatoid arthritis. Nutrition (Burbank, Los Angeles County, Calif.) 2021, 86, 111183. [Google Scholar] [CrossRef] [PubMed]
  324. 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. European Journal of Clinical Nutrition 2017, 71, 92–96. [Google Scholar] [CrossRef] [PubMed]
  325. Khojah, H.M.; Ahmed, S.; Abdel-Rahman, M.S.; Elhakeim, E.H. Resveratrol as an effective adjuvant therapy in the management of rheumatoid arthritis: a clinical study. Clinical rheumatology 2018, 37, 2035–2042. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Main dietary polyphenols and simple phenols, and also their most notable health-benefits (OH: hydroxyl group) (adapted from Mamari [30], Gonçalves et al. [31] and Matsumura et al. [32].
Figure 1. Main dietary polyphenols and simple phenols, and also their most notable health-benefits (OH: hydroxyl group) (adapted from Mamari [30], Gonçalves et al. [31] and Matsumura et al. [32].
Preprints 103419 g001
Figure 2. Effects of polyphenols against rheumatoid arthritis (RA: rheumatoid arthritis; APC: antigen-presenting cell; IL: interleukin; TNF-α: tumor necrosis factor alpha; MMP: matrix metalloproteinases; RASF: rheumatoid arthritis synovial fibroblasts; RANKL; receptor activator of nuclear factor kappa-Β ligand) [Portions of Figure 2 were drawn using images from BioRender.com (https://biorender.com/) (accessed on 8 March 2024)] (adapted from Weyand & Goronzy [132] and Long et al. [6]).
Figure 2. Effects of polyphenols against rheumatoid arthritis (RA: rheumatoid arthritis; APC: antigen-presenting cell; IL: interleukin; TNF-α: tumor necrosis factor alpha; MMP: matrix metalloproteinases; RASF: rheumatoid arthritis synovial fibroblasts; RANKL; receptor activator of nuclear factor kappa-Β ligand) [Portions of Figure 2 were drawn using images from BioRender.com (https://biorender.com/) (accessed on 8 March 2024)] (adapted from Weyand & Goronzy [132] and Long et al. [6]).
Preprints 103419 g002
Figure 3. Representation of phenolics antioxidant abilities explained by Bors criteria, involving their structure. Bors 1: catechol group on the B ring (green); Bors 2: 4-oxo group and 2,3 double bond on the C ring (blue); Bors 3: hydroxyl (OH) groups at 3 and 5 groups on A and C rings combined with a 4-oxo group on C ring (adapted from Platzer and co-workers [139] and created with ChemDraw Professional 16.0 (CambridgeSoft, Perkin Elmer Inc., Waltham, MA, USA).
Figure 3. Representation of phenolics antioxidant abilities explained by Bors criteria, involving their structure. Bors 1: catechol group on the B ring (green); Bors 2: 4-oxo group and 2,3 double bond on the C ring (blue); Bors 3: hydroxyl (OH) groups at 3 and 5 groups on A and C rings combined with a 4-oxo group on C ring (adapted from Platzer and co-workers [139] and created with ChemDraw Professional 16.0 (CambridgeSoft, Perkin Elmer Inc., Waltham, MA, USA).
Preprints 103419 g003
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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Alerts
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

© 2025 MDPI (Basel, Switzerland) unless otherwise stated